{ "Contributors": [ "HoC" ], "Source": [ "HoC" ], "URL": [ "https://github.com/sb895/Hallmarks-of-Cancer" ], "Categories": [ "Text Classification" ], "Definition": [ "Given the biomedical publication abstracts, you are required to classify the hallmarks of cancer. Each article may have one or more of the ten hallmarks. The ten hallmarks are as follows: 1. Sustaining proliferative signaling, 2. Evading growth suppressors, 3. Resisting cell death, 4. Enabling replicative immortality, 5. Inducing angiogenesis, 6. Activating invasion and metastasis, 7. Genomic instability and mutation, 8. Tumor promoting inflammation, 9. Cellular energetics, 10. Avoiding immune destruction." ], "Reasoning": [], "Input_language": [ "English" ], "Output_language": [ "English" ], "Instruction_language": [ "English" ], "Domains": [ "Medical Knowledge" ], "Positive Examples": [], "Negative Examples": [], "Instances": [ { "input": "Ghrelin was identified in the stomach as an endogenous ligand specific for the growth hormone secretagogue receptor ( GHS-R ) .\n\nGHS-R is found in various tissues , but its function is unknown .\n\nHere we show that GHS-R is found in hepatoma cells .\n\nExposure of these cells to ghrelin caused up-regulation of several insulin-induced activities including tyrosine phosphorylation of insulin receptor substrate-1 ( IRS-1 ) , association of the adapter molecule growth factor receptor-bound protein 2 with IRS-1 , mitogen-activated protein kinase activity , and cell proliferation .\n\nUnlike insulin , ghrelin inhibited Akt kinase activity as well as up-regulated gluconeogenesis .\n\nThese findings raise the possibility that ghrelin modulates insulin activities in humans .", "output": "Sustaining proliferative signaling" }, { "input": "PURPOSE The epidermal growth factor receptor ( EGFR ) tyrosine kinase inhibitor ZD1839 ( Iressa ; AstraZeneca Pharmaceuticals , Alderley Park , United Kingdom ) is under development as an anticancer agent .\n\nWe studied the pharmacodynamic effects of ZD1839 on EGFR in the skin , an EGFR-dependent tissue , in cancer patients participating in ZD1839 phase I clinical trials .\n\nPATIENTS AND METHODS We studied 104 pre- and/or on-ZD1839 therapy ( approximately at day 28 of therapy ) skin biopsies from 65 patients receiving escalating doses of daily oral ZD1839 .\n\nWe measured ZD1839 effects on EGFR activation by immunohistochemistry using an antibody specific for the activated ( phosphorylated ) EGFR .\n\nEffects on receptor signaling ( activated mitogen-activated protein kinase [ MAPK] ) , proliferation , p27(KIP1) , and maturation were also assessed .\n\nRESULTS Histopathologically , the stratum corneum of the epidermis was thinner during therapy ( P <.001 ) .\n\nIn hair follicles , prominent keratin plugs and microorganisms were found in dilated infundibula .\n\nZD1839 suppressed EGFR phosphorylation in all EGFR-expressing cells ( P <.001 ) .\n\nIn addition , ZD1839 inhibited MAPK activation ( P <.001 ) and reduced keratinocyte proliferation index ( P <.001 ) .\n\nConcomitantly , ZD1839 increased the expression of p27(KIP1) ( P <.001 ) and maturation markers ( P <.001 ) and increased apoptosis ( P <.001 ) .\n\nThese effects were observed at all dose levels , before reaching dose-limiting toxicities .\n\nCONCLUSION ZD1839 inhibits EGFR activation and affects downstream receptor-dependent processes in vivo .\n\nThese effects were profound at doses well below the one producing unacceptable toxicity , a finding that strongly supports pharmacodynamic assessments to select optimal doses instead of a maximum-tolerated dose for definitive efficacy and safety trials .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Adoptive transfer of immunity against hepatitis B surface antigen ( HBsAg ) was previously shown to facilitate suppression of experimental human hepatocellular carcinoma ( HCC ) expressing HBsAg in athymic mice .\n\nWe have shown that oral tolerance induces antigen-specific immune suppression of HBsAg by feeding hepatitis B virus ( HBV ) antigens .\n\nIn the present study we evaluated the effect of oral tolerance induction toward HBV or HCC antigens on the growth of experimental HCC-expressing HBsAg in mice .\n\nTolerance induction was induced in mice by 5 oral feedings of 1 microg HBV antigens or HCC-extracted proteins ( 50 microg protein ) before vaccination with recombinant HBsAg .\n\nSplenocytes ( 2 x 10(6) ) from these mice were transferred to sublethally irradiated athymic BALB/c mice previously transplanted subcutaneously with 10(7) human hepatoma Hep3B cells .\n\nAdoptive transfer of splenocytes immunized toward HBsAg prevented tumor growth .\n\nAt 4 weeks after splenocyte transplantation , tumor volume and serum alpha-fetoprotein ( AFP ) levels in athymic mice transplanted with splenocytes immunized to HBsAg were undetectable as compared with 1,048 +/- 738 mm(3) and 2,500 +/- 1,431 ng/ml in recipients of na\ufffdve splenocytes ( p < 0.0001 ) .\n\nMice receiving splenocytes tolerized toward Hep3B cells , as manifested by reduced serum HBs antibody levels , reduced HBV-specific stimulation index and reduced HBV-specific-IFN gamma spot-forming cells , had early tumor growth evident by elevated AFP serum levels , weight loss and mortality , which were suppressed at 6 weeks .\n\nMice transplanted with splenocytes tolerized toward HBV antigens did not have direct evidence of tumor growth .\n\nInduction of oral tolerance toward HCC-extracted proteins enabled transient tumor growth in this model .\n\nThis effect was mediated through downregulation of the anti-HBV immune response .", "output": "Avoiding immune destruction" }, { "input": "The secretion of immunosuppressive factors like transforming growth factor-beta ( TGF-beta ) by tumor cells has been recognized as one of the mechanisms involved in tumor immunological escape .\n\nThis study aimed to examine whether dendritic cell ( DC ) immunization could reverse TGF-beta-induced immunosuppression by simulating the in vivo interaction among infused DCs , host T cells , and tumor-secreted TGF-beta in an in vitro study .\n\nWe found that both immature and mature DCs were relatively resistant to TGF-beta .\n\nThe addition of TGF-beta to naive human CD4+ T cells , which are required by genetically modified DC to elicit antitumor immunity , resulted in their hyporesponsiveness to DC stimulation in a dose-dependent manner .\n\nWhen activated by allogeneic DCs in the presence of TGF-beta , CD4+ T cells displayed a reduced capacity to proliferate .\n\nMore importantly , activated CD4+ T cells induced by DC stimulation were very sensitive to TGF-beta , and this susceptibility was enhanced by their previous exposure to TGF-beta .\n\nThe underlying mechanism was linked to TGF-beta-induced apoptosis of activated T cells .\n\nHowever , the presence of stimulation from DC or antibodies to CD3 plus CD28 could partly reverse the immunosuppressive effect of TGF-beta on activated CD4+ T cells .\n\nTaken together , our results indicate that the efficacy of DC immunization may be impaired by tumor-derived TGF-beta .", "output": "Sustaining proliferative signaling, Avoiding immune destruction, Resisting cell death" }, { "input": "To characterize the impact of increased production of TGF-beta in a xenograft model of human breast cancer , TGF-beta-responsive MDA-231 cells were genetically modified by stable transfection so as to increase their production of active TGF-beta1 .\n\nCompared with control cells , cells that produced increased amounts of TGF-beta proliferated in vitro more slowly .\n\nIn vivo , however , tumors derived from these cells exhibited increased proliferation and grew at an accelerated pace .\n\nTo evaluate the role of autocrine TGF-beta signaling , cells were also transfected with a dominant-negative truncated type II TGF-beta receptor ( TbetaRII ) .\n\nDisruption of autocrine TGF-beta signaling in the TGF-beta-overexpressing cells reduced their in vivo growth rate .\n\nCo-inoculation of Matrigel with the TGF-beta-overexpressing cells expressing the truncated TbetaRII compensated for their diminished in vivo growth capacity , compared with the TGF-beta-overexpressing cells with an intact autocrine loop .\n\nTissue invasion by the tumor was a distinctive feature of the TGF-beta-overexpressing cells , whether or not the autocrine loop was intact .\n\nFurthermore , tumors derived from TGF-beta-overexpressing cells , irrespective of the status of the autocrine TGF-beta-signaling pathway , had a higher incidence of lung metastasis .\n\nConsistent with the suggestion that TGF-beta's enhancement of invasion and metastasis is paracrine-based , we observed no significant differences among the cell clones in an in vitro invasion assay .\n\nThus , in this experimental model system in vitro assays of cell proliferation and invasion do not accurately reflect in vivo observations , perhaps due to autocrine and paracrine effects of TGF-beta that influence the important in vivo-based phenomena of tumor growth , invasion , and metastasis .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "The involvement of PRL in regulating monocyte/macrophage functions is suggested by the presence of PRL-Rs in these cells .\n\nHere , we show that PRL , though it failed to activate mouse peritoneal resident macrophages ( RMs ) , acted as a second signal and activated mouse peritoneal inflammatory macrophages ( EMs ) to a tumoricidal state .\n\nThe cytotoxicity of mouse tumor-associated macrophages ( TAMs ) isolated at day 1 of tumor ( Ehrlich ascites carcinoma , EAC ) growth was enhanced by PRL .\n\nHowever , with progression of tumor growth , TAMs became nonresponsive to the hormone .\n\nPRL-induced killing of P815 target cells by EMs and TAMs was independent of TNF but correlated with the hormone-induced augmentation of NO2(-) and O2(-) release in these macrophages .\n\nAdministration of PRL in vivo inhibited EAC growth and augmented NO2(-) release by TAMs .\n\nPRL synergized with the TH1 cytokine IFN-gamma , a known activator of macrophages , in inducing tumor killing and release of NO2(-) from EMs and TAMs .\n\nThe hormone might activate macrophages at least partially , through the release of IFN-gamma as anti-IFN-gamma blocked IFN-gamma- as well as PRL-induced cytotoxicity in EMs .\n\nThe TH2 cytokine IL-4 suppressed PRL-induced activation of macrophages .\n\nPRL induced release of IL-12 from EMs also , which suggested that the hormone might drive the TH1 response through IL-12 .\n\nOur observations further suggest that PRL alone and in synergy with IFN-gamma , released through induction of IL-12 , may generate tumoricidal macrophages and thus regulate the antitumor immune response of tumor hosts .", "output": "Tumor promoting inflammation" }, { "input": "Primary fibroblasts respond to activated H-RAS(V12) by undergoing premature arrest , which resembles replicative senescence .\n\nThis irreversible ' fail-safe mechanism ' requires p19(ARF) , p53 and the Retinoblastoma ( Rb ) family : upon their disruption , RAS(V12)-expressing cells fail to undergo senescence and continue to proliferate .\n\nSimilarly , co-expression of oncogenes such as c-MYC or E1A rescues RAS(V12)-induced senescence .\n\nTo identify novel genes that allow escape from RAS(V12)-induced senescence , we designed an unbiased , retroviral complementary DNA library screen .\n\nWe report on the identification of DRIL1 , the human orthologue of the mouse Bright and Drosophila dead ringer transcriptional regulators .\n\nDRIL1 renders primary murine fibroblasts unresponsive to RAS(V12)-induced anti-proliferative signalling by p19(ARF)/p53/p21(CIP1) , as well as by p16(INK4a) .\n\nIn this way , DRIL1 not only rescues RAS(V12)-induced senescence but also causes these fibroblasts to become highly oncogenic .\n\nFurthermore , DRIL1 immortalizes mouse fibroblasts , in the presence of high levels of p16(INK4a) .\n\nImmortalization by DRIL1 , whose product binds the pRB-controlled transcription factor E2F1 ( ref. 8 ) , is correlated with induction of E2F1 activity .\n\nCorrespondingly , DRIL1 induces the E2F1 target Cyclin E1 , overexpression of which is sufficient to trigger escape from senescence .\n\nThus , DRIL1 disrupts cellular protection against RAS(V12)-induced proliferation downstream of the p19(ARF)/p53 pathway .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "Oncostatin M ( OSM ) , an interleukin-6 type cytokine , acts via the gp130 signaling receptor to inhibit proliferation and induce differentiation of breast cancer cells .\n\nEGF , a mitogen for breast cells , signals via EGFR/ErbB tyrosine kinase receptors which are implicated in breast cancer pathogenesis .\n\nHere we show paradoxically that EGF enhanced the OSM-induced inhibition of proliferation and induction of cellular differentiation in both estrogen receptor positive and negative breast cancer cells .\n\nThis functional synergism was also seen with heregulin but not SCF , PDGF or IGF-1 , indicating that it was specific to EGF-related growth factors .\n\nImmunoprecipitation experiments revealed that gp130 was constitutively associated with ErbB-2 and ErbB-3 .\n\nThere was a similar association between the OSMRbeta and ErbB-2 .\n\nFurthermore , EGF unexpectedly induced tyrosine phosphorylation of gp130 .\n\nWe show that OSM induced phosphorylation of STAT3 .\n\nBoth OSM and EGF activated the p42/44 MAP kinases , but while the MEK inhibitor , PD98059 , ablated the OSM-induced inhibition , it only partially ablated the inhibitory effects of OSM plus EGF .\n\nThus , we have demonstrated that the receptors and signalling pathways of two apparently unrelated growth factors were intimately linked , resulting in an unexpected biological effect .\n\nThis provides a new mechanism for generating signalling diversity and has potential clinical implications in breast cancer .", "output": "Sustaining proliferative signaling" }, { "input": "p53 is a tumor suppressor gene that is mutated in many human malignancies , including gastric cancer .\n\nIt remains unclear why patients with germ-line p53 mutations ( i.e. , Li-Fraumeni syndrome ) are not at increased risk for gastric adenocarcinoma , despite the fact that they show a high rate of many other tumors .\n\nFurthermore , the precise relationship between germ-line p53 mutations and the response to chronic bacterial infections ( such as Helicobacter spp. ) has not been investigated .\n\nTo assess the role of germ-line p53 deletions in modulating the progression to gastric cancer , p53(+/-) and wild-type ( WT ) C57BL/6 mice were infected with H. felis .\n\nThe gastric pathology and immune response in these two groups of mice were analyzed for up to 15 months postinfection .\n\nThe gastric fundus and antrum were evaluated independently using a 0-4 scale to score inflammation , parietal and chief cell loss , mucus metaplasia , and helicobacter colonization .\n\nNonparametric statistical analysis was performed to determine the effects of p53(+/-) , infection status , and postinoculation ( p.i. ) time on inflammation , preneoplastic changes , invasive lesions , and helicobacter colonization. mRNA expression for gammaIFN , interleukin ( IL)-1 , IL-10 , and IL-4 was quantified by PCR .\n\nSera were also evaluated for H. felis antibody by ELISA .\n\nAntral inflammation increased significantly with time in infected mice .\n\nThere was a significant , protective effect on the development of preneoplastic fundic lesions and invasive carcinoma attributable to the deletion of one p53 allele ( P < 0.05 ) .\n\nSubmucosal invasive foci were observed in 9 of 11 WT-infected mice ranging from 13 to 15 months p.i. ; invasion of adjacent submucosal blood vessels by glandular epithelia also was present in 5 of these mice .\n\nNone of these lesions were observed in 33 p53(+/-) mice , infected or not , at any time p.i. p53(+/-) mice had significantly higher helicobacter colonization consistent with a Th2 host response .\n\nIn sera from WT mice , IgG2a , considered a proinflammatory Th1 response , continued to rise throughout the 15-month study ( P < 0.004 ) .\n\nIn contrast , IgG2a levels of the p53(+/-) mice were 50-60% lower than those of the WT mice at each time point ( P range , <0.012 to 0.002 ) and did not progress in magnitude between 12 and 15 months of chronic H. felis infection ( P = 0.167). mRNA levels for gammaIFN and IL-1 were significantly up-regulated in WT mice infected with H. felis ( P < 0.05 ) but were slightly elevated or were at background levels in p53(+/-) mice .\n\nIL-10 and IL-4 mRNA expression was not significantly different from control samples .\n\nOur results support the hypothesis that germ-line deletion of one p53 allele results in a down-regulated Th1 response to gastric helicobacter infection , possibly because of T-cell senescence , which may indirectly protect against the development of gastric cancer and other epithelial-derived neoplasms associated with chronic inflammation .", "output": "Tumor promoting inflammation, Enabling replicative immortality, Activating invasion and metastasis" }, { "input": "Cellular senescence forms a barrier that inhibits the acquisition of an immortal phenotype , a critical feature in tumorigenesis .\n\nThe inactivation of multiple pathways that positively regulate senescence are required for immortalization .\n\nTo identify these pathways in an unbiased manner , we performed DNA microarray analyses to assess the expression of 20,000 genes in human prostate epithelial cells ( HPECs ) passaged to senescence .\n\nThese gene expression patterns were then compared with those of HPECs immortalized with the human Papillomavirus 16 E7 oncoprotein .\n\nSenescent cells display gene expression patterns that reflect their nonproliferative , differentiated phenotype and express secretory proteases and extracellular matrix components .\n\nA comparison of genes transcriptionally up-regulated in senescence to those in which expression is significantly down-regulated in immortalized HPECs identified three genes : the chemokine BRAK , DOC1 , and a member of the insulin-like growth factor axis , IGFBP-3 .\n\nExpression of these genes is found to be uniformly lost in human prostate cancer cell lines and xenografts , and previously , their inactivation was documented in tumor samples .\n\nThus , these genes may function in novel pathways that regulate senescence and are inactivated during immortalization .\n\nThese changes may be critical not only in allowing cells to bypass senescence in vitro but in the progression of prostate cancer in vivo .", "output": "Enabling replicative immortality" }, { "input": "Overexpression and amplification of cyclin D1 were investigated by immunohistochemistry and differential polymerase chain reaction ( dPCR ) in 440 formalin-fixed primary breast carcinoma tissues .\n\nOverexpression of cyclin D1 was detected in 60% ( 263/440 ) and amplification of cyclin D1 was noted in 27% ( 119/440 ) of the primary breast carcinomas .\n\nMolecular analysis demonstrated that cyclin D1 was amplified in 30% ( 7/23 ) of the comedo DCIS , 22% ( 9/41 ) of the comedo DCIS and 32% ( 13/41 ) of the adjacent invasive ductal carcinomas , 30% ( 82/270 ) of the invasive ductal carcinomas , 27% ( 9/33 ) of the invasive lobular carcinomas , 19% ( 4/21 ) of the colloid carcinomas and 13% ( 2/15 ) of the medullary carcinomas .\n\nCyclin D1 was amplified in 11% ( 2/19 ) of the invasive ductal carcinomas but not in the adjacent non-comedo DCIS lesions .\n\nOur observation showed that cyclin D1 was strongly positive in 61% ( 14/23 ) of the comedo subtype , 61% ( 11/18 ) of the non-comedo subtype , 59% ( 24/41 ) of the comedo DCIS and 63% ( 26/41 ) of the adjacent invasive ductal carcinomas , 53% ( 10/19 ) of the non-comedo DCIS and 58% ( 11/19 ) of the adjacent invasive lesions , 58% ( 157/270 ) of the invasive ductal carcinomas , 73% ( 24/33 ) of the invasive lobular carcinomas , 52% ( 11/21 ) of the colloid carcinomas and 27% ( 4/15 ) of the medullary carcinomas .\n\nA significant association was observed between in situ components and adjacent invasive lesions for cyclin D1 expression ( p<0.05 ) and amplification ( p<0.05 ) .\n\nA significant relationship was noted between amplification of cyclin D1 and lymph node metastases ( p<0.05 ) but not with histological grade ( p>0.05 ) , estrogen receptor status ( p>0.05 ) and proliferation index ( Ki-67 and PCNA ) ( p>0.05 ) .\n\nHowever , overexpression of cyclin D1 was statistically associated with well differentiated tumors ( p<0.05 ) and estrogen receptor positivity ( p<0.05 ) .\n\nNo relationship was seen with nodal status ( p>0.05 ) and proliferation index ( Ki-67 and PCNA ) ( p>0.05 ) .\n\nThese observations suggest that tumors positive for cyclin D1 protein may have features of good prognosis but amplification of cyclin D1 gene could be an indicator of tumors with poor prognostic features .\n\nAlthough majority of the Malaysian patients belong to younger age group ( <50 years old ) , amplification and expression of cyclin D1 was not statistically associated with patient age ( p>0.05 ) .\n\nThese observations indicate that amplification and up-regulation of cyclin D1 may be independent of patient age .\n\nMoreover , overexpression and amplification of cyclin D1 in preinvasive , preinvasive and adjacent invasive lesions , and invasive carcinomas suggest that the gene may play an important role in early and late stages of breast carcinogenesis .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Interferon-gamma ( IFN-gamma ) has pleiotropic activities other than its antivirus action , including cell growth inhibition , natural killer ( NK ) cell and cytotoxic T lymphocyte ( CTL ) activation , and angiogenesis inhibitory activity , and these activities are supposed to be involved in its antitumour activity .\n\nHowever , it has not been completely elucidated which activity is mainly involved in the tumour suppression in vivo .\n\nIn this study , we analysed inhibitory mechanisms of endogenous IFN-gamma against B16 melanoma experimental metastasis .\n\nAfter intravenous injection of tumour cells , tumour deposits in the lungs and liver were increased and life span was shorter in IFN-gamma(-/-) mice , indicating important roles for IFN-gamma in antitumour mechanisms .\n\nInterestingly , tumour deposits were not increased in IFN-gamma receptor ( R)(-/- ) mice .\n\nFurthermore , only low levels of cell-mediated immunity against the tumour and activation of NK cells were observed , indicating that antimetastatic effects of IFN-gamma is not mediated by host cells .\n\nThe survival period of B16 melanoma-bearing IFN-gamma R(-/-) mice was , however , shorter than wild-type mice .\n\nThese observations suggest that IFN-gamma prevents B16 melanoma experimental metastasis by directly inhibiting the cell growth , although antitumour host functions may also be involved in a later phase .", "output": "Activating invasion and metastasis" }, { "input": "P53 activity plays a key role in mammalian cells when they undergo replicative senescence at their Hayflick limit .\n\nTo determine whether p63 proteins , members of the family of p53-related genes , are also involved in this process , we examined their expression in serially passaged rat embryo fibroblasts .\n\nUpon senescence , two truncated DeltaNp63 proteins decreased in abundance whereas two TAp63 isoforms accumulated. 2-D gel analysis showed that the DeltaNp63 proteins underwent post-translational modifications in both proliferating and senescent cells .\n\nDirect binding of DeltaNp63 proteins to a p53 consensus motif was greater in proliferating cells than senescent cells .\n\nIn contrast p63alpha isoforms bound to DNA in a p53 dependent manner and this was higher in senescent cells than proliferating cells .\n\nAn interaction of p63alpha proteins with SV40 large tumour antigen was also detected and ectopic expression of DeltaNp63alpha can extend the lifespan of rat embryo fibroblasts .\n\nTaken together the results indicate that p63 proteins may play a role in replicative senescence either by competition for p53 DNA binding sites or by direct interaction with p53 protein bound to DNA .", "output": "Enabling replicative immortality" }, { "input": "Mesotheliomas are malignant tumors of the pleural and peritoneal membranes which are often associated with asbestos exposure and with Simian virus 40 ( SV40 ) infection .\n\nTelomerase activity is repressed in somatic cells and tissues but is activated in immortal and malignant cells .\n\nWe evaluated telomerase activity in seven primary malignant mesothelioma biopsies and matched lung specimens and 20 mesothelioma cell lines and eight corresponding primary tumor cultures .\n\nAll the tumor biopsies , and nearly all primary cell mesothelioma cultures and cell lines were telomerase positive .\n\nThe findings in cell lines paralleled those observed in primary cultures in cases where paired samples were available .\n\nNext , we found that SV40 , a DNA tumor virus present in approximately 50% of mesothelioma biopsies in the USA , induced telomerase activity in primary human mesothelial cells , but not in primary fibroblasts .\n\nTelomerase activity became detectable as early as 72 h following wild-type ( strain 776 ) SV40 infection , and a clear DNA ladder was detectable 1 week after infection .\n\nThe amount of telomerase activity increased during passage in cell culture and appeared to parallel increases in the cellular amounts of the SV40 large T-antigen .\n\nThus , SV40 infection leads to telomerase activity before the infected mesothelial cells become transformed and immortalized .\n\nSV40 infection of human fibroblasts did not cause detectable telomerase activity .\n\nWe also determined that the SV40 small t-antigen ( tag ) plays an important role in inducing telomerase activity because this activity was undetectable or minimal in mesothelial cells infected and/or transformed by SV40 tag mutants .\n\nAsbestos alone did not induce telomerase activity , and asbestos did not influence telomerase activity in mesothelial cells infected with SV40 .\n\nInduction of telomerase activity by SV40 may be related to the very high rate of mesothelial cell immortalization that is characteristically associated with SV40 infection of mesothelial cells .", "output": "Enabling replicative immortality" }, { "input": "Vascular endothelial growth factor ( VEGF ) has emerged as one of the most important angiogenic growth factors from experimental in vitro and in vivo studies .\n\nIn the present study , we investigated the relationship between VEGF expression and microvessel density ( MVD ) and defined their prognostic relevance on a series of 242 patients with node-negative breast cancer , using immunohistochemical methods .\n\nIn parallel , estrogen and progesterone receptors were quantitatively assessed using the dextran-charcoal technique and cell proliferation was evaluated as S-phase cell fraction according to ( 3)H-thymidine-labeling index ( TLI ) .\n\nThe percentage of VEGF-expressing cells varied from 0-95% in the different tumors and was unrelated to menopausal status , tumor size or steroid receptor status .\n\nConversely , a significant inverse relation was observed with patient age or tumor cell proliferation , albeit with very poor correlation coefficients .\n\nA significant relation was observed between VEGF expression and MVD ( r(s) = 0.55 , p < 0.001 ) .\n\nClinical outcome analyzed as a function of high and low VEGF expression showed slight differences in terms of both disease-free survival ( DFS ) and overall survival ( OS ) that never reached statistical significance .\n\nMoreover , the trend was paradoxically in favor of patients with highly VEGF-expressing tumors .\n\nFinally , DFS and OS curves , when analyzed as a function of VEGF expression or MVD , were superimposable .\n\nIn conclusion , our study did not highlight a prognostic relevance of VEGF expression in patients with node-negative breast cancer , as already observed for MVD .", "output": "Inducing angiogenesis" }, { "input": "PURPOSE We recently reported that overexpression of epidermal growth factor receptor ( EGFR ) positively correlated with radioresistance of murine carcinomas .\n\nBecause cyclin D1 is a downstream sensor of EGFR activation , the present study investigated whether a relationship exists between the extent of cyclin D1 expression and in vivo radiocurability of murine tumors .\n\nWe further investigated the influence of radiation on cyclin D1 expression and the expression of p27 , an inhibitor of the cyclin D1 downstream pathway , as well as the relationship of these molecular determinants to cell proliferation and induced apoptosis in tumors exposed to radiation .\n\nMETHODS AND MATERIALS Cyclin D1 expression was assayed in nine carcinomas syngeneic to C3Hf/Kam mice using Western blot analysis .\n\nThese tumors greatly differed in their radioresponse as assessed by TCD(50) .\n\nThe expression of cyclin D1 and p27 proteins was determined by Western blotting .\n\nCell proliferative activity in tumors was determined by proliferating cell nuclear antigen ( PCNA ) immunochemistry .\n\nThe effect of irradiation on the expression of cyclin D1 or p27 proteins and on PCNA positivity was determined in the radiosensitive OCa-I and in the radioresistant SCC-VII tumors .\n\nRESULTS Cyclin D1 expression varied among tumors by 40-fold , and its magnitude positively correlated with poorer tumor radioresponse ( higher TCD(50) values ) .\n\nThe level of cyclin D1 expression paralleled that of EGFR .\n\nA 15-Gy dose reduced constitutive expression of cyclin D1 in the radiosensitive OCa-I tumors , but had no influence on expression of cyclin D1 in the radioresistant SCC-VII tumors .\n\nIn contrast , 15 Gy increased the expression of p27 in radiosensitive tumors and reduced it in radioresistant tumors .\n\nRadiation induced no significant apoptosis or change in the percentage of PCNA-positive ( proliferating ) cells in SCC-VII tumors with high cyclin D1 levels , but it induced significant apoptosis and a decrease in the percentage of proliferating cells in OCa-I tumors with low cyclin D1 expression .\n\nCONCLUSION Our findings show a positive correlation between cyclin D1 expression and tumor radioresistance .\n\nThe expression of cyclin D1 and p27 was modified by radiation and was associated with cellular response to radiation , but this depended on the pretreatment level of cyclin D1 expression .\n\nThese findings may have important clinical implications : The pretreatment assessment of cyclin D1 expression could serve as a useful predictor of radiotherapy outcome and assist in selecting an effective treatment modality .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Treatment of cells with the anti-cancer drug camptothecin ( CPT ) induces topoisomerase I ( Top1)-mediated DNA damage , which in turn affects cell proliferation and survival .\n\nIn this report , we demonstrate that treatment of the wild-type HCT116 ( wt HCT116 ) human colon cancer cell line and the isogenic p53(-/-) HCT116 and p21(-/-) HCT116 cell lines with a high concentration ( 250 nm ) of CPT resulted in apoptosis , indicating that apoptosis occurred by a p53- and p21-independent mechanism .\n\nIn contrast , treatment with a low concentration ( 20 nm ) of CPT induced cell cycle arrest and senescence of the wt HCT116 cells , but apoptosis of the p53(-/-) HCT116 and p21(-/-) HCT116 cells .\n\nFurther investigations indicated that p53-dependent expression of p21 blocked apoptosis of wt HCT116 cells treated with 20 nm , but not 250 nm CPT .\n\nInterestingly , blocking of the apoptotic pathway , by Z-VAD-FMK , in p21(-/-) HCT116 cells following treatment with 20 nm CPT did not permit the cells to develop properties of senescence .\n\nThese observations demonstrated that p21 was required for senescence development of HCT116 cells following treatment with low concentrations of CPT .", "output": "Sustaining proliferative signaling, Enabling replicative immortality, Resisting cell death" }, { "input": "OBJECTIVE To elucidate the direct effects of gonadotropin-releasing hormone agonist ( GnRHa ) on the growth of human uterine leiomyoma cells , cell proliferation and apoptosis in cultured leiomyoma cells treated with GnRHa were investigated .\n\nMETHODS Isolated leiomyoma cells were subcultured in DMEM supplemented with 10% FBS for 5 days and stepped down to serum-free conditions for an additional 6 days in the presence or absence of graded concentrations of GnRHa ( 10(-9) mol/l to 10(-12) mol/l ) .\n\nThe effects of GnRHa on the number of viable cells , expression of proliferating cell nuclear antigen ( PCNA ) , Fas and Fas ligand , and apoptosis in cultured leiomyoma cells were examined by MTT ( 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrasodium bromide ) assay , immunocytochemical analysis , Western blot analysis and TUNEL assay respectively .\n\nRT-PCR was performed to detect the expression of GnRH receptor mRNA in cultured leiomyoma cells .\n\nRESULTS Treatment with GnRHa resulted in a decrease in the number of cultured viable leiomyoma cells assessed by MTT assay in a dose-dependent manner compared with that in control cultures ( P<0.01 ) .\n\nThe growth inhibition of cultured leiomyoma cells treated with GnRHa in concentrations higher than 10(-10) mol/l was associated with the suppression of the proliferative potential characterized by a decrease in PCNA-positive rate of the cultured cells ( P<0.01 ) and an increase in the apoptosis-positive rate assessed by TUNEL assay ( P<0.05 and P<0.01 ) .\n\nGnRHa markedly increased the expression of Fas and induced the expression of Fas ligand in the cultured leiomyoma cells on the basis of Western blot analysis .\n\nThese direct effects of GnRHa on the number of viable cultured leiomyoma cells , PCNA-positive rate , apoptosis-positive rate and Fas/Fas ligand expression in the cultured leiomyoma cells were only attained after the 4-day treatment .\n\nRT-PCR analysis revealed that GnRH receptor mRNA was expressed in cultured leiomyoma cells .\n\nCONCLUSIONS The present results demonstrate that GnRHa directly inhibits the growth of human uterine leiomyoma cells by suppressing cell proliferation and inducing apoptosis , which might be associated with the increase in Fas expression and the induction of Fas ligand expression in the cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "AIMS Tumour cell growth results from a disturbance in the balance between the rate of proliferation and cell death .\n\nIn this study , proteins involved in the regulation of cell cycle arrest and apoptosis were studied as possible factors responsible for uncontrolled cell growth in colorectal cancer .\n\nMETHODS The expression of proteins involved in these processes was investigated in 48 metastases from patients with colorectal cancer and compared with eight normal colon mucosa samples and 14 primary tumours .\n\nBoth primary tumours and metastases were obtained from eight patients .\n\nThe expression of thymidylate synthase ( TS ) , p53 , retinoblastoma protein ( Rb ) , Fas receptor , Fas ligand , bcl-2 , mcl-1 , bax , and bcl-x was measured using immunohistochemistry .\n\nProliferation was determined by Ki67 staining , whereas apoptosis was assessed by M30 immunostaining , which recognises cleaved cytokeratin 18 .\n\nRESULTS In the limited number of cases in which paired comparisons were possible , the expression of TS and Ki67 was significantly higher in metastases than in the matched primary tumour samples ( p = 0.014 and 0.016 , respectively ) , whereas Rb expression was lower in metastases than in primary tumours ( p = 0.024 ) .\n\nFas receptor expression was high in normal mucosa but absent in primary tumours and metastases , whereas the opposite was seen for p53 .\n\nThe expression of bax , mcl-1 , and bcl-x in normal mucosa was more apical than that seen in malignant cells , where a more diffuse expression pattern was seen ( p < 0.04 ) .\n\nApoptosis was more abundant in primary tumours than in metastases .\n\nCONCLUSIONS These results demonstrate that proliferation and apoptosis are disturbed during colorectal cancer progression , and this is accompanied by loss of Rb and Fas expression , the accumulation of p53 and TS , and changes in the expression patterns of bax , mcl-1 , and bcl-xl .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "It is well known that cell-mediated immunity is suppressed in patients with neoplastic diseases .\n\nWe have reported that soluble receptors for interleukin-2 ( sIL-2R ) and tumor necrosis factor ( sTNF-R1 ) are elevated in the serum of patients with advanced colorectal cancer .\n\nThe presence of these soluble receptors and immunosuppressive cytokines , including interleukin-10 ( IL-10 ) , might be important in the mechanisms of immunosuppression. cis-Diaminedichloroplatinum ( cisplatin ) has been reported to immunomodulate , especially when used in low dose in combination with 5-Fluorouracil ( 5-FU ) .\n\nIn this study , cisplatin and UFT , a form of uracil and tegafur which is a prodrug of 5-FU , were administered with immunomodulator Polysaccharide K ( PSK ) to ten patients with colorectal cancer , who showed distant metastasis in the liver or lung , and the serum levels of sIL-2R and sTNF-R1 and the production of gamma-interferon ( gamma-INF ) and interleukin-10 by peripheral blood mononuclear cells were measured .\n\nThe serum concentrations of sIL-2R and the production of IL-10 were reduced ( p < 0.05 ) after 2 months of treatment .\n\nThus , this combination appeared to have immunomodulative potential in patients with advanced colorectal cancer .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "Often the use of cytotoxic drugs in cancer therapy results in stable disease rather than regression of the tumor , and this is typically seen as a failure of treatment .\n\nWe now show that DNA damage is able to induce senescence in tumor cells expressing wild-type p53 .\n\nWe also show that cytotoxics are capable of inducing senescence in tumor tissue in vivo .\n\nOur results suggest that p53 and p21 play a central role in the onset of senescence , whereas p16(INK4a) function may be involved in maintaining senescence .\n\nThus , like apoptosis , senescence appears to be a p53-induced cellular response to DNA damage and an important factor in determining treatment outcome .", "output": "Genomic instability and mutation, Enabling replicative immortality, Resisting cell death" }, { "input": "Senescence limits the proliferative capacity of primary cells in culture .\n\nWe describe here a genetic screen to identify genes that allow bypass of this checkpoint .\n\nUsing retroviral cDNA expression libraries , we identify BCL6 as a potent inhibitor of senescence .\n\nBCL6 is frequently activated in non-Hodgkin's lymphoma , but its mechanism of action has remained unclear .\n\nBCL6 efficiently immortalizes primary mouse embryonic fibroblasts and cooperates with RAS in oncogenic transformation .\n\nBCL6 overrides the senescence response downstream of p53 through a process that requires induction of cyclin D1 expression , as cyclin D1 knockout fibroblasts are specifically resistant to BCL6 immortalization .\n\nWe show that BCL6 expression also dramatically extends the replicative lifespan of primary human B cells in culture and induces cyclin D1 expression , indicating that BCL6 has a similar activity in lymphoid cells .\n\nOur results suggest that BCL6 contributes to oncogenesis by rendering cells unresponsive to antiproliferative signals from the p19(ARF)-p53 pathway .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "Proliferator-activated receptor gamma ( PPARgamma ) is a nuclear receptor , which mainly associates with adipogenesis , but also appears to facilitate cell differentiation or apoptosis in certain malignant cells .\n\nThis apoptosis induction by PPARgamma is increased by co-stimulation with tumor necrosis factor ( TNF)-alpha-related apoptosis-inducing ligand ( TRAIL ) , a member of the TNF family .\n\nIn this study , we investigated the effect of PPARgamma on Fas-mediated apoptosis in hepatocellular carcinoma ( HCC ) cell lines .\n\nPPARgamma was expressed on all seven HCC cell lines and located in their nuclei. 15-Deoxy-Delta-12,14-prostaglandin J2 ( 15d- PGJ2 ) , a PPARgamma ligand , inhibited cellular proliferation in HepG2 , SK-Hep1 or HLE cells , unlike pioglitazone , another PPARgamma ligand , which did not have a significant influence on proliferation of these cells .\n\nHowever , 15d-PGJ2 facilitated Fas-mediated HCC apoptosis that could not be induced by Fas alone .\n\nThese results suggest that PPARgamma can augment TNF-family-induced apoptosis .", "output": "Resisting cell death" }, { "input": "We have used a combination of vitamin A ( all-trans-retinyl palmitate ) , 5-fluorouracil ( 5-FU ) and radiation to treat human head and neck squamous cell carcinoma ( HNSCC ) .\n\nThis chemoradiotherapy is called \" FAR therapy. \"\n\nIn this study we examined the effects of all-trans-retinoic acid ( ATRA ) , the active metabolite of vitamin A , and ATRA plus 5-FU on two HNSCC cell lines ( YCU-N861 and YCU-H891 ) to gain insight into the molecular mechanisms of FAR therapy .\n\nATRA at 1 mM ( the order of concentration found in HNSCC tumors treated with FAR therapy ) inhibited cell proliferation and caused G1 cell cycle arrest in both cell lines .\n\nThis was associated with a decrease in cyclin D1 , an increase in p27(Kip1) and a reduction in the hyperphosphorylated form of retinoblastoma protein ( pRB ) .\n\nWith YCU-N861 cells , ATRA also caused a decrease in Bcl-2 and Bcl-X(L) and an increase in Bax .\n\nBoth ATRA and 5-FU activated c-Jun N-terminal kinase ( JNK ) 1 and the combination of both agents resulted in additive or synergistic activation of JNK1 , and also enhanced the induction of apoptosis .\n\nThe YCU-H891 cells , in which the epidermal growth factor receptor ( EGFR)-signal transducer and activator of transcription 3 ( Stat3 ) pathway is constitutively activated , were more resistant to treatments with ATRA , 5-FU and the combination of both agents than YCU-N861 cells .\n\nA dominant negative Stat3 construct strongly enhanced the cellular sensitivity of this cell line to 5-FU but not to ATRA .\n\nIn addition there is evidence that activation of Stat3 is associated with cellular resistance to radiation in HNSCC .\n\nTherefore , the addition to FAR therapy of agents that inhibit activation of the Stat3 pathway may enhance the clinical response of patients with HNSCC to FAR therapy .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Inflammatory events have been associated with senile plaques , one of the pathological hallmarks of Alzheimer's disease ( AD ) .\n\nIt is believed that aggregated beta-amyloid ( betaA ) proteins , which form the core of these plaques , may be responsible for triggering the inflammatory reaction .\n\nIn the present study , the ability of aluminum ( Al ) to initiate similar inflammatory events was investigated in a human glioblastoma cell line .\n\nA 6-day exposure to either lipopolysaccharide ( LPS ) or aluminum sulfate caused a significant increase in the rate of proliferation of the glioblastoma cells .\n\nBoth treatments also caused activation of the immune-responsive transcription factor NF-kappaB although there were time-related differences .\n\nThe levels of secreted cytokines , interleukin-6 ( IL-6 ) and tumor necrosis factor alpha ( TNF-alpha ) were both increased by the LPS treatment although exposure to Al decreased the secretion of the former while elevating the levels of the latter .\n\nThese events may be due to the activation of glial cells and subsequent stress response to either Al complexes or LPS .\n\nAlthough exposure to either stress factor caused a stimulation of inflammatory markers , there were time-dependent differences in the response .\n\nThis may reflect the ability of the cells to discern different stress factors and thus orchestrate an innate immune response profile distinct to each immunogen .", "output": "Tumor promoting inflammation" }, { "input": "Telomerase is a ribonucleoprotein enzyme that functions to maintain telomeres , the terminal DNA that protects chromosomal integrity , regulating cellular replicative life span .\n\nTelomerase is not expressed in most normal human somatic cells but is active in stabilizing telomeres of certain self-renewing cell populations and most malignant cells , making the enzyme an appealing target for anticancer therapy .\n\nWe describe here a novel cross-species approach to telomerase inhibition .\n\nEctopic expression of the human telomerase catalytic reverse transcriptase component in murine cells inhibited endogenous murine telomerase activity .\n\nUsing this approach , telomerase inhibition in immortal murine fibroblasts resulted in critical telomere shortening , leading to slowed proliferation , abnormal morphology , altered cell cycle , and telomere dysfunction with cytogenetic instability , followed by apoptotic cell death .\n\nSubpopulations of two telomerase-inhibited clones escaped widespread apoptosis , showing proliferative recovery in culture despite persistently inhibited telomerase activity with progressive telomere shortening and dysfunction .\n\nThis study , by targeting immortal murine cells for telomerase inhibition , demonstrates the importance of telomerase to murine cell immortalization and telomere maintenance .\n\nMoreover , the murine model used here should prove useful in further evaluating telomerase inhibition as an anticancer therapy .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "BACKGROUND Helicobacter pylori is the main risk factor for the development of non-cardia gastric cancer .\n\nIncreased proliferation of the gastric mucosa is a feature of H. pylori infection .\n\nMucosal interkeukin-1beta production is increased in H. pylori infection and IL-1beta genotypes associated with increased pro-inflammatory activity are risk factors for the development of gastric cancer .\n\nThe effect of IL-1beta on gastric epithelial cell proliferation has been examined in this study .\n\nMETHODS AGS cells were cultured with IL-1beta .\n\nDNA synthesis was assed by [ 3H]thymidine incorporation and total viable cell numbers by MTT assay .\n\nRESULTS IL-1beta dose dependently increased DNA synthesis and cell numbers .\n\nThe enhanced proliferation was blocked by interleukin-1 receptor antagonist .\n\nAddition of neutralising antibody to GM-CSF reduced IL-1beta-stimulated proliferation by 31 +/- 4 % .\n\nGM-CSF alone significantly stimulated proliferation .\n\nAddition or neutralisation of IL-8 had no effect on basal or IL-1beta-stimulated proliferation .\n\nThe tyrosine kinase inhibitor genistein completely blocked IL-1beta-stimulated proliferation and inhibition of the extracellular signal related kinase pathway with PD 98059 inhibited IL-1beta stimulated proliferation by 58 +/- 5 % .\n\nCONCLUSIONS IL-1beta stimulates proliferation in gastric epithelial cells .\n\nAutocrine stimulation by GM-CSF contributes to this proliferative response .\n\nSignalling via tyrosine kinase activity is essential to the mitogenic response to IL-1beta .\n\nThe extracellular signal related kinase pathway is involved in , but not essential to downstream signalling .\n\nIL-1beta may contribute to the hyperproliferation seen in H. pylori- infected gastric mucosa , and be involved in the carcinogenic process .", "output": "Sustaining proliferative signaling" }, { "input": "A rare immunohistochemical study using 28 surgical sections of human chondrosarcoma revealed that 67.9% of tumour cells had weak ( 10-40% ) or strong ( >40% ) positive immunoreaction for peroxisome proliferator-activated receptor-gamma .\n\nThe expression of peroxisome proliferator-activated receptor-gamma mRNA and protein in human chondrosarcoma cell line OUMS-27 was also determined by reverse transcription-polymerase chain reaction and immunocytochemistry , respectively .\n\nFurthermore , the effects of peroxisome proliferator-activated receptor-gamma ligands on cell proliferation and survival were investigated in OUMS-27 cells .\n\nPioglitazone , a selective ligand for peroxisome proliferator-activated receptor-gamma , and 15-deoxy-Delta(12,14)-prostaglandin J(2) ( 15d-PGJ(2) ) , a putative endogenous ligand for peroxisome proliferator-activated receptor-gamma , inhibited the proliferation of OUMS-27 cells in a dose-dependent manner .\n\nThe mechanism of cytotoxic effects of 15d-PGJ(2) was via apoptosis as shown by DNA fragmentation using TUNEL stain and DNA ladder formation , and by ultrastructural analysis using transmission electron microscopy .\n\nFlow-cytometric analysis using annexin-V-fluorescein and propidium iodide detected the early change of apoptosis , as well as necrosis of OUMS-27 cells at 4 h after co-incubation with 15d-PGJ(2) .\n\nThese results suggest that peroxisome proliferator-activated receptor-gamma may play a significant role in the pathogenesis of chondrosarcoma , and peroxisome proliferator-activated receptor-gamma ligands , especially 15d-PGJ(2) , may be of therapeutic value in the treatment of human chondrosarcoma .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Notch signaling controls cell fate decisions of hematopoietic progenitors by inhibiting certain steps of differentiation and inducing either self-renewal or differentiation toward lymphoid or myeloid lineages .\n\nIn addition , truncated Notch1 alleles could be associated with 10% of all cases of human T lymphoblastic leukemia and , when introduced into mouse bone marrow stem cells , cause T-cell neoplasms .\n\nHowever , functional links between the abundant expression of intact Notch1 and oncogenesis are still lacking .\n\nHere we show that Notch1 is highly expressed in B- and T-cell-derived tumor cells of Hodgkin and anaplastic large cell lymphoma .\n\nWe demonstrate a novel mechanism for the oncogenic capacity of Notch1 by showing that the interaction between intact Notch1 on tumor cells and its ligand Jagged1 dramatically induces proliferation and inhibition of apoptosis in vitro .\n\nWe further provide evidence that in Hodgkin and anaplastic large cell lymphoma , Jagged1 is expressed in malignant and in bystander cells colocalizing with Notch1-positive tumor cells .\n\nNotch1 signaling may therefore be activated in tumor cells by Jagged1 through homotypic or heterotypic cell-cell interactions , and it seems likely that these interactions contribute to lymphomagenesis in vivo .\n\nThus , our data suggest that activated Notch1 signaling plays an important role in the pathobiology of Hodgkin and anaplastic large cell lymphoma and that it might be a potential new target for treatment .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "UVB from solar radiation is both an initiating and promoting agent for skin cancer .\n\nWe have found that primary human keratinocytes undergo an apoptotic response to UVB .\n\nTo determine whether these responses are altered during the course of immortalization , we examined markers of apoptosis in primary human foreskin keratinocytes ( HFK ) transduced with either a retroviral vector expressing the E6 and E7 genes of HPV-16 or with empty vector alone ( LXSN-HFK ) .\n\nWhereas LXSN-HFK as well as early passage keratinocytes expressing HPV-16 E6 and E7 ( p7 E6/7-HFK ) were both moderately responsive to UVB irradiation , late passage-immortalized keratinocytes ( p27 E6/7-HFK ) were exquisitely sensitive to UVB-induced apoptosis .\n\nAfter exposure to UVB , enhanced annexin V-positivity and internucleosomal DNA fragmentation were observed in p27 E6/7-HFK compared with either LXSN- or p7 E6/7-HFK .\n\nCaspase-3 fluorometric activity assays as well as immunoblot analysis with antibodies to caspase-3 and poly(ADP-ribose) polymerase revealed elevated caspase-3 activity and processing at lower UVB doses in p27 E6/7-HFK compared with LXSN- or p7 E6/7-HFK .\n\nIn addition , the caspase inhibitor DEVD-CHO reduced the apoptotic response and increased survival of all three HFK types .\n\nImmunoblot analysis revealed that caspase-8 was activated in all three cell types , but caspase-9 was only activated in p27 E6/7-HFK .\n\nCell cycle analysis further showed that only p27 E6/7-HFK exhibit G(2)/M accumulation that is enhanced by UVB treatment .\n\nThis accumulation was associated with a rapid down-regulation of Bcl-2 in these cells .\n\nThe immortalization process subsequent to the expression of HPV E6 and E7 may therefore determine UVB sensitivity by switching the mode of apoptosis from a caspase-8 to a Bcl-2-caspase-9-mediated pathway of apoptosis .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "The in vitro immortalization of primary human mammary epithelial ( HME ) cells solely by the exogenous introduction of the catalytic subunit of human telomerase ( hTERT ) has been achieved .\n\nEarly passage hTERT-transfected HME ( T-HME ) cells continuously decreased the length and density of telomeres even in the presence of telomerase activity , with a significant number of cells staining positive for senescence-associated beta-galactosidase ( SA-beta-gal ) .\n\nSubsequently , with the increase in cell passages , the copy number of the exogenously transfected hTERT gene and the percentage of SA-beta-gal positive cells were found to decrease .\n\nEventually , a single copy of the exogenous hTERT gene was observed in the relatively later passage T-HME cells in which telomere length was elongated and stabilized without obvious activation of endogenous hTERT and c-Myc expression .\n\nIn T-HME cells , the expression of two p53 regulated genes p21(WAF) and HDM2 increased ( as in primary senescent HME cells ) , and was found to be further elevated as the function of p53 was activated by treatment with DNA-damaging agents. p16(INK4a) was shown to be significantly higher in the primary senescent HME and the early passage T-HME cells when compared with the primary presenescent HME cells , with a dramatic repression of p16(INK4a) observed in the later passage T-HME cells .\n\nIn addition , the expression of E2F1 and its transcription factor activity were found to be significantly higher in the later passage T-HME cells when compared with the earlier passage T-HME cells .\n\nTogether , our results indicate that in vitro immortalization in HME cells may require the activation of the function of telomerase and other genetic alterations such as the spontaneous loss of p16(INK4a) expression .", "output": "Enabling replicative immortality" }, { "input": "Prostate cancer ( PCA ) is the most common histological malignancy and the second leading cause of cancer deaths among North American men .\n\nThere has been considerable interest in the chemopreventative properties of selenium .\n\nIn this study , we assessed whether selenium inhibits cell growth and associated cell cycle regulatory proteins .\n\nHuman PCA cells ( LNCaP , PC3 , PC3-AR2 , and PC3-M ) were incubated with and without selenium ( Seleno-DL-methionine , 150 microM ) for 24 , 48 , and 72 h .\n\nCells were fixed and stained with propidium iodide for flow cytometry analysis .\n\nIn parallel experiments , total protein was extracted , immunoprecipitated with cyclin E antibody , and analyzed by Western blot for the expression of cell cycle markers .\n\nTreatment with selenium caused G1 arrest and an 80% reduction in the S phase of LNCaP with no effect on PC3 .\n\nHowever , PC3 cells transfected with the androgen receptor ( PC3-AR2 ) exhibited a G2/M arrest and a marked reduction ( 57% ) in the S phase during cell cycle progression .\n\nIn the analysis of cell cycle regulatory molecules , selenium-treated cells demonstrated a significant induction of cyclin-dependent kinase inhibitors Cip1/p21 and Kip1/p27 .\n\nThese data suggest that selenium possesses strong antiproliferative properties in regard to human PCA .\n\nThis effect appears to be dependent on the presence of a functioning androgen receptor .\n\nThis provides a theoretical basis for Phase III studies of selenium in PCA prevention .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Interactions of CD70 , a tumor necrosis factor-related cell surface ligand and its receptor , CD27 , are thought to play an important role for T- , B- , and natural killer-cell activation .\n\nHowever , ligation of CD27 can also induce apoptosis .\n\nHuman glioblastoma is paradigmatic for cancer-associated immunosuppression .\n\nWe identified CD70 as a radioinducible gene in U87 MG glioma cells .\n\nA screening of a panel of human glioma cell lines revealed that 11 of 12 cell lines expressed CD70 mRNA and protein .\n\nTwo human neuroblastoma cell lines did not express CD70 .\n\nCD70 mRNA expression was enhanced by irradiation in 8 of 12 glioma cell lines in a p53-independent manner .\n\nNo alteration in CD70 expression was observed after glioma cell exposure to cytotoxic drugs such as lomustine .\n\nCD70 protein was also detected by immunocytochemistry in 5 of 12 glioblastomas and 3 of 4 anaplastic astrocytomas in vivo .\n\nCD27 expression was not detected in any glioma cell line , and there was no evidence for autocrine or backward signaling of the CD70 system in human glioma cells .\n\nUnexpectedly , CD70 expressed on glioma cells did not increase the immunogenicity of glioma cells in vitro .\n\nIn contrast , CD70-positive glioma cells induced apoptosis in peripheral blood mononuclear cells ( PBMCs ) in a CD70-dependent manner .\n\nNeutralization of CD70 expressed on glioma cells prevented apoptosis and enhanced the release of tumor necrosis factor-alpha in cocultures of glioma cells and PBMCs .\n\nThe effects of CD70-expressing glioma cells on PBMCs were mimicked by agonistic CD27 antibodies .\n\nConversely , the shedding of CD27 by PBMCs was identified as a possible escape mechanism from glioma cell-induced CD70-dependent apoptosis .\n\nThus , induction of B-cell and T-cell apoptosis via interactions of CD70 expressed on glioma cells and CD27 expressed on B and T cells may be a novel way for the immune escape of malignant gliomas .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "The cyclin-dependent kinase ( CDK ) inhibitor p21(Waf1/Cip1/Sdi1) was identified initially as a gene induced in senescent cells and itself has been shown to cause permanent growth arrest/senescence .\n\nReactive oxygen species ( ROS ) , a byproduct of oxidative processes , can also induce an irreversible growth arrest similar to senescence .\n\nHere we show that p21 increased intracellular levels of ROS both in normal fibroblasts and in p53-negative cancer cells .\n\nN-acetyl-L-cysteine , an ROS inhibitor , rescued p21-induced senescence , showing that ROS elevation is necessary for induction of the permanent growth arrest phenotype. p16(Ink4a) , a CDK4- and CDK6-specific inhibitor , failed to increase ROS levels , and cell cycle arrest induced by p16 was reversible following its down-regulation , demonstrating the specificity of this p21 effect .\n\nA p21 mutant that lacked the ability to bind proliferating cell nuclear antigen ( PCNA ) retained the ability to induce both ROS and permanent growth arrest .\n\nAll of these findings establish that p21 mediates senescence by a mechanism involving ROS accumulation which does not require either its PCNA binding or the CDK inhibitory functions shared with p16 .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "The t(10;11)(p12;q23) chromosomal translocation in human acute myeloid leukemia results in the fusion of the MLL and AF10 genes .\n\nThe latter codes for a novel leucine zipper protein , one of many MLL fusion partners of unknown function .\n\nIn this report , we demonstrate that retroviral-mediated transduction of an MLL-AF10 complementary DNA into primary murine myeloid progenitors enhanced their clonogenic potential in serial replating assays and led to their efficient immortalization at a primitive stage of myeloid differentiation .\n\nFurthermore , MLL-AF10-transduced cells rapidly induced acute myeloid leukemia in syngeneic or severe combined immunodeficiency recipient mice .\n\nStructure/function analysis showed that a highly conserved 82-amino acid portion of AF10 , comprising 2 adjacent alpha-helical domains , was sufficient for immortalizing activity when fused to MLL .\n\nNeither helical domain alone mediated immortalization , and deletion of the 29-amino acid leucine zipper within this region completely abrogated transforming activity .\n\nSimilarly , the minimal oncogenic domain of AF10 exhibited transcriptional activation properties when fused to the MLL or GAL4 DNA-binding domains , while neither helical domain alone did .\n\nHowever , transcriptional activation per se was not sufficient because a second activation domain of AF10 was neither required nor competent for transformation .\n\nThe requirement for alpha-helical transcriptional effector domains is similar to the oncogenic contributions of unrelated MLL partners ENL and ELL , suggesting a general mechanism of myeloid leukemogenesis by a subset of MLL fusion proteins , possibly through specific recruitment of the transcriptional machinery .", "output": "Enabling replicative immortality" }, { "input": "The retinoic acid receptor beta2 ( RARbeta2 ) protein is a putative tumor suppressor that inhibits proliferation and can induce apoptosis when introduced into breast , cervical , lung , and pancreatic cancer cell lines .\n\nTo determine if RARbeta2 suppresses proliferation of mammary-derived cancer cells in vivo , we transduced MDA-MB-435 breast cancer cells with the LXSN retroviral vector containing RARbeta2 and implanted LXSN vector- or RARbeta2-transduced cells into the mammary fat pads of nude and severe combined immune deficiency ( SCID ) mice .\n\nWe analyzed the xenografts for several tumor parameters , including tumor size , inflammation , vascularity , mitoses , tumor recurrence at the primary site following resection , and metastases .\n\nWe found that 19 of 52 mice inoculated with vector-transduced cells developed metastases in multiple organs while only one of 55 mice receiving RARbeta2-transduced cells displayed evidence of metastases ( p < 0.000001 , combined experiments , two-tailed Fisher's exact test ) .\n\nMoreover , RARbeta2-tumor cell recipient mice had a lower incidence of post-resection tumor recurrence ( 8/55 vs. 25/52 , p = 0.0004 ) , 34% less necrosis ( in three of four experiments , p = 0.001 ) , and 39% fewer mitoses in tumor tissue ( p < 0.000001 ) .\n\nOur findings suggest that RARbeta2 may play a role in inhibiting the metastatic cascade in a mouse mammary gland xenograft tumor model and is a potential candidate for therapeutic intervention in human breast cancer .", "output": "Activating invasion and metastasis" }, { "input": "The yeast Sir2 protein mediates chromatin silencing through an intrinsic NAD-dependent histone deacetylase activity .\n\nSir2 is a conserved protein and was recently shown to regulate lifespan extension both in budding yeast and worms .\n\nHere , we show that SIRT1 , the human Sir2 homolog , is recruited to the promyelocytic leukemia protein ( PML ) nuclear bodies of mammalian cells upon overexpression of either PML or oncogenic Ras ( Ha-rasV12 ) .\n\nSIRT1 binds and deacetylates p53 , a component of PML nuclear bodies , and it can repress p53-mediated transactivation .\n\nMoreover , we show that SIRT1 and p53 co-localize in nuclear bodies upon PML upregulation .\n\nWhen overexpressed in primary mouse embryo fibroblasts ( MEFs ) , SIRT1 antagonizes PML-induced acetylation of p53 and rescues PML-mediated premature cellular senescence .\n\nTaken together , our data establish the SIRT1 deacetylase as a novel negative regulator of p53 function capable of modulating cellular senescence .", "output": "Enabling replicative immortality" }, { "input": "BACKGROUND Insulin-like growth factor I ( IGF-I ) stimulates cell proliferation and inhibits apoptosis in the lung and other tissues by interacting with the IGF-I receptor .\n\nThe major binding protein for IGF-I , insulin-like growth factor-binding protein 3 ( IGFBP-3 ) , modulates the effects of IGF-I but also inhibits cell growth and induces apoptosis independent of IGF-I and its receptor .\n\nIn a prospective study of men in Shanghai , China , we examined the association between serum levels of IGF-I and IGFBP-3 and the subsequent risk of lung cancer .\n\nMETHODS From 1986 to 1989 , serum was collected from 18,244 men aged 45-64 years living in Shanghai without a history of cancer .\n\nWe analyzed IGF-I and IGFBP-3 levels in serum from 230 case patients who developed incident lung cancer during follow-up and from 740 control subjects .\n\nRESULTS Among 230 case patients and 659 matched control subjects , increased IGF-I levels were not associated with increased risk of lung cancer .\n\nHowever , for subjects in the highest quartile relative to the lowest quartile of IGFBP-3 , the odds ratio ( OR ) for lung cancer , adjusted for smoking and IGF-I , was 0.50 ( 95% confidence interval [ CI ] = 0.25 to 1.02 ) .\n\nWhen the analysis was restricted to ever smokers ( 184 case patients and 344 matched control subjects ) , the OR for lung cancer in men in the highest quartile of IGFBP-3 relative to those in the lowest quartile , adjusted for smoking and IGF-I , was 0.41 ( 95% CI = 0.18 to 0.92 ) .\n\nCONCLUSIONS In this prospective study of Chinese men , higher serum levels of IGF-I did not increase the risk of lung cancer .\n\nHowever , subjects with higher serum levels of IGFBP-3 were at reduced risk of lung cancer .\n\nThis finding is consistent with experimental data that indicate that IGFBP-3 can inhibit cellular proliferation and induce apoptosis independent of IGF-I and the IGF-I receptor .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "p53 and INK4a/ARF mutations promote tumorigenesis and drug resistance , in part , by disabling apoptosis .\n\nWe show that primary murine lymphomas also respond to chemotherapy by engaging a senescence program controlled by p53 and p16(INK4a) .\n\nHence , tumors with p53 or INK4a/ARF mutations-but not those lacking ARF alone-respond poorly to cyclophosphamide therapy in vivo .\n\nMoreover , tumors harboring a Bcl2-mediated apoptotic block undergo a drug-induced cytostasis involving the accumulation of p53 , p16(INK4a) , and senescence markers , and typically acquire p53 or INK4a mutations upon progression to a terminal stage .\n\nFinally , mice bearing tumors capable of drug-induced senescence have a much better prognosis following chemotherapy than those harboring tumors with senescence defects .\n\nTherefore , cellular senescence contributes to treatment outcome in vivo .", "output": "Genomic instability and mutation, Enabling replicative immortality, Resisting cell death" }, { "input": "Insulin-like growth factor I ( IGF-I ) and IGF-II stimulate cancer cell proliferation via interaction with the type I IGF receptor ( IGF-IR ) .\n\nWe put forward the hypothesis that IGF-IR mediates cancer cell growth by regulating amino acid transport , both when sufficient nutrients are present and when key nutrients such as glutamine are in limited supply .\n\nWe examined the effects of alphaIR3 , the monoclonal antibody recognizing IGF-IR , on cell growth and amino acid transport across the cell membrane in a human neuroblastoma cell line , SK-N-SH .\n\nIn the presence of alphaIR3 ( 2 micro/ml ) , cell proliferation was significantly attenuated in both control ( 2 mM glutamine ) and glutamine-deprived ( 0 mM glutamine ) groups .\n\nGlutamine deprivation resulted in significantly increased glutamate ( system X(AG)(-) ) , MeAIB ( system A ) , and leucine ( system L ) transport , which was blocked by alphaIR3 .\n\nGlutamine ( system ASC ) and MeAIB transport was significantly decreased by alphaIR3 in the control group .\n\nAddition of alphaIR3 significantly decreased DNA and protein biosynthesis in both groups .\n\nGlutamine deprivation increased the IGF-IR protein on the cell surface .\n\nOur results suggest that activation of IGF-IR promotes neuroblastoma cell proliferation by regulating trans-membrane amino acid transport .", "output": "Sustaining proliferative signaling" }, { "input": "In order to elucidate the mechanisms by which tumour-specific CD4+ T-cell responses are impaired during tumour development , an attempt was made to identify factors which impair CD4+ T-cell responses at a late tumour-bearing stage .\n\nPlasma from mice bearing B16 melanoma for 30 days ( plasma d30 ) showed a more profound immunosuppressive effect on the in vitro proliferation of unrelated antigen-specific CD4+ T cells in the presence of both antigen and antigen-presenting cells ( APC ) than plasma from na\ufffdve mice .\n\nThe level of plasma transforming growth factor ( TGF)- was elevated in mice bearing B16 melanoma for 30 days compared with na\ufffdve mice , and the suppressive effect of plasma d30 was partially diminished by the neutralization of TGF- .\n\nInterestingly , immunoglobulin ( IgG)-bound TGF- , but not IgG-unbound TGF- , in plasma d30 was suggested to be responsible for the immunosuppressive activity .\n\nIn addition , no suppressive effect of plasma d30 was observed when antigen was added as a class II peptide , thus suggesting that the impaired proliferation of CD4+ T cells in the presence of plasma d30 was due to a dysfunction of antigen uptake/processing by APC .\n\nFurthermore , dissociation between IgG and TGF- resulted in a loss of the suppressive activity of plasma d30 .\n\nTaken together , these results suggest that circulating IgG-bound TGF- is , at least in part , responsible for the impaired responses of CD4+ T cells at the late tumour-bearing stage by suppressing antigen uptake/ processing by APC .", "output": "Sustaining proliferative signaling, Avoiding immune destruction" }, { "input": "AIM The goal of this study was to characterize the AFP receptor , its possible signal transduction pathway and its proliferative functions in human hepatoma cell line Bel 7402 .\n\nMETHODS Cell proliferation enhanced by AFP was detected by MTT assay , 3H-thymidine incorporation and S-stage percentage of cell cycle analysis .\n\nWith radioactive labeled 125I-AFP for receptor binding assay ; cAMP accumulation , protein kinase A activity were detected by radioactive immunosorbent assay and the change of intracellular free calcium ( Ca2+i ) was monitored by scanning fluorescence intensity under TCS-NT confocal microscope .\n\nThe expression of oncogenes N- ras , p 53 , and p21( ras ) in the cultured cells in vitro were detected by Northern blotting and Western blotting respectively .\n\nRESULTS It was demonstrated that AFP enhanced the proliferation of human hepatoma Bel 7402 cell in a dose dependent fashion as shown in MTT assay , ( 3)H-thymidine incorporation and S-phase percentage up to 2-fold .\n\nTwo subtypes of AFP receptors were identified in the cells with Kds of 1.3 x 10(-9)mol.L(-1) and 9.9 x10(-8)mol .\n\n( -1)L respectively .\n\nPretreatment of cells with AFP resulted in a significant increase ( 625% ) in cAMP accumulation .\n\nThe activity of protein kinase A activity were increased up to 37.5 , 122.6 , 73.7 and 61.2% at treatment time point 2 , 6 , 12 and 24 hours .\n\nThe level of intracellular calcium were elevated after the treatment of alpha-fetoprotein and achieved to 204% at 4 min .\n\nThe results also showed that AFP(20mg.L(-1)) could upregulate the expression of N- ras oncogenes and p 53 and p21( ras ) in Bel 7402 cells .\n\nIn the later case,the alteration were 81.1%(12h) and 97.3%(12h) respectively compared with control .\n\nCONCLUSION These results demonstrate that AFP is a potential growth factor to promote the proliferation of human hepatoma Bel 7402 cells .\n\nIts growth-regulatory effects are mediated by its specific plasma membrane receptors coupled with its transmembrane signaling transduction through the pathway of cAMP-PKA and intracellular calcium to regulate the expression of oncogenes .", "output": "Sustaining proliferative signaling" }, { "input": "PURPOSE Astrocytoma arises in the central nervous system as a tumor of great lethality , in part because of the invasive potential of the neoplastic cells that are able to release extracellular matrix-degrading enzymes .\n\nFurin convertase activates several precursor matrix metalloproteases involved in the breakdown of the extracellular matrix .\n\nIn the present study inhibition of furin was achieved by gene transfer of alpha(1)-antitrypsin Portland ( PDX ) cDNA .\n\nEXPERIMENTAL DESIGN This furin inhibitor was transfected into two tumorigenic astrocytoma cell lines .\n\nThe inhibitory effect was evaluated using in vivo tumorigenicity , invasion , and proliferation assays , as well as by investigating impairment of furin substrate processing .\n\nRESULTS Expression of PDX prevented the s.c. growth of the transfected cells .\n\nInvasion assays demonstrated that PDX-transfected cells exhibited a reduced invasive ability in vitro and in vivo .\n\nFurthermore , s.c. growth of PDX transfectant xenotransplants showed a significant reduction in size that coincided with a significant decrease of the in vitro doubling time and of the in vivo cell proliferation ability .\n\nAdditional studies showed that the furin substrates insulin-like growth factor IR , transforming growth factor beta and membrane type 1-matrix metalloprotease were not activated in PDX-expressing astrocytoma cells .\n\nCONCLUSIONS PDX expression in astrocytoma cells demonstrated a direct mechanistic link between furin inhibition , and decreased astrocytoma proliferation and invasive ability .\n\nBecause furin inhibition inhibits both invasiveness and cell growth in astrocytoma , furin should be considered a promising target for glioblastoma therapy .", "output": "Activating invasion and metastasis" }, { "input": "Elevated insulin-like growth factor binding protein-related protein 1 ( IGFBP-rP1 ) mRNA in senescent human mammary epithelial cells suggested that the IGFBP-3 gene product may inhibit cell proliferation .\n\nTo test this hypothesis , we used a retroviral vector to express IGFBP-rP1 cDNA in the IGFBP-rP1-deficient MCF-7 breast cancer cell line .\n\nCompared with control vector-transduced cells , cumulative cell numbers for IGFBP-rP1-transduced polyclonal or clonal cell cultures were reduced by 39 and 74% , respectively , after 1 week .\n\nMedium conditioned by IGFBP-rP1-producing cultures reduced cumulative cell numbers in parental MCF-7 cultures by 20% compared with medium from cultures of a control vector-transduced cell line .\n\nNuclear fragmentation analysis and cell proliferation assays completed in the presence of the pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone excluded apoptosis as the responsible mechanism .\n\nThe percentage of cells containing senescence-associated beta-galactosidase activity was doubled compared with control cell cultures .\n\nFlow cytometry analysis indicated that twice as many noncycling cells were present in the IGFBP-rP1-transduced MCF-7 cell cultures compared with controls .\n\nThese findings indicate that IGFBP-rP1 is an inhibitor of MCF-7 breast cancer cell proliferation and may act via a cellular senescence-like mechanism .", "output": "Enabling replicative immortality, Sustaining proliferative signaling, Resisting cell death" }, { "input": "In vitro model systems for studying uterine leiomyomas are limited in that human-derived leiomyoma cells grow poorly in culture compared with normal myometrial cells and begin to senesce early , at approximately passage 10 in our studies .\n\nTo our knowledge , a good in vitro human-derived cell culturing system for leiomyomas does not exist .\n\nIn an attempt to fill this void , we have immortalized a uterine leiomyoma cell line by inducing telomerase activity , which allows cells to bypass their normal programmed senescence .\n\nTelomerase activity was induced by infecting the target ( uterine leiomyoma and normal myometrial ) cells with a retroviral vector containing hTERT , the gene for the catalytic subunit of telomerase .\n\nSubsequent analysis by RT-PCR and the telomeric repeat amplification protocol assay confirmed expression of the inserted gene and induction of telomerase activity in leiomyoma and myometrial cells .\n\nAnalysis of cells for estrogen receptor-alpha and progesterone receptor proteins by Western blotting showed no change in expression of these proteins between the immortalized and parental leiomyoma and myometrial cells .\n\nBoth immortalized and parental myometrial and leiomyoma cells expressed the smooth muscle-specific cytoskeletal protein alpha-actin and were negative for mutant p53 protein as evidenced by immunocytochemical staining .\n\nThe immortalized leiomyoma and myometrial cells showed no anchorage-independent growth , with the exception of a small subpopulation of immortalized leiomyoma cells at a higher passage that did form two to three small colonies ( per 50,000 cells ) in soft agar .\n\nNone of the immortalized cells were tumorigenic in nude mice .\n\nIn conclusion , our data show the successful insertion of the hTERT gene into leiomyoma and myometrial cells and the immortalization of these cell lines without phenotypic alteration from the parental cell types ( up to 200 population doublings ) .\n\nThese cells should help to advance research in understanding the molecular pathways involved in the conversion of a normal myometrial cell to a leiomyoma cell and the mechanisms responsible for the growth of uterine leiomyomas .\n\nAnswers to these questions will undoubtedly lead to the development of more effective treatment and intervention regimens for clinical cases of uterine leiomyoma .", "output": "Enabling replicative immortality" }, { "input": "BACKGROUND Modulation of the expression of retinoic acid receptors ( RAR ) alpha and gamma in adult rat prostate by testosterone ( T ) suggests that RAR signaling events might mediate some of the androgen effects on prostate cells .\n\nMETHOD In this study , we examined the interactions between T and retinoic acid ( RA ) in cell growth of human prostate carcinoma cells , LNCaP , and their relationship with the expression of RAR and epidermal growth factor receptor ( EGF-R ) .\n\nRESULTS Both T and RA , when administered alone , stimulated 3H-thymidine incorporation in LNCaP cells in a dose-dependent manner ; the effect of each agent was reciprocally attenuated by the other agent .\n\nTestosterone treatment of LNCaP cells also resulted in dose dependent , biphasic increases in RAR alpha and gamma mRNAs ; increases paralleled that of 3H-thymidine incorporation and were attenuated by the presence of 100 nM RA .\n\nThese results suggest a link between RAR signaling and the effect of T on LNCaP cell growth .\n\nGel electrophoretic mobility shift assays revealed the presence of putative androgen responsive element ( ARE ) in the promoter region of RAR alpha gene , suggesting that a direct AR-DNA interaction might mediate the effects of T on RAR alpha gene .\n\nFurthermore , treatment of LNCaP cells with 20 nM T resulted in an increase in EGF-R .\n\nIn contrast , EGF-R was suppressed by 100 nM RA that also suppressed the effect of T. CONCLUSIONS Current results demonstrate interactions between T and RA in the expression of RARs and cell growth in LNCaP cells .\n\nThe presence of putative ARE in the promoter of the RAR alpha gene suggests that AR-DNA interaction might mediate the effects of T on RAR alpha gene .\n\nThe opposite effects of T and RA on the expression of RAR and EGF-R suggest that signal events of these receptors might be involved in the interaction between T and RA in the control of LNCaP cell growth .", "output": "Sustaining proliferative signaling" }, { "input": "We have modelled multiple stages of malignant transformation of human endothelial cells ( ECs ) by overexpressing the catalytic subunit of human telomerase ( hTERT ) , together with SV40 T antigen ( SV40T ) and oncogenic N-ras .\n\nTransfection with hTERT alone , led to the immortalization of two out of three cultures of bone marrow-derived ECs ( BMECs ) .\n\nOne hTERT transduced BMEC culture underwent a long proliferative lag before resuming proliferation .\n\nBMECs transfected with hTERT alone were functionally and phenotypically normal .\n\nBMECs transfected with SV40T ( BMSVTs ) had an extended lifespan , but eventually succumbed to crisis .\n\nBMSVTs exhibited a partially transformed phenotype , demonstrating growth factor independence , altered antigen expression and forming tiny , infrequent colonies in vitro .\n\nTransduction of BMSVTs with hTERT resulted in immortalization of 4 out of 4 cultures .\n\nBMSVTs immortalized with hTERT formed large colonies in vitro and small transient tumours in vivo .\n\nBMECs co-expressing SV40T , hTERT and N-ras exhibited an overtly transformed phenotype ; forming very large colonies with an altered morphology and generating rapidly growing tumours in vivo .\n\nThese investigations demonstrate transformation of human ECs to an overtly malignant phenotype .\n\nThis model will be useful for understanding mechanisms underlying vascular and angiogenic neoplasias , as well as for testing drugs designed to curtail aberrant EC growth .", "output": "Inducing angiogenesis, Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "The association between acute myeloid leukaemia ( AML ) and the aberrant expression of Hoxa9 is evidenced by ( 1 ) proviral activation of Hoxa9 and Meis1 in BXH-2 murine AML , ( 2 ) formation of the chimeric Nup98-HoxA9 transactivator protein as a consequence of the t(7;11) translocation in human AML , and ( 3 ) the strong expression of HoxA9 and Meis1 in human AML .\n\nIn mouse models , enforced retroviral expression of Hoxa9 alone in marrow is not sufficient to cause rapid AML , while co-expression of Meis1 and Hoxa9 induces rapid AML .\n\nIn contrast , retroviral expression of Nup98-HoxA9 is sufficient to cause rapid AML in the absence of enforced Meis1 expression .\n\nPreviously , we demonstrated that Hoxa9 could block the differentiation of murine marrow progenitors cultured in granulocyte-macrophage colony-simulating factor ( GM-CSF ) .\n\nThese progenitors lacked Meis1 expression , could not proliferate in stem cell factor ( SCF ) , but could differentiate into neutrophils when switched into granulocyte colony-simulating factor ( G-CSF ) .\n\nEctopic expression of Meis1 in these Hoxa9 cells suppressed their G-CSF-induced differentiation , permitted proliferation in SCF , and therein offered a potential explanation of cooperative function .\n\nBecause Meis1 binds N-terminal Hoxa9 sequences that are replaced by Nup98 , we hypothesized that Nup98-HoxA9 might consolidate the biochemical functions of both Hoxa9 and Meis1 on target gene promoters and might evoke their same lymphokine-responsive profile in immortalized progenitors .\n\nHere we report that Nup98-HoxA9 , indeed mimicks Hoxa9 plus Meis1 coexpression - it immortalizes myeloid progenitors , prevents differentiation in response to GM-CSF , IL-3 , G-CSF , and permits proliferation in SCF .\n\nUnexpectedly , however , Nup98-Hoxa9 also enforced strong transcription of the cellular Hoxa9 , Hoxa7 and Meis1 genes at levels similar to those found in mouse AML's generated by proviral activation of Hoxa9 and Meis1 .\n\nUsing Hoxa9(-/-) marrow , we demonstrate that expression of Hoxa9 is not required for myeloid immortalization by Nup98-HoxA9 .\n\nRapid leukaemogenesis by Nup98-HoxA9 may therefore result from both the intrinsic functions of Nup98-HoxA9 , as well as of those of coexpressed HOX and MEIS1 genes .", "output": "Enabling replicative immortality" }, { "input": "BACKGROUND : Previously , we have observed that highly unsaturated dietary ( n-3 ) fatty acids inhibit cell proliferation in conjunction with stimulation of insulin-like growth factor-binding protein ( IGFBP)-6 secretion in Caco-2 cells , a human colon carcinoma cell line .\n\nMETHODS : To test the converse hypothesis that inhibition of endogenous IGFBP-6 secretion stimulates Caco-2 cell proliferation , cells were transfected with the antisense IGFBP-6 expression construct or pcDNA3 vector only , and single colonies resistant to G418 sulfate were isolated .\n\nRESULTS : Our initial studies indicated that three antisense clones grew faster and produced less IGFBP-6 than two pcDNA3 clones , so antisense IGFBP-6 #5 and pcDNA3 #8 were selected for further detailed analysis .\n\nBoth the control and antisense clones grew in serum-free medium reaching a plateau density at day eight .\n\nHowever , the antisense clone grew at a rate faster than that of the control and reached a final density that was 31 +/- 3% higher than the control .\n\nNorthern blot , ligand blot and immunoblot analyses revealed that accumulation of IGFBP-6 mRNA and concentrations of IGFBP-6 peptide produced by the antisense clone were decreased by 80-90% compared to the control .\n\nThe doubling times of the antisense and control clones were 21.9 +/- 0.4 and 24.8 +/- 0.3 h ( P < 0.05 ) , respectively .\n\nExogenous IGF-I and IGF-II ( 0.2-200 nmol/L ) stimulated proliferation of both the control and antisense clones in a dose-dependent manner , but the relative potency and efficacy of IGF-II was higher in the antisense clone compared to the control .\n\nThese results indicate that suppression of IGFBP-6 secretion correlates with an increase in the basal rate of Caco-2 cell growth .\n\nCONCLUSIONS : Our findings are consistent with the hypothesis that IGFBP-6 inhibits cell growth by binding to endogenously produced IGF-II , thereby preventing IGF-II from interacting with the IGF-I receptor to stimulate cellular proliferation by an autocrine mechanism .", "output": "Sustaining proliferative signaling" }, { "input": "Glioblastoma multiforme , the most common form of malignant brain tumor,is resistant to all forms of therapy and causes death within 9-12 months of diagnosis .\n\nGlioblastomas are known to contain numerous genetic and physiological alterations affecting cell survival and proliferation ; one of the most common alterations being platelet-derived growth factor ( PDGF ) autocrine signaling characterized by coexpression of PDGF and its receptor .\n\nThe PDGF family consists of four members , PDGF-A , -B , -C , and -D , that signal through the alpha and beta PDGF receptor ( PDGFR ) tyrosine kinases .\n\nNumerous studies have demonstrated expression of PDGF-A , PDGF-B , and the PDGFRs in gliomablastomas , but such studies have not been conducted for the newly identified PDGF-C and -D .\n\nTherefore , we examined the expression of all PDGF ligands and receptors in 11 glioma cell lines and 5 primary glioblastoma tumor tissues by quantitative reverse transcription-PCR .\n\nExpression of PDGF/PDGFR pairs that are known to functionally interact were identified in all of the samples .\n\nInterestingly , PDGF-C expression was ubiquitous in brain tumor cells and tissues but was very low or absent in normal adult and fetal brain .\n\nPDGF-D was expressed in 10 of 11 brain tumor cell lines and 3 of 5 primary brain tumor samples .\n\nAs a strategy for blocking PDGFR signaling , CT52923 , a potent selective small molecule piperazinyl quinazoline kinase inhibitor of the PDGFR , was identified .\n\nIn model systems using NIH/3T3 cells , CT52923 blocked PDGF autocrine-mediated phosphorylation of PDGFR , Akt , and mitogen-activated protein kinase ( MAPK ) , while having no effect on v-fms or V12-ras-mediated Akt or extracellular signal-regulated protein kinase ( Erk ) phosphorylation .\n\nMore importantly , p.o. administration of CT52923 to nude mice caused a significant 61% reduction ( P < 0.006 ) in tumor growth of NIH/3T3 cells transformed by PDGF , whereas tumor formation by cells expressing v-fms was unaffected .\n\nWe next characterized PDGF autocrine signaling in five glioblastoma cell lines .\n\nIn all of the cases , PDGF autocrine signaling was evident because treatment with 1-10 microM CT52923 inhibited PDGFR autophosphorylation when present at a detectable level and blocked downstream Akt and/or Erk phosphorylation .\n\nThe functional significance of PDGF autocrine signaling in these cells was demonstrated by the fact that the CT52923 inhibited soft agar colony formation , and , when given p.o. to nude mice , it effectively reduced tumor formation by 44% ( P < 0.0019 ) after s.c. injection of C6 glioblastoma cells .\n\nThis study of glioblastoma cells and primary tissues is the first to implicate PDGF-C and -D in brain tumor formation and confirms the existence of autocrine signaling by PDGF-A and -B .\n\nMore importantly , treatment with the PDGFR antagonist CT52923 inhibited survival and/or mitogenic pathways in all of the glioblastoma cell lines tested and prevented glioma formation in a nude mouse xenograft model .\n\nTogether these findings demonstrate the potential therapeutic utility of this class of compounds for the treatment of glioblastoma .", "output": "Sustaining proliferative signaling" }, { "input": "Direct experimental evidence implicates telomere erosion as a primary cause of cellular senescence .\n\nUsing a well characterized model system for breast cancer , we define here the molecular and cellular consequences of adriamycin treatment in breast tumor cells .\n\nCells acutely exposed to adriamycin exhibited an increase in p53 activity , a decline in telomerase activity , and a dramatic increase in beta-galactosidase , a marker of senescence .\n\nInactivation of wild-type p53 resulted in a transition of the cellular response to adriamycin treatment from replicative senescence to delayed apoptosis , demonstrating that p53 plays an integral role in the fate of breast tumor cells treated with DNA-damaging agents .\n\nStable introduction of hTERT , the catalytic protein component of telomerase , into MCF-7 cells caused an increase in telomerase activity and telomere length .\n\nTreatment of MCF-7-hTERT cells with adriamycin produced an identical senescence response as controls without signs of telomere shortening , indicating that the senescence after treatment is telomere length-independent .\n\nHowever , we found that exposure to adriamycin resulted in an overrepresentation of cytogenetic changes involving telomeres , showing an altered telomere state induced by adriamycin is probably a causal factor leading to the senescence phenotype .\n\nTo our knowledge , these data are the first to demonstrate that the mechanism of adriamycin-induced senescence is dependent on both functional p53 and telomere dysfunction rather than overall shortening .", "output": "Genomic instability and mutation, Enabling replicative immortality, Resisting cell death" }, { "input": "The relationship between thyroid hormone ( triiodothyronine , T(3) ) and breast cancer is unclear .\n\nWe studied the effect of the c-erbA/TR alpha proto-oncogene encoding a functional T(3) receptor ( TR alpha 1 ) , of its ligand T(3) , and of its retroviral , mutated counterpart , the v-erbA oncogene , on the proliferation capacity of nontumorigenic mammary epithelial cells ( EpH4 ) .\n\nWe found that EpH4 cells expressing ectopically TR ( EpH4 + TR alpha 1 ) or v-erbA ( EpH4 + v-erbA ) proliferated faster than parental EpH4 cells that contained low levels of endogenous TR .\n\nT(3) inhibited DNA synthesis and proliferation in EpH4 + TR alpha 1 cells but not EpH4 or EpH4 + v-erbA cells .\n\nThe study of cell-cycle genes showed that T(3) decreased cyclin D1 RNA and protein levels in EpH4 + TR alpha 1 cells .\n\nIn addition , T(3) downregulated the expression of T1 , a gene that is overexpressed in human breast adenocarcinomas and is induced by mitogens , serum , and several oncogenes and cytokines .\n\nInhibition of the T1 gene by T(3) required both de novo mRNA and protein synthesis .\n\nFurthermore , T(3) abolished the induction of T1 by the tumor promoter 12-O-tetradecanoylphorbol-13-acetate and inhibited the activity of an activation protein 1-dependent promoter ( -73-Col-CAT ) in EpH4 + TR alpha 1 cells , suggesting that interference with activation protein 1 transcription factor plays a part in the inhibition of the T1 gene .\n\nOur results showed that T(3) reduced the proliferation of mammary epithelial cells and inhibited the expression of cyclin D1 and T1 genes .", "output": "Sustaining proliferative signaling" }, { "input": "The metabolic detoxification capacity may critically regulate the susceptibility of human tissues to cancer development .\n\nWe used standardized and quantitative , reverse transcription-polymerase chain reaction ( StaRT-PCR ) and microarray chip techniques to analyze transcript levels of multiple detoxification enzymes in cultured normal human oral keratinocytes ( NOK ) and the Siman virus 40 T antigen-immortalized oral keratinocyte line SVpgC2a , viewing the latter as a model of a benign tumor state .\n\nWith good agreement between the 2 methodologies , NOK and SVpgC2a were found to express transcripts for cytochrome P450 enzymes ( CYPs ) , factors related to CYP induction and enzymes involved in conjugation reactions or detoxification of reactive oxygen .\n\nThe cell types expressed similar levels of CYP 2B6/7 , CYP 2E1 , P450 oxidoreductase , the aryl hydrocarbon receptor nuclear translocator , sulfotransferase 1A1 , sulfotransferase 1A3 , epoxide hydrolase , glutathione S-transferase M3 , glutathione S-transferase pi and catalase , superoxide dismutase 1 , glutathione peroxidase 1 and glutathione peroxidase 3 .\n\nIn contrast , SVpgC2a exhibited comparatively higher levels of CYP1A1 , 1B1 , aryl hydrocarbon receptor , glutathione S-transferase M1 , 2 , 4 , 5 , glutathione S-transferase theta 1 and superoxide dismutase 2 and comparatively lower levels of UDP glycosyltransferase 2 and microsomal glutathione S-transferase 1 .\n\nSome transcripts , e.g. , CYP 2A6/7 , were not detected by either standard , non quantitative RT-PCR or the above methods , whereas others were barely quantifiable by StaRT-PCR , i.e. , were present at 1-10 molecules/10(6) molecules of actin .\n\nOverall , the expression analysis demonstrated presence of mRNA for multiple enzymes involved in foreign compound metabolism and detoxification pathways , including several enzymes not previously reported for oral epithelium .\n\nGenerally , the comparison of NOK from 2 individuals indicated relatively similar transcript levels of these enzymes .\n\nIn contrast , differences between NOK and SVpgC2a , e.g. , for CYP1B1 , may reflect alteration caused by immortalization and aid identification of early stage tumor markers in oral epithelium .", "output": "Enabling replicative immortality" }, { "input": "Urokinase plasminogen activator receptor ( uPAR ) activates alpha5beta1 integrin and ERK signaling , inducing in vivo proliferation of HEp3 human carcinoma .\n\nHere we demonstrate that EGFR mediates the uPAR/integrin/fibronectin ( FN ) induced growth pathway .\n\nIts activation is ligand-independent and does not require high EGFR , but does require high uPAR expression .\n\nOnly when uPAR level is constitutively elevated does EGFR become alpha5beta1-associated and activated .\n\nDomain 1 of uPAR is crucial for EGFR activation , and FAK links integrin and EGFR signaling .\n\nInhibition of EGFR kinase blocks uPAR induced signal to ERK , implicating EGFR as an important effector of the pathway .\n\nDisruption of uPAR or EGFR signaling reduces HEp3 proliferation in vivo .\n\nThese findings unveil a mechanism whereby uPAR subverts ligand-regulated EGFR signaling , providing cancer cells with proliferative advantage .", "output": "Sustaining proliferative signaling" }, { "input": "We have recently developed surface-shielded transferrin-polyethylenimine ( Tf-PEI)/DNA delivery systems that target reporter gene expression to distant tumors after systemic application .\n\nIn the present study , we used surface-shielded Tf-PEI/DNA complexes for delivering the gene for a highly potent cytokine , tumor necrosis factor-alpha ( TNFalpha ) .\n\nTNFalpha is known for its ability to induce hemorrhagic tumor necrosis and tumor regression .\n\nHowever , the therapeutic application of TNFalpha is hampered by its high systemic toxicity dictating the need to target TNFalpha activity to the tumor .\n\nSystemic application of surface-shielded Tf-PEI complexes with the TNFalpha gene resulted in preferential expression of TNFalpha in the tumor without detectable TNFalpha serum levels , in contrast to the application of nontargeted complexes .\n\nTumor-targeted TNFalpha gene delivery induced pronounced hemorrhagic tumor necrosis and inhibition of tumor growth in three murine tumor models of different tissue origins , Neuro2a neuroblastoma , MethA fibrosarcoma , and M-3 melanoma , with complete tumor regressions observed in the MethA model .\n\nNo systemic TNF-related toxicity was observed due to the localization of the TNFalpha activity to the tumor .\n\nTargeted gene therapy may be an attractive strategy applicable to highly active , yet toxic , molecules such as TNFalpha .", "output": "Resisting cell death" }, { "input": "Clinical data and biological samples were prospectively collected in 42 children with lymphoproliferative disease ( LPD ) secondary to organ/bone marrow transplant-related immunosuppression ( 30 : 11 liver , 10 heart/lung , 8 kidney and 1 bone marrow ) , other drug-induced immunosuppression ( 2 ) , congenital immunodeficiency ( 8 ) or human immunodeficiency virus ( HIV)-related immune dysfunction ( 2 ) .\n\nAges ranged from 10 months to 17 years and there were 15 girls .\n\nPathology was centrally reviewed and showed polymorphic features in 5 cases , monomorphic in 23 , mixed pattern in 5 patients and 9 other types .\n\nUsing the Revised European-American Classification of Lymphoid Neoplasms , 5 were B lymphoblastoid , 24 were high-grade B and 14 were other subtypes .\n\nUsing the Pittsburgh classification , 9 were lymphadenopathic , 10 were systemic , 25 were lymphomatous and , with the Murphy grouping for non-Hodgkin's lymphoma ( NHL ) , 10 were localized and 32 non-localized .\n\nTwenty-four out of 38 evaluable cases were Epstein-Barr virus positive .\n\nThirty-five patients were evaluable for clonality ; 24 were monoclonal and 11 were polyclonal .\n\nReduced immunosuppression in solid organ transplant patients resulted in resolution of disease in 14/24 , which was sustained in 11 .\n\nNineteen patients received chemotherapy , 14/18 evaluable responded , which was sustained in 8 cases .\n\nSeven out of 29 solid organ transplant and 10/13 other immune-deficient patients died .\n\nIn the largest group of patients , solid organ transplants , no significant clinical or biological characteristics that predicted outcome were identified .\n\nIn the transplant group close monitoring of response during reduction in immunosuppression is essential and the early use of B NHL chemotherapy may be effective .", "output": "Avoiding immune destruction" }, { "input": "Epidemiological studies have indicated that females may be at greater risk of smoking associated lung cancer compared with males .\n\nSeveral lines of biochemical evidence support these observations .\n\nA possible role of circulating steroid hormones in the etiology of lung cancer has been hypothesized .\n\nIn the present paper , we have studied the expression of the estrogen receptors ( ER)-alpha and ER beta in histologically normal human lung tissue and lung tumor cell lines .\n\nRelative ER mRNA levels were measured by reverse transcriptase-PCR and normalized to the level of expression of the glyceraldehyde 3-phosphate dehydrogenase gene ( GAPDH ) .\n\nIn lung tissue , an ER alpha transcript was found at various levels in 38 out of 46 cases ( 83% ) .\n\nER beta was expressed in all cases .\n\nThe ERs were expressed at similar levels in females and males , and the levels of ER alpha and ER beta mRNA were significantly related ( P<0.0001 ) .\n\nCompared with the lung tissue , ER expression levels were lower in 16 human lung tumor cell lines and two immortalized human bronchial epithelial cell lines .\n\nFive of the tumor cell lines ( 31% ) expressed detectable levels of ER alpha and both of the immortalized cell lines showed a weak ER alpha expression level .\n\nAll cell lines expressed the ER beta .\n\nThe lung cell lines BEAS-2B and DB354 showed significantly reduced cell proliferation in response to tamoxifen and a minor increased growth in response to 17 beta-estradiol .\n\nIn conclusion , ER genes are abundantly expressed in both histologically normal human lung and lung tumor cell lines .\n\nThis indicates a possible role of ERs in lung carcinogenesis .", "output": "Sustaining proliferative signaling" }, { "input": "The beneficial effect of yoghurt consumption on health and on the improvement of the mucosal immune system is well established , as is the diet-associated risk of colon cancer .\n\nIn an experimental model in BALB/c mice we demonstrated that yoghurt added to the diet for 10 consecutive days , with the procedure repeated each 10 days for 6 months , inhibited the development of a colorectal carcinoma induced by 1,2 dimethylhydrazine ( DMH ) .\n\nThe immunoregulatory mechanisms involved in the inhibition of tumour growth by yoghurt were also examined in these studies .\n\nWe determined B lymphocytes IgA(+) and IgG(+) , as well as CD4(+) and CD8(+) T cells in the large intestine .\n\nWe measured cellular apoptosis and the cytokines TNF-alpha , IFN-gamma and IL-10 .\n\nAn increase in the number of IgA(+) ( P<0.01 ) was observed , but not in IgG(+) ( P<0.01 ) , or in the CD4(+) population ( P<0.01 ) in the mice treated with DMH and yoghurt .\n\nWhile in the group with the carcinogen there was an enhancement in the IgG(+) B cells ( P<0.01 ) and CD8(+) T cells ( P<0.01 ) .\n\nYoghurt increased the number of apoptotic cells and induced IFN-gamma and TNF-alpha cytokine release , their production being regulated by an increase in IL-10 ( P<0.001 ) .\n\nWe demonstrated that yoghurt may exert antitumour activity by a decrease in the inflammatory immune response mediated by IgA(+) increase , apoptosis induction and IL-10 release .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Ovarian steroids are one of the strongest risk factors for breast cancer .\n\nSex hormone-binding globulin ( SHBG ) binds and transports sex steroids in the blood , regulating their bioavailable fraction and access to target cells .\n\nIt can also inhibit the estradiol-induced proliferation of breast cancer cells through its membrane receptor .\n\nThree coding-region polymorphisms , which lead to an amino acid change , have been reported .\n\nWe studied the influence of these three polymorphisms on breast cancer risk in three different populations : Polish familial breast cancer cases , 27% of them carrying a BRCA1/BRCA2 mutation , Nordic familial and sporadic breast cancer cases .\n\nThe reported G to A polymorphism in exon 1 was not found in the 423 analyzed samples .\n\nInstead , we found a C to T transition causing an arg to cys amino acid change within the same codon in one Polish breast cancer patient and her daughter .\n\nBoth of them were heterozygotes for the exon 8 G to A polymorphism as well .\n\nThey were diagnosed for bilateral breast cancer and carried a BRCA1 mutation ( 5382insC ) .\n\nAnalysis of the tumor samples showed that they had lost the wild-type allele both at exons 1 and 8 of the SHBG gene .\n\nAnalysis of the other Polish samples showed no correlation of the exon 8 polymorphism to breast cancer , bilateral breast cancer , BRCA1/BRCA2 mutations or age at diagnosis .\n\nNo association of the exon 8 polymorphism with breast cancer in the Nordic familial or sporadic cases was found .\n\nThe C to T polymorphism located in exon 4 was rare in all the studied populations ( overall allele frequency 0.011 ) .\n\nHowever , in each of the study populations there was a trend for a lower variant allele frequency in cancer cases than in controls .\n\nVariant allele frequency in all the breast cancer cases was significantly lower than in all the controls ( chi(2) = 5.27 , P-value 0.02 ; odds ratio = 0.23 , 95% confidence interval 0.05-0.84 ) .", "output": "Genomic instability and mutation" }, { "input": "We investigated the direct effects of LH-releasing hormone ( LH-RH ) antagonist , Cetrorelix , on the growth of HTOA human epithelial ovarian cancer cell line .\n\nRT-PCR revealed the expression of mRNA for LH-RH and its receptor in HTOA cells .\n\nCetrorelix , at concentrations between 10(-9) and 10(-5) M , exerted a dose-dependent antiproliferative action on HTOA cells , as measured by 5-bromo-2'-deoxyuridine incorporation into DNA .\n\nFlow cytometric analysis indicated that Cetrorelix , at 10(-5) M , arrested cell cycle in HTOA cells , at G1 phase , after 24 h of treatment .\n\nWestern blot analysis of cell cycle-regulatory proteins demonstrated that treatment with Cetrorelix ( 10(-5) M ) for 24 h did not change the steady-state levels of cyclin D1 , cyclin E , and cyclin-dependent kinase ( Cdk)4 but decreased the levels of cyclin A and Cdk2 .\n\nThe protein levels of p21 ( a Cdk inhibitor ) and p53 ( a suppressor of tumor cell growth and a positive regulator for p21 expression ) were increased by Cetrorelix , but the levels of p27 ( a Cdk inhibitor ) did not change significantly .\n\nFlow cytometric analysis and terminal deoxynucleotidyltransferase-mediated deoxyuridine 5-triphosphate nick end labeling staining demonstrated that Cetrorelix ( 10(-5) M ) induced apoptosis in HTOA cells .\n\nIn conclusion , Cetrorelix directly inhibits the proliferation of human epithelial ovarian cancer cells through mechanisms mediated by LH-RH receptor and involving multiple events in cell cycle progression , including G1 phase cell cycle arrest coupled with down-regulation of cyclin A-Cdk2 complex levels , presumably attributable to an up-regulation of p53 and p21 protein levels and apoptosis .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "PURPOSE Current antibody-based immunotherapeutic approaches under evaluation for breast carcinoma are limited in target scope .\n\nFor example , administration of the human epidermal growth factor receptor ( EGFR ) antibody , alone or in combination with a chemotherapeutic drug , is thought to primarily inhibit tumor cell proliferation .\n\nThe aim of this study was to assess the effects of a combined blockade designed to inhibit tumor growth by inhibition of proliferation rate and the proinflammatory effects of interleukin ( IL ) 8 .\n\nEXPERIMENTAL DESIGN A human breast carcinoma cell line that produces high levels of IL-8 was injected s.c. into severe combined immunodeficient mice .\n\nIL-8 has been reported to augment the progression of some human tumors ; thus , we used a human IL-8 antibody , ABXIL8 , in combination with anti-EGFR , ABXEGFR , to inhibit the metastasis of MDA231 tumors .\n\nRESULTS Whereas anti-IL-8 alone had no appreciable antimetastatic effect , the combination of ABXIL8 significantly enhanced the antitumor effects of ABXEGFR , resulting in greater survival of SCID tumor-bearing mice .\n\nThis effect on survival was correlated with decreased metastatic spread and decreased tumor size in mice receiving both antibodies .\n\nIntriguingly , in vitro studies indicate that this antibody combination markedly inhibited matrix metalloproteinase activity associated with MDA-231 cells to a greater degree than either antibody alone .\n\nCONCLUSION Combined administration of these two human antibodies using growth factor blockade in conjunction with chemokine blockade may thus provide a more effective approach for treatment of metastatic human breast carcinoma .", "output": "Activating invasion and metastasis" }, { "input": "Id proteins are negative regulators of basic helix-loop-helix transcription factors , which are critical for expression of genes associated with cellular differentiation .\n\nPrevious studies have shown that overexpression of Id-1 delays cellular senescence in several cell types , including fibroblasts , mammary epithelial cells , and keratinocytes .\n\nAlthough previous studies have demonstrated the expression of Id-1 in endothelium , the regulation of Id-1 has not been studied in these cells .\n\nIn this report , a retroviral vector was used to overexpress Id-1 in human endothelial cells .\n\nSustained expression of Id-1 resulted in a 2- to 3-fold increase in the total number of population doublings ( replicative capacity ) of the cells compared with vector-treated controls , which correlated with low levels of p16 , p21 , and p27 expression .\n\nThe cells , however , were not immortalized and did eventually undergo senescence despite elevated Id-1 levels .\n\nSenescence was characterized by a dramatic increase in p16 , but not p21 and p27 .\n\nUnder these experimental conditions , telomerase activity was not detected and the telomeres became progressively shorter with time .\n\nThese results demonstrate the importance of Id-1 in endothelial cell proliferation and indicate that Id-1 represses p16 expression , resulting in delayed senescence .\n\nThese findings may have implications in the development of endothelial cell-derived tumors .", "output": "Enabling replicative immortality" }, { "input": "The HOX11 gene encodes a homeodomain transcription factor that is essential for spleen development during embryogenesis .\n\nHOX11 is also leukaemogenic , both through its clinical association with childhood T-cell acute lymphoblastic leukaemia , and its ability to immortalize other haematopoietic cell lineages experimentally .\n\nTo examine the pathological role of HOX11 in tumorigenesis , we constitutively expressed HOX11 cDNA in J2E murine erythroleukaemic cells , which are capable of terminal differentiation .\n\nEnforced HOX11 expression was found to induce a profound alteration in J2E cellular morphology and differentiation status .\n\nOur analyses revealed that HOX11 produced clones with a preponderance of less differentiated cells that were highly adherent to plastic .\n\nMorphologically , the cells overexpressing HOX11 were larger and had decreased globin levels , as well as a reduction in haemoglobin synthesis in response to erythropoietin ( EPO ) .\n\nImmunocytochemical analysis confirmed the immature erythroid phenotype imposed by HOX11 , with clones transfected with HOX11 demonstrating expression of the c-Kit stem cell marker , while retaining EPO receptor expression .\n\nTaken together , these results show that HOX11 alters erythroid differentiation , favouring a less mature progenitor-like stage .\n\nThis supports the notion that disrupted haematopoietic cell differentiation is responsible for pre-leukaemic immortalization by the HOX11 oncoprotein .", "output": "Enabling replicative immortality" }, { "input": "The aim of this study was to extend our previous observations on the soy modulation of biochemical parameters in 7,12-dimethylbenz[a]anthracene ( DMBA)-induced rat mammary tumors , by simultaneously investigating the expression of estrogen receptor-alpha ( ERalpha ) , estrogen receptor-beta ( ERbeta ) , progesterone receptor ( PR ) , apoptosis , neu , and markers of cell proliferation , such as proliferating cell nuclear antigen ( PCNA ) by immunohistochemistry .\n\nThe percentage of ERalpha positive tumors was 65.8% in masses from control animals , and significantly dropped to 36.8% in tumors from soy treated rats ( P=0.010 ) .\n\nThe percentage of ERbeta positive tumors was 70.3% in masses from control animals vs. 50.0% in tumors from soy exposed animals ( P=0.066 ) .\n\nMoreover , the percentage of cases which were both ERalpha and ERbeta positive was significantly lower ( 17.6% ) in soy treated than in control animals ( 51.3% ) ( P=0.006 ) .\n\nThe percentage of PR positive tumors was 34.2% in control animals vs. 2.6% in tumors from soy treated rats ( P=0.0006 ) .\n\nThere were no statistically significant differences in the percentage of tumors positively stained for neu , apoptosis , or PCNA , in control vs. soy treated rats .\n\nHowever , when analyzing the reciprocal correlation among the different biochemical parameters we showed that , in treated animals , the majority of ERalpha positive tumors ( 91.7% ) were also PCNA positive ( P=0.036 ) .\n\nThe median percentage of PCNA positivity was significantly higher in ERalpha positive than in ERalpha negative tumors ( 25 vs. 5% ) ( P=0.0031 ) .\n\nMoreover , an association was found between PCNA and neu status since all neu positive tumors were also PCNA positive ( P=0.011 ) .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The vast majority of breast cancers are carcinomas that arise from mammary epithelial cells ( MECs ) .\n\nOne of the key early events in tumorigenic transformation is the ability of cells to overcome replicative senescence .\n\nHowever , the precise genetic changes that are responsible for this event in MECs is largely unknown .\n\nHere , we report that Bmi-1 , originally identified as a c-Myc cooperating oncoprotein , can bypass senescence , extend the replicative life span , and immortalize MECs .\n\nFurthermore , Bmi-1 was overexpressed in immortal MECs and several breast cancer cell lines .\n\nOverexpression of Bmi-1 in MECs led to activation of human telomerase reverse transcriptase ( hTERT ) transcription and induction of telomerase activity .\n\nTelomerase induction by Bmi-1 was an early event in the extension of the replicative life span and immortalization .\n\nBmi-1 was not overexpressed in hTERT-immortalized MECs , suggesting that Bmi-1 functions upstream of hTERT .\n\nAlthough , c-Myc has been reported to induce telomerase in MECs , Bmi-1 appeared to act independently of c-Myc binding sequences in the hTERT promoter .\n\nDeletion analysis of the Bmi-1 protein suggested that the RING finger , as well as a conserved helix-turn-helix-turn domain , were required for its ability to induce telomerase and immortalize MECs .\n\nThese data suggest that Bmi-1 regulates telomerase expression in MECs and plays a role in the development of human breast cancer .", "output": "Enabling replicative immortality" }, { "input": "When cells were incubated in the presence of both interferon-gamma ( IFN-gamma ) and all-trans retinoic acid ( ATRA ) , the concentration of IFN-gamma required to induce apoptosis of B-cell lymphoma cells was much lower than that required for myeloid or erythroid cell lines .\n\nThe concentration of IFN-gamma that effectively inhibited the proliferation of BALM-3 cells was 1/40 of that required for BALM-1 cells .\n\nSTAT-1 phosphorylation , IRF-1 mRNA and protein expression and RAR-beta expression were enhanced to a greater degree in BALM-3 cells treated with IFN-gamma and ATRA than in BALM-1 cells treated with IFN-gamma and ATRA , suggesting that these IFN-gamma related genes were involved in the induction of apoptosis of BALM-3 cells .", "output": "Resisting cell death" }, { "input": "Immunosuppression has been related to the incidence of tumor apparition , including endocrine tumors .\n\nThe intrasplenic ovarian tumor ( luteoma ) is a typical benign endocrine tumor that develops under high gonadotropin stimulation and , from the immunological perspective , is located in a critical organ involved in immune response .\n\nTo establish if immunosuppression could alter the development of this experimental tumor , the effects of cyclosporin A ( CsA ) and dexamethasone ( Dex ) were evaluated .\n\nAfter surgery , tumor-bearing and sham animals were kept without treatment for 4 weeks ; thereafter , they were distributed into CsA ( 25 mg/kg ) , Dex ( 0.1 mg/kg ) , or vehicle ( 75:25 castor oil:ethanol ) groups and were injected on alternate days for 50 days .\n\nBody weight was evaluated weekly .\n\nAnimals were sacrificed after a jugular vein blood sample was obtained .\n\nThymi were weighed .\n\nTumors were measured and placed in formaline for histological studies .\n\nSerum luteinizing hormone ( LH ) , follicle-stimulating hormone ( FSH ) , prolactin ( PRL ) , and estradiol were measured by radioimmunoassay .\n\nHematological parameters were determined .\n\nCsA induced a significant decrease in survival rates both in tumor-bearing and sham animals ( P < 0.01 ) .\n\nDex significantly impaired weight increase in both groups of animals .\n\nCsA induced a significant weight loss in sham animals , not observed in tumor-bearing animals .\n\nDex induced thymus weight loss in both groups , whereas CsA induced thymus weight loss only in sham animals .\n\nOnly Dex induced a decrease in lymphocyte number in both groups .\n\nCsA induced an increase in monocyte number only in sham animals .\n\nTreatments did not alter LH , FSH , or estradiol , whereas PRL was increased by CsA only in sham rats .\n\nNeither Dex nor CsA induced any significant variations in tumor volume , nor did they alter tumor histology .\n\nIn addition , no visible metastases or alterations in other organs were observed .\n\nWe conclude that , though immunological parameters were altered by the treatments , immunosuppressor drugs did not condition tumor development .\n\nIn addition , tumors secrete one or more factor/s that counteract CsA effect .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "The knowledge of alterations in regulation of autophagy during tumorigenesis may also help our understanding of its normal control .\n\nWe established an experimental system and reported recently that autophagic capacity , measured as the cell's capability of increasing segregation ( formation of autophagosomes ) and subsequent degradation of cytoplasmic quanta were highly increased in premalignant nodule cells 6 months after initiation by azaserine in the rat pancreas in vivo .\n\nIn the present study , we followed changes of these autophagic functions throughout the tumour progression .\n\nWe carried out electron-microscopic morphometrical analysis of the expansion of autophagic vacuole compartment and subcompartments induced by vinblastine ( an in vivo segregation enhancer ) , as well as their regression upon segregation-inhibitor cycloheximide post-treatment .\n\nPremalignant tumour samples were taken at month 5 , month 8 ( nodules ) , month 10 and month 15 ( adenomas ) after initiation .\n\nIn all these stages , a highly increased and varying autophagic capacity was found compared with the host tissue .\n\nThe basal ( non-stimulated ) autophagic compartment was measurable only at month 5 and month 15 , and its regression upon cycloheximide was consistent with increased basal autophagic activity .\n\nCompared with the host tissue , autophagic capacity profoundly decreased in the differentiated and anaplastic adenocarcinomas at month 20 , when , surprisingly , cycloheximide was unable to inhibit segregation .\n\nOur conclusion is that down-regulation of the cycloheximide sensitive segregation and a partly compensatory up-regulation of an alternative pathway of segregation might occur along with malignant transformation .", "output": "Resisting cell death" }, { "input": "Dimethyl sulfoxide ( DMSO ) , a well-known differentiation inducer in several myeloid cells , also induces a reversible G(1) arrest in many cell lines .\n\nWe recently showed that DMSO induces a G(1) phase arrest in Chinese hamster ovary ( CHO ) cells , by restoring contact inhibition and preventing high density-dependent apoptosis .\n\nCHO cells are frequently used in cell biology and mutagenesis studies due to their good growth capacity and ease of manipulation but are very difficult to synchronize by serum starvation since they detach from monolayers when they reach confluence .\n\nIn this study we investigated the possibility of using DMSO to reversibly synchronize CHO cells in the G(1) phase of the cell cycle and analysed whether toxic effects follow the arrest using growth curve , sister chromatid exchange and micronuclei assays .\n\nWe carried out a kinetic analysis of the arrest by DMSO and re-entry into the cell cycle after drug release by cytofluorimetric analysis of DNA content and bromodeoxyuridine incorporation .\n\nWe show that CHO cells are efficiently and reversibly arrested in G(1) by DMSO in concentrations ranging between 1 and 2% .\n\nIn our experiments , >90% of cells grown for 96 h in presence of the drug were arrested in G(1) and synchronously re-entered S phase approximately 8-12 h after release .\n\nFurthermore , expression levels of p27 were down-regulated during G(1) progression and cyclin D3 and E expression patterns were similar to those observed after serum starvation .\n\nNo detectable cytotoxicity or genetic damage were induced in G(1) released cells as revealed by the tests employed .\n\nOur results show that DMSO is a very powerful inducer of G(1) synchronization in CHO cells without detectable cytotoxic or genetic effects in cell populations released from G(1) arrest .\n\nDMSO synchronization represents a model system in which to analyse protein activities regulating G(1) progression and investigate the response of G(1) cells to mutagen treatments .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The widespread use of synthetic musk fragrances and the resultant presence of these substances and their metabolites in the aquatic environment ( as well as their accumulation in human adipose tissue ) raises the question of whether musk fragrances display endocrine and in particular estrogenic activity .\n\nA variety of musk fragrances were tested using the E-screen assay .\n\nA statistically significant increase in proliferation rate of human MCF-7 breast cancer cells was detected for two nitro musks ( musk xylene , musk ketone ) , a major metabolite of musk xylene ( p-amino-musk xylene ) , and the polycyclic musk fragrance AHTN .\n\nThis indicates that these substances do , in fact , demonstrate estrogenic activity .\n\nCoincubation with the antiestrogen tamoxifen showed that the increase in proliferation rate by the musk fragrances is estrogen receptor-mediated .\n\nIt must be noted , however , that the effective estrogenic strength and estrogenic potency were low compared to 17 b-estradiol .\n\nThe naturally occurring fragrance muscone from the group of macrocyclic musk fragrances , a group of substances that have not yet been well characterized in respect to their toxicological properties , has also been shown to be weakly estrogenically active in vitro .\n\nE-screen analysis showed that the nitro musk metabolites o-amino musk xylene and 2-amino-MK , the macrocyclic musk fragrances ethylene brassylate , ethylene dodecandioate , and cyclopentadecanolide , are not estrogenically active .", "output": "Sustaining proliferative signaling" }, { "input": "Mutations in the Nf2 tumor suppressor gene lead to tumor formation in humans and mice and cellular overproliferation phenotypes in Drosophila .\n\nThe Nf2 encoded protein , merlin , shares close sequence similarity in its amino terminus to members of the band 4.1 family of membrane-cytoskeletal linkers .\n\nSimilarities between merlin and this family suggest a role for merlin in regulating cytoskeletal function .\n\nHowever , the mechanism of the tumor suppressing activity of merlin is not yet understood .\n\nMutational analysis of Nf2 in flies has led to the identification of a dominant-negative allele , which harbors mutations in the amino terminus of the protein .\n\nHere , we report that expression of a murine analog of this amino-terminal mutant of Nf2 leads to complete transformation of NIH3T3 fibroblasts in culture .\n\nCells that express this Nf2 mutant allele display disruptions of the actin cytoskeleton , lack of contact inhibition of growth , and anchorage-independent growth .\n\nFinally , fibroblasts that express this mutant Nf2 allele form tumors when injected into nude mice .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "p107 Links to cyclin A/CDK2 ( cyclin-dependent kinase 2 ) and cyclin E/CDK2 that are important cell cycle regulators .\n\nHowever , p107 expression remains unclear in almost all kinds of human solid tumours .\n\nTo clarify the expression of p107 in colorectal tumours , 22 normal mucosae , 9 hyperplastic polyps , 60 adenomas , 198 primary carcinomas , 21 lymph-nodal metastases , and 10 hepatic metastases were immunohistochemically stained for p107 , cyclin A , cyclin E , CDK2 and Ki67 .\n\nResults were measured using labelling indices ( LIs). p107 LIs surpassed the highest value in normal tissues in six of nine hyperplastic polyps , 54 of 60 adenomas , 144 of 198 primary cancers , 13 of 21 nodal foci and three of 10 hepatic foci. p107 LIs also apparently rose from normal through hyperplasia and adenoma to early carcinoma .\n\nHowever , they declined in liver-metastatic foci , and in primary cancers showing large size , mucinous type , venous invasion , lymphatic invasion , poorly differentiated type , deep invasion , lymph-nodal metastasis , hepatic metastasis or advanced stage .\n\nLow p107 LIs were also linked to a poor survival , particularly in stage-III patients .\n\nAs the p107 LI gradually rose , the CDK2 ( in primary cancers only ) , cyclin A , cyclin E and Ki67 LIs were elevated concurrently-in both adenomas and primary cancers .\n\nThus , in colorectal tumours , p107 expression rises abnormally and gradually during carcinogenesis and then falls during invasion , and thereby probably perturbs the cell-cycle control and promotes carcinogenesis and invasion .\n\nClinically , reduced p107 may indicate a poorer prognosis .", "output": "Activating invasion and metastasis" }, { "input": "Cellular senescence , initially observed during subculturing of normal diploid fibroblasts , can also be induced by chronic exposure to cellular stress , such as UV light , oxidative stress , or DNA damaging agents .\n\nHere we demonstrate that stable expression of an activated form of MKK6 ( MKK6EE ) , a direct activator of the stress-induced p38(HOG) mitogen-activated protein kinase pathway , is sufficient for inducing features of senescence including a flattened , vacuolated , and irregular morphology , staining for acidic beta-galactosidase , and accumulation of age-associated pigments .\n\nConsistent with the senescent phenotype , p38(HOG) activation induces a G(1) cell cycle arrest , which is permanent and irreversible after 4 days .\n\nMKK6EE also induces biochemical features of senescence in a p38-dependent manner , including enhanced expression of p21(CIP) , a cyclin-dependent kinase inhibitor .\n\nMicroarray analysis of MKK6EE cells showed a pattern of gene expression noted previously in Werner Syndrome and senescent fibroblasts .\n\nThese results define p38(HOG) as an intracellular pathway that activates a senescence checkpoint in tumor cells and may play a role in Ras- or stress-induced senescence .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "Malignant mesothelioma ( MM ) still remains a therapeutic and diagnostic problem to which new therapeutic perspectives are being continuously tried and tested .\n\nThree different primary cultures ( MMGe-1 , MES MM 98 , and MES 1 ) and one immortalized cell line ( MSTO 211 H ) of human MM were studied in order to evaluate the HER-2/neu expression .\n\nThree out of four cell lines showed a different level of c-erbB-2 expression , the highest being detected on the MSTO 211 H cell line ( fibroblastic phenotype ) , whereas MMGe-1 resulted negative .\n\nThe effect of the anti-HER-2/neu antibody ( Trastuzumab ) alone , and in combination with cisplatin ( CDDP ) at different doses ( ranging from 0.1 to 100 microg/ml ) , was studied on all the c-erB-2 positive cell lines .\n\nTrastuzumab was able to inhibit cell proliferation in a time-dependent manner , with growth inhibition also obtained at low concentrations ( 0.1-1 microg/ml ) .\n\nCombined treatment with Trastuzumab ( 10 microg/ml ) and CDDP ( 1 microg/ml ) showed synergism .\n\nOur results were encouraging , and suggest a rationale for further investigations in a clinical setting .", "output": "Sustaining proliferative signaling" }, { "input": "Enhanced glycolysis represents a striking feature of cancers and can therefore serve to indicate a malignant transformation .\n\nWe have developed a noninvasive , quantitative method to characterize tumor glycolysis by monitoring ( 13)C-labeled glucose and lactate with magnetic resonance spectroscopy .\n\nThis method was applied in MCF7 human breast cancer implanted in the mammary gland of female CD1-NU mice and was further employed to assess tumor response to hormonal manipulation with the antiestrogen tamoxifen .\n\nAnalysis of the kinetic data based on a unique physiological-metabolic model yielded the rate parameters of glycolysis , glucose perfusion , and lactate clearance in the tumor , as well as glucose pharmacokinetics in the plasma .\n\nTreatment with tamoxifen induced a twofold reduction in the rate of glycolysis and of lactate clearance but did not affect the other parameters .\n\nThis metabolic monitoring can thus serve to evaluate the efficacy of new selective estrogen receptor modulators and may be further extended to improve diagnosis and prognosis of breast cancer .", "output": "Cellular energetics" }, { "input": "PD-1 is a receptor of the Ig superfamily that negatively regulates T cell antigen receptor signaling by interacting with the specific ligands ( PD-L ) and is suggested to play a role in the maintenance of self-tolerance .\n\nIn the present study , we examined possible roles of the PD-1/PD-L system in tumor immunity .\n\nTransgenic expression of PD-L1 , one of the PD-L , in P815 tumor cells rendered them less susceptible to the specific T cell antigen receptor-mediated lysis by cytotoxic T cells in vitro , and markedly enhanced their tumorigenesis and invasiveness in vivo in the syngeneic hosts as compared with the parental tumor cells that lacked endogenous PD-L .\n\nBoth effects could be reversed by anti-PD-L1 Ab .\n\nSurvey of murine tumor lines revealed that all of the myeloma cell lines examined naturally expressed PD-L1 .\n\nGrowth of the myeloma cells in normal syngeneic mice was inhibited significantly albeit transiently by the administration of anti-PD-L1 Ab in vivo and was suppressed completely in the syngeneic PD-1-deficient mice .\n\nThese results suggest that the expression of PD-L1 can serve as a potent mechanism for potentially immunogenic tumors to escape from host immune responses and that blockade of interaction between PD-1 and PD-L may provide a promising strategy for specific tumor immunotherapy .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND Thrombospondin-1 ( TSP-1 ) promotes breast cancer cell invasion of collagen by upregulating matrix metalloproteinase-9 ( MMP-9 ) production .\n\nStromal TSP-1 may play a role in regulating tumor cell invasion .\n\nWe hypothesize that fibroblasts promote breast cancer cell invasion by upregulating the production of MMP-9 through TSP-1 .\n\nMETHODS MDA-MB-231 human breast carcinoma cells were grown alone or in coculture with human fibroblasts .\n\nGelatin zymography and Western immunoblot analysis for MMP-9 were performed on the coculture cell media and the single cell media .\n\nInhibition of fibroblast-mediated breast tumor cell invasion by an anti-TSP-1 or an anti-MMP-9 antibody was evaluated using a modified Boyden chamber .\n\nRESULTS Coculture experiments showed an increased production of MMP-9 when compared with breast cancer single cell culture or fibroblast single cell culture experiments as demonstrated by zymography and Western immunoblot analysis .\n\nFibroblast-stimulated MMP-9 production was comparable with TSP-1-stimulated MMP-9 production .\n\nAnti-TSP-1 antibody and anti-MMP-9 antibody inhibited fibroblast-stimulated tumor cell invasion to 30% and 26% of controls , respectively ( P <.05 ) .\n\nCONCLUSIONS Fibroblasts may regulate breast cancer cell invasion by promoting tumor MMP-9 production through TSP-1 .\n\nInhibition of stromal TSP-1 stimulation of MMP-9 synthesis may prevent matrix degradation necessary for tumor invasion and metastasis .", "output": "Activating invasion and metastasis" }, { "input": "The metastatic spread of malignant neoplasms is associated with active migration of cancer cells .\n\nThe migration of neoplastic cells during the metastatic process may be affected by various extracellular factors , including chemoattractants , haptotactic signals , electric fields , substrate anisotropy , and cell-to-cell contacts .\n\nWe examined the effect of homotypic collisions and heterotypic interactions with normal human skin fibroblasts on the motile activity of Walker carcinosarcoma cells .\n\nIt was found that Walker carcinosarcoma cells moving in a dense population neither show contact inhibition of movement when colliding with one another nor increase their motile activity as a result of contact stimulation of motility .\n\nOn the other hand , when plated onto the surface of aligned fibroblasts , Walker carcinosarcoma cells migrated mainly along the long axes of underlying fibroblasts as a result of contact guidance .\n\nThe directional character of movement ( but not the speed of migration ) of Walker carcinosarcoma cells on the surface of aligned fibroblasts was completely effaced by RGD-containing synthetic peptide at a concentration of 1 mg/ml but not by 5 microM verapamil ( selective voltage-gated calcium channel inhibitor ) or 10 microM gadolinium chloride ( non-specific blocker of mechanosensitive ion channels ) .\n\nThe suppression of directional character of migration of tumour cells by RGD-containing peptide was associated with the decrease in the amount of fibronectin macromolecules attached to fibroblasts .\n\nThis suggests that alignment and anisotropic distribution of fibronectin macromolecules may be responsible for contact guidance of tumour cells moving on the surface of fibroblasts .", "output": "Evading growth suppressors" }, { "input": "The G protein-coupled human gastrin-releasing peptide receptor ( hGRP-R ) is frequently found aberrantly expressed in human cancers of the colon , stomach , and lung , and its ligand-specific activation has been implicated in cell proliferation and differentiation .\n\nHere , we demonstrated hGRP-R activation stimulated sustained cyclic AMP response element binding protein ( CREB ) phosphorylation and transactivation in duodenal cancer cells through a protein kinase C and partially p38 mitogen-activated protein kinase-dependent pathway .\n\nIn contrast , intracellular calcium , ERK1/2 , protein kinase A , and PI3 kinase were not involved .\n\nThis novel signaling mechanism might be of importance for regulation of CREB-dependent gene expression in human cancer expressing functional hGRP-R .", "output": "Sustaining proliferative signaling" }, { "input": "A rat colonic adenocarcinoma was implanted subcutaneously ( s.c. ) in nude mice .\n\nAfter 7 days , the animals were divided into different groups .\n\nTwo groups received subcutaneous injections twice daily with 3 or 6 micro g/kg body weight octreotide , galanin and serotonin .\n\nThree groups were respectively treated with 20 , 30 , and 40 micro g/kg body weight of the previously mentioned bioactive substances .\n\nControl group received only saline solution in the same fashion as treated animals .\n\nThe treatment lasted for 5 days .\n\nThe tumour volume and weight , the relative density of blood vessels , of tumour necrotic tissue , of apoptotic nuclei and of proliferating nuclei were measured .\n\nApoptosis was detected by in situ labelling of nuclear DNA fragmentation according to TUNEL method , and proliferation by immunocytochemistry .\n\nMorphometry was done with the classical stereological point-counting method .\n\nFood consumption , animal weight , faeces weight and its water content were measured for 3 days before and after treatment .\n\nTriple therapy with 3 and 6 micro g/kg body weight had no effect on any of the parameters measured , except in reducing the relative volume density of tumour blood vessels .\n\nTreatment with 20 , 30 and 40 micro g/kg body weight of the previously mentioned bioactive substances reduced the tumour volume , the relative volume density of blood vessels and increased the relative volume density of necrotic tissue and of apoptotic nuclei ( in the 20 micro g group ) .\n\nHowever , there was no difference between treated mice and controls regarding the relative volume density of proliferating nuclei .\n\nThere was no statistical difference between treated animals regarding food consumption , body weight , faeces weight and its water content before and during treatment .\n\nThe present study confirms that triple therapy with octreotide , galanin and serotonin causes regression of a rat colon carcinoma .\n\nIt further showed that optimum treatment dose is 20 micro g/kg body weight of each bioactive substance .\n\nMoreover , this therapy regime does not show apparent side effects in the experiments carried out on mice .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "A cell line designated HUUCLEC was established from a human uterine cervical lymphoepithelial carcinoma obtained from a 61-year-old Japanese woman .\n\nThe cell line has grown slowly without interruption and serial passages were successively carried out 60 times within 3 years .\n\nThe cultured cells were spindle or round in shape , showing anaplastic and pleomorphic features , a pavement cell arrangement and multilayering without contact inhibition .\n\nThe population doubling time of the HUUCLEC line was 72 hours while the chromosomal number varied widely and showed aneuploidy .\n\nThe modal chromosomal number was stable at the triploid range and marker chromosomes were present ; the Ebstein-Barr virus was absent in the cultured cells .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "The ultraviolet radiation present in sunlight is immune suppressive .\n\nRecently we showed that solar-simulated ultraviolet radiation ( ultraviolet A + B ; 295-400 nm ) , applied after immunization , suppressed immunologic memory and the elicitation of delayed-type hypersensitivity to the common opportunistic pathogen , Candida albicans .\n\nFurther , we found that wavelengths in the ultraviolet A region of the solar spectrum ( 320-400 nm ) , devoid of ultraviolet B , were equally effective in activating immune suppression as ultraviolet A + B radiation .\n\nHere we report on the mechanisms involved .\n\nMaximal immune suppression was found when mice were exposed to solar-simulated ultraviolet radiation 7-9 d post immunization .\n\nNo immune suppression was found in ultraviolet-irradiated mice injected with monoclonal anti-interleukin-10 antibody , or mice exposed to solar-simulated ultraviolet radiation and injected with recombinant interleukin-12 .\n\nSuppressor lymphocytes were found in the spleens of mice exposed to ultraviolet A + B radiation .\n\nIn addition , antigen-specific suppressor T cells ( CD3+ , CD4+ , DX5+ ) were found in the spleens of mice exposed to ultraviolet A radiation .\n\nApplying liposomes containing bacteriophage T4N5 to the skin of mice exposed to solar-simulated ultraviolet A + B radiation , or mice exposed to ultraviolet A radiation , blocked immune suppression , demonstrating an essential role for ultraviolet-induced DNA damage in the suppression of established immune reactions .\n\nThese findings indicate that overlapping immune suppressive mechanisms are activated by ultraviolet A and ultraviolet A + B radiation .\n\nMoreover , our findings demonstrate that ultraviolet radiation activates similar immunologic pathways to suppress the induction of , or the elicitation of , the immune response .", "output": "Genomic instability and mutation, Avoiding immune destruction" }, { "input": "Exposure to ultraviolet B ( UVB ) radiation ( 280-320 nm ) is the primary etiologic factor associated with the development of basal cell carcinoma ( BCC ) .\n\nThe outgrowth of these keratinocyte-derived skin lesions is enhanced by the ability of UVB to impair an immune response that would otherwise eliminate them .\n\nStudies in a range of inbred mouse strains as well as mast cell-depleted mice reconstituted with mast cell precursors support a functional link between histamine-staining dermal mast cells and the extent of susceptibility to UVB-induced systemic immunomodulation .\n\nHumans , like mouse strains , display variations in dermal mast cell prevalence .\n\nIn a study of Danish and South Australian BCC patients and control subjects , one 4-mm punch biopsy of non-sun-exposed buttock skin was sampled from each participant .\n\nThis skin site was investigated to avoid any changes in mast cell prevalence caused by sun exposure .\n\nTwo sections ( 4 microm ) per biopsy were immunohistochemically stained for detection of histamine-containing dermal mast cells .\n\nComputer-generated image analysis evaluated dermal mast cell prevalence in both sections by quantifying the total number of mast cells according to the total dermal area ( expressed as mast cells per square millimeter ) .\n\nThis technique enabled us to detect heterogeneity of dermal mast cell prevalence in buttock skin between individuals and provided evidence of an association between high dermal mast cell prevalence and BCC development in two diverse populations .\n\nWe hypothesize that mast cells function in humans , as in mouse strains , by initiating immunosuppression following UV irradiation and , thereby , allowing a permissive environment for the development of BCC .\n\nThus , a high dermal mast cell prevalence as demonstrable in buttock skin is a significant predisposing factor for development of BCC in humans .", "output": "Avoiding immune destruction" }, { "input": "OBJECTIVE Overexpression of epidermal growth factor receptor ( EGFR ) in glioblastoma multiforme ( GBM ) secondary to EGFR gene amplification is associated with a more aggressive tumor phenotype and a worse clinical outcome .\n\nThe purpose of this study was to analyze whether blocking this receptor with the anti-EGFR chimeric monoclonal antibody C225 would decrease proliferation and increase apoptosis in GBM cells .\n\nMETHODS EGFR expression and amplification were analyzed for seven human GBM cell lines .\n\nThese lines were then exposed to different concentrations of C225 for 48 hours , 72 hours , and 7 days , after which time cytotoxicity , apoptosis , and vascular endothelial growth factor expression were assessed in vitro .\n\nTwo EGFR-amplified human GBM were implanted in the flanks of nude mice , and the animals received C225 twice per week intraperitoneally for 5 weeks .\n\nTumor volumes and survival times were compared with those of sham-treated mice .\n\nRESULTS EGFR gene amplification was demonstrated in three of the primary GBM lines .\n\nC225 treatment produced significant cytotoxicity in all three EGFR-amplified GBM lines , but not in unamplified lines .\n\nFlow cytometry demonstrated increased apoptosis in C225-treated , EGFR-amplified GBM lines , but not in unamplified lines .\n\nThere was a decrease in vascular endothelial growth factor expression in all GBM lines with exposure to C225 .\n\nTumor-bearing mice treated with C225 experienced significant inhibition of tumor growth as well as a 200% increase in median survival .\n\nCONCLUSION Blocking EGFR in GBM cells that overexpress this receptor significantly changes tumor cell biology by promoting apoptosis while decreasing proliferation and vascular endothelial growth factor expression .\n\nThis approach holds great promise for the treatment of patients with GBMs .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "We showed in a previous study that soluble low-molecular-mass tumor-associated antigens ( sTAA ) promote the anti-tumor effect of the anticancer drug cyclophosphamide ( CPA ) on rat mammary carcinogenesis .\n\nIn this report , we analyzed the possible mechanism underlying this phenomenon .\n\nStudies were performed on the bone marrow and thymus from the following groups of rats : i ) control rats , ii ) rats treated with sTAA , iii ) rats treated with CPA , iv ) rats treated with CPA and sTAA .\n\nThe cellular content of the bone marrow and thymus ( CD4+ and CD8+ lymphocytes ) was analyzed morphometrically and immunohistochemically .\n\nIn the bone marrow , CPA caused significant substitution of cellular components with fatty tissue whereas sTAA repaired this process .\n\nWe found that CPA affects mainly the process of myelogenesis whereas sTAA protect the production of lymphocytes .\n\nIn the thymus , CPA alone or in combination with sTAA repaired the inhibition effect of DMBA on synthesis of CD4+ and CD8+ thymocytes. sTAA further increased the amount of CD8+ T lymphocytes in the medulla of the thymus .\n\nData in the literature as well as the findings presented here demonstrate that the tested treatment , including vaccination with sTAA , actively promotes the generation of the host's antitumor immune response .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND/AIMS Endoscopic mucosal resection and laparoscopic wedge resection have become more common in the treatment of early gastric cancer .\n\nHowever , lymph node metastasis is a major poor prognostic factor influencing tumor recurrence and survival .\n\nTo predict the risk of lymph node metastasis in early gastric cancer , the authors conducted a study to investigate the clinicopathologic characteristics of early gastric cancer with lymph node metastasis .\n\nMETHODOLOGY From 1982 to 1998 , 181 patients of early gastric cancer underwent primary surgery and were included in the study .\n\nPatient data was postoperatively reviewed regarding age , gender , tumor size , depth of invasion , histologic differentiation , macroscopic classification and anatomic level of lymph node metastasis .\n\nThe chi 2 test or Student's t test was used for statistical analysis .\n\nLogistic regression analysis was used to evaluate the independent risk factors for lymph node metastasis .\n\nRESULTS Lymph node metastasis was observed in 19 cases ( 11% ) .\n\nEarly gastric cancer with size larger than 4 cm ( P < 0.05 ) , with submucosal invasion ( P < 0.01 ) , and with poor differentiation ( P < 0.05 ) was associated with higher risk of lymph node metastasis .\n\nThe macroscopic classification had no predictive value .\n\nMultivariate analysis showed that submucosal invasion correlated best with lymph node spread ( OR 10.25 , 95% CI : 2.10-49.96 ) , followed by tumor size larger than 4 cm ( OR 4.99 , 95% CI : 1.46-17.05 ) , and poorly differentiated histological subtype ( OR 3.31 , 95% CI : 1.16-9.45 ) .\n\nCONCLUSIONS Poor differentiation , submucosal invasion and large tumor size were independent risk factors for lymph node metastasis in early gastric cancer .\n\nMacroscopic classification was not correlated with lymph node metastasis .", "output": "Activating invasion and metastasis" }, { "input": "Glycolysis is known to be the primary energy source in most cancer cells .\n\nWe investigated here the effect of clotrimazole ( 1-(alpha-2-chlorotrityl)imidazole ) , the antifungal azole derivative , which was recently recognized as calmodulin antagonist , on the levels of glucose 1,6-bisphosphate and fructose 1,6-bisphosphate , the two stimulatory signal molecules of glycolysis , and on ATP content and cell viability in LL/2 Lewis lung carcinoma cells and CT-26 colon adenocarcinoma cells .\n\nWe found that clotrimazole induced a significant , dose- and time-dependent reduction in the levels of glucose 1,6-bisphosphate , fructose 1,6-bisphosphate , ATP , and cell viability .\n\nThese findings suggest that clotrimazole causes a reduction in glycolysis and ATP levels , which eventually leads to cell destruction after 3 h of treatment .\n\nSince cell proliferation was also reported to be inhibited by calmodulin antagonists , this substance is most promising agent in treatment of cancer by inhibiting both cell proliferation and the glycolytic supply of ATP required for cancer cell growth .", "output": "Cellular energetics" }, { "input": "The immune system of vertebrates is able to detect bacterial DNA based on the presence of unmethylated CpG motifs .\n\nWe examined the therapeutic potential of oligodeoxynucleotides with CpG motifs ( CpG ODN ) in a colon carcinoma model in BALB/c mice .\n\nTumors were induced by s.c. injection of syngeneic C26 cells or Renca kidney cancer cells as a control .\n\nInjection of CpG ODN alone or in combination with irradiated tumor cells did not protect mice against subsequent tumor challenge .\n\nIn contrast , weekly injections of CpG ODN into the margin of already established tumors resulted in regression of tumors and complete cure of mice .\n\nThe injection site was critical , since injection of CpG ODN at distant sites was not effective .\n\nMice with two bilateral C26 tumors rejected both tumors upon peritumoral injection of one tumor , indicating the development of a systemic immune response .\n\nThe tumor specificity of the immune response was demonstrated in mice bearing a C26 tumor and a Renca tumor at the same time .\n\nMice that rejected a tumor upon peritumoral CpG treatment remained tumor free and were protected against rechallenge with the same tumor cells , but not with the other tumor , demonstrating long term memory .\n\nTumor-specific CD8 T cells as well as innate effector cells contributed to the antitumor activity of treatment .\n\nIn conclusion , peritumoral CpG ODN monotherapy elicits a strong CD8 T cell response and innate effector mechanisms that seem to act in concert to overcome unresponsiveness of the immune system toward a growing tumor .", "output": "Avoiding immune destruction" }, { "input": "OBJECT The intracellular events transducing mitogenic signals from platelet-derived growth factor-beta ( PDGFbeta ) receptor tyrosine kinases are not precisely known .\n\nIn this study the authors evaluated whether the phosphatidylinositol 3-kinase ( PI3-K)-Akt-p70S6K pathway is expressed in meningiomas , regulates their growth , and transduces mitogenic signals of PDGF-BB .\n\nMETHODS Nine meningioma tumors obtained in humans were evaluated using Western blot analysis for phosphorylated ( activated ) Akt and phosphorylated p70S6K .\n\nCells cultured from seven of these meningiomas were also screened using Western blot analysis for Akt and for phosphorylated Akt and p70S6K .\n\nThe authors also evaluated whether PDGF-BB stimulation of meningioma cells was associated with the phosphorylation of Akt and p70S6K known to activate these kinases .\n\nIn addition , the effects of wortmannin , an inhibitor of P13-K , on proliferation and activation of Akt and p70S6K in meningioma cells stimulated with PDGF-BB were evaluated .\n\nWestern blots of lysates from meningiomas demonstrated phosphorylated Akt and p70S6K .\n\nTreatment with PDGF-BB stimulated phosphorylation of Akt and p70S6K in each meningioma cell culture .\n\nWortmannin ( 500 and 1000 nM ) significantly decreased PDGF-BB stimulation of meningioma cells ( p < 0.001 ) while it reduced Akt and p70S6K phosphorylation but not mitogen-activated protein kinase/extracellular signal-regulated kinase ( MAPK/ERK ) phosphorylation .\n\nCONCLUSIONS These findings indicate that Akt and p70S6K are constitutively expressed and activated in meningioma cells and that the PI3-K-Akt-p70S6K pathway may participate in transduction of mitogenic signals in meningiomas independent of the Raf-1-MEK-1-MAPK/ERK cascade .", "output": "Sustaining proliferative signaling" }, { "input": "It is still difficult to decide on the treatment modalities for advanced esophageal carcinoma when the prognostic factors of T4 esophageal cancer are not fully understood .\n\nIn this article , we report that among 71 patients with T4 thoracic esophageal cancer , 49 underwent esophagectomy , 9 had curative resection ( R0 group ) , and 40 had palliative resection ( R1/2 group ) .\n\nA total of 22 patients had palliative treatments : bypass in 5 ( bypass group ) , gastrostomy or jejunostomy in 6 ( stoma group ) , and radiochemotherapy alone in 11 ( nonoperation group ) .\n\nClinicopathologic characteristics were retrospectively investigated .\n\nTreatment-related deaths occurred in 7 ( 10% ) : none in R0 , 3 ( 8% ) in R1/2 , 3 ( 60% ) in bypass , and 1 ( 17% ) in stoma group .\n\nSwallowing was improved in 50 ( 70% ) patients : 9 ( 100% ) in R0 , 30 ( 75% ) in R1/2 , 1 ( 20% ) in bypass , 3 ( 50% ) in stoma , and 7 ( 64% ) in the nonoperation group .\n\nOne- , two- , and three-year overall survival rates were 56% , 22% , and 22% in the R0 group and 35% , 19% and 6% in the R1/2 group , respectively ( p = 0.19 ) .\n\nIn the bypass , stoma , and nonoperation groups , none survived 1.6 years .\n\nThe factors influencing the survival rate of the 49 patients undergoing esophagectomy were grade of lymph node metastasis , amount of perioperative blood transfusion , lymph vessel , and blood vessel invasion .\n\nAmong these , independent prognostic factors for survival were amount of blood transfusion ( -6 units vs. -7 units , p <0.0001 ) and grade of lymph node metastasis [ none- or peritumoral [ lymph nodes adjacent to the main tumor or at a nearby location ( <3 cm ) from the tumor ] metastasis vs. more distant metastasis [ lymph nodes at a distant location ( > 3 cm) ] , p = 0.016 ] .\n\nBypass and stoma operation neither prolonged the survival nor improved the difficulty of swallowing compared with radiochemotherapy alone .\n\nEsophagectomy can achieve the best improvement of swallowing and the longest survival with an acceptable mortality rate .\n\nEsophageal carcinoma patients with T4 disease and distinct metastasis in the lymph nodes at a distant location ( >3 cm ) from the primary tumor may not benefit from an esophageal resection .", "output": "Activating invasion and metastasis" }, { "input": "A key issue in the development of the central nervous system ( CNS ) is understanding the molecular mechanisms regulating cell number .\n\nThe present study examines the role of CD81 ( previously known as TAPA , the target of the antiproliferative antibody ) in the control of brain size and glial cell number .\n\nCD81 is a member of the tetraspanin family of proteins .\n\nThis group of small membrane proteins is associated with the regulation of cell migration and mitotic activity .\n\nGlial cells express CD81 , and antibodies directed against this protein suppress the mitotic activity of cultured cells .\n\nIn this study , we examine the effects of the CD81 -/- mutation on the CNS of mature mice .\n\nThese mice have extremely large brains , as much as 30% larger than the brains of wild-type ( +/+ ) littermates .\n\nThe increase in brain weight is accompanied by an increase in the number astrocytes and microglia , whereas the number of neurons and oligodendrocytes in the CD81 -/- animals appears to be normal .\n\nWhen the CD81 -/- mutation is placed on different genetic backgrounds , there is a remarkable range in the penetrance of the null allele phenotype , demonstrating that the mutation can be affected by modifier loci .\n\nThis work provides support for the role of CD81 in the regulation of astrocyte and microglial number , perhaps by regulating cell proliferation by a contact inhibition-dependent mechanism .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "We used cDNA-based genomic microarrays to examine DNA copy number changes in a panel of prostate tumors and found a previously undescribed amplicon on chromosome 17 containing a novel overexpressed gene that we termed prostate cancer gene 17 ( PRC17 ) .\n\nWhen overexpressed in 3T3 mouse fibroblast cells , PRC17 induced growth in low serum , loss of contact inhibition , and tumor formation in nude mice .\n\nThe PRC17 gene product contains a GTPase-activating protein ( GAP ) catalytic core motif found in various Rab/Ypt GAPs , including RN-Tre .\n\nSimilar to RN-Tre , we found that PRC17 protein interacts directly with Rab5 and stimulates its GTP hydrolysis .\n\nPoint mutations that alter conserved amino acid residues within the PRC17 GAP domain abolished its transforming abilities , suggesting that GAP activity is essential for its oncogenic function .\n\nWhereas PRC17 is amplified in 15% of prostate cancers , it is highly overexpressed in approximately one-half of metastatic prostate tumors .\n\nThe potent oncogenic activity of PRC17 is likely to influence the tumorigenic phenotype of these prostate cancers .", "output": "Genomic instability and mutation, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Hormone-independent tumor growth and metastasis are associated with increased mortality in human prostate cancer .\n\nIn this study , we evaluate a potential role for ligand-mediated activation of HER2 receptor tyrosine kinase in androgen-independent prostate cancers .\n\nHER2 , HER3 , and epidermal growth factor receptor were detected in the androgen-independent cell line 22Rv1 .\n\nHeregulin stimulation results in receptor phosphorylation and cell proliferation that is inhibited by increasing concentrations of anti-HER2 recombinant humanized monoclonal antibody ( rhuMAb ) 2C4 .\n\nFurthermore , inhibition of tumor growth was observed in xenografts derived from 22Rv1 cells when treated with rhuMAb 2C4 in a dose-dependent manner .\n\nThese studies provide a framework , both in vitro and in vivo , to examine the molecular mechanisms of ligand-driven HER2 activation in androgen-independent tumorigenesis .", "output": "Sustaining proliferative signaling" }, { "input": "We have previously reported the frequent occurrence of bile duct invasion by liver metastases from colorectal cancer .\n\nWe found that patients with macroscopic intrabiliary cancer growth survive longer after hepatectomy than those without this feature .\n\nIn the present study , we analyzed the clinicopathological features of primary colorectal cancer showing macroscopic intrabiliary extension of liver metastases .\n\nWe reviewed 217 patients who underwent initial hepatic resection for colorectal liver metastasis between 1992 and 1998 , and analyzed the corresponding primary colorectal cancers clinicopathologically .\n\nMicroscopic bile duct invasion was found in 89 of 217 cases ( 40.6% ) and , of these cases , 23 ( 10.6% ) had macroscopic intrabiliary extension .\n\nHistological sections of the corresponding primary colorectal cancer were available in eight ( group A ) of these 23 cases .\n\nThese were compared with 20 cases , selected randomly , of colorectal cancer that did not show bile duct invasion and were diagnosed as liver metastases .\n\nThese patients underwent hepatectomy during the same period as group A and were used as a control ( group B ) .\n\nThe histology of the primary tumors revealed well-differentiated adenocarcinoma in 100% of group A and in 25% of group B. The average maximum diameter of the primary tumor was 5.32 cm in group A and 3.61 cm in group B. Venous invasion was detected in 25% of group A and in 90% of group B ( P < 0.01 ) , while the incidences of lymphatic vessel invasion and lymph node metastases were similar between the groups .\n\nThese data suggest that macroscopic intrabiliary extension could be a good indicator of a unique subgroup of colorectal cancers showing less aggressive features even though they develop liver metastases .\n\nCareful histological evaluation is important even for metastatic tumors .", "output": "Activating invasion and metastasis" }, { "input": "Antithymocyte globulin is widely used before haematopoietic transplantation with HLA-matched unrelated donors or mismatched relatives to prevent rejection and graft-versus-host disease ( GVHD ) .\n\nHowever , optimal dosage is still under debate .\n\nThirty-one consecutive children , mainly with haematological malignancies , were transplanted in a single institution with such donors , selected by HLA-A -B compatibility by serology and DRB1* by DNA typing .\n\nAntithymocyte globulin ( Thymoglobuline ; Sangstat ) was infused at days -3 , -2 , -1 .\n\nTotal dosage varied : 16 patients received a median of 7.5 mg/kg ( 2.5 to 10.5 : low-dose group ) , and 15 a median of 15.5 mg/kg ( 14.4 to 19.4 : high-dose group ) .\n\nPost-transplant GVHD prophylaxis consisted of cyclosporine , short-course methotrexate and steroids .\n\nCD3(+) , CD4(+) and CD19(+) cell reconstitution was slower in the high-dose group .\n\nMedian time to reach 100 CD4(+) cells was 8 months vs 4 months ( P = 0.03 ) .\n\nMedian time to normal CD19(+) cells was 16 months vs 8 months ( P = 0.01 ) .\n\nCD16(+)CD56(+) and CD8(+) cell reconstitution was similar .\n\nNine patients in the high-dose group and two in the low-dose group experienced life-threatening opportunistic infections ( P = 0.009 ) .\n\nAlthough obtained from a limited number of patients , our data suggest that a higher pre-graft dose of antithymocyte globulin may negatively influence immune reconstitution .", "output": "Avoiding immune destruction" }, { "input": "Human fibroblasts undergo cellular senescence after a finite number of divisions , in response to the erosion of telomeres .\n\nIn addition to being terminally arrested in the cell cycle , senescent fibroblasts express genes that are normally induced upon wounding , including genes that remodel the extracellular matrix .\n\nWe have identified the novel zinc finger protein APA-1 , whose expression increased in senescent human fibroblasts independent of telomere shortening .\n\nExtended passage , telomerase-immortalized fibroblasts had increased levels of APA-1 as well as the cyclin-dependent kinase inhibitor p16 .\n\nIn fibroblasts , APA-1 was modified by the ubiquitin-like protein SUMO-1 , which increased APA-1 half-life , possibly by blocking ubiquitin-mediated degradation .\n\nOverexpression of APA-1 did not cause cell cycle arrest ; but , it induced transcription of the extracellular matrix-remodeling genes MMP1 and PAI2 , which are associated with fibroblast senescence .\n\nMMP1 and PAI2 transcript levels also increased in telomerase-immortalized fibroblasts that had high levels of APA-1 , demonstrating that the matrix-remodeling phenotype of senescent fibroblasts was not induced by telomere attrition alone .\n\nAPA-1 was able to transactivate and bind to the MMP1 promoter , suggesting that APA-1 is a transcription factor that regulates expression of matrix-remodeling genes during fibroblast senescence .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "We investigated the mode of cell death induced by the anthracyclines , aclarubicin , doxorubicin and daunorubicin in the human leukemia cell lines , HL60 and Jurkat .\n\nThe cells were incubated with drug concentrations up to 500 nM for periods between 3 and 24 hours , followed by morphological and biochemical analyses .\n\nAll three substances induced DNA fragmentation , evident as DNA laddering and appearance of cells with hypodiploid DNA content , externalization of phosphatidyl serine , activation of caspases and degradation of the apoptosis-specific endonuclease inhibitor DFF45 .\n\nHowever , concentrations and times necessary for these effects to occur were different , aclarubicin being the quickest acting drug with a lag phase of 3 h , followed by daunorubicin with 6 h and doxorubicin with 24 h .\n\nMore importantly , aclarubicin induced these effects while the cell membrane was intact , whereas doxorubicin and daunorubicin led to immediate loss of membrane integrity .\n\nProgrammed cell death is characterised by preservation of membrane integrity in order to allow removal of apoptotic bodies , whereas cell rupture is an early event in necrosis .\n\nWe therefore suggest that , in our experimental settings , doxorubicin- and daunorubicin-induced cell death occurs by necrosis , while aclarubicin induces programmed cell death .", "output": "Resisting cell death" }, { "input": "In human colorectal adenomas or polyps , cyclooxygenase-2 is expressed predominantly by stromal ( or interstitial ) macrophages .\n\nTherefore , we tested the hypothesis that macrophage cyclooxygenase-2 has paracrine pro-tumorigenic activity using in vitro models of macrophage-epithelial cell interactions .\n\nWe report that macrophages can promote tumorigenic progression of intestinal epithelial cells ( evidenced by decreased cell-cell contact inhibition , increased proliferation and apoptosis , gain of anchorage-independent growth capability , decreased membranous E-cadherin expression , up-regulation of cyclooxygenase-2 expression , down-regulation of transforming growth factor-beta type II receptor expression and resistance to the anti-proliferative activity of transforming growth factor-beta(1) ) in a paracrine , cyclooxygenase-2-dependent manner .\n\nPharmacologically relevant concentrations ( 1-2 microM ) of a selective cyclooxygenase-2 inhibitor had no detectable , direct effect on intestinal epithelial cells but inhibited the macrophage-epithelial cell signal mediating tumorigenic progression .\n\nCyclooxygenase-2-mediated stromal-epithelial cell signalling during the early stages of intestinal tumorigenesis provides a novel target for chemoprevention of colorectal cancer ( and other gastro-intestinal epithelial malignancies , which arise on a background of chronic inflammation , such as gastric cancer ) and may explain the discrepancy between the concentrations of cyclooxygenase inhibitors required to produce anti-neoplastic effects in vitro and in vivo .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "A hallmark of cancer cells is the ability to proliferate indefinitely .\n\nThis acquisition of an immortal lifespan usually requires the activation of telomerase , the enzyme that elongates telomeres .\n\nHuman telomerase is minimally composed of the reverse transcriptase subunit hTERT , and the RNA subunit hTR .\n\nWhile hTR is ubiquitously expressed in human cells , the hTERT subunit is generally transcriptionally repressed in most normal somatic cells , but is illegitimately activated to restore telomerase activity in cancer cells .\n\nIndeed , in the thousands of different human tumours assayed , 85% were scored positive for telomerase activity .\n\nHowever , the levels of telomerase activity detected in tumour samples can vary substantially and even some normal somatic cells have been found to have low levels of enzyme activity .\n\nAs the functional significance of low levels of telomerase activity is unclear , we investigated whether there is a minimum level of telomerase activity required for tumourigenesis .\n\nUsing mutants of hTERT that induce varying levels of telomerase activity , we show that there does indeed exist a threshold of activity required for the processes of immortalization , transformation and tumourigenesis .\n\nThus , low levels of activity detected in certain somatic cells would not be expected to contribute to tumourigenesis , nor does the mere detection of telomerase in cancer cells necessarily signify an immortal lifespan .", "output": "Enabling replicative immortality" }, { "input": "Telomerase activity , a cardinal requirement for immortalization , is a crucial step in the development of cancer and has been studied in many kinds of malignant tumours for clinical diagnostic and/or prognostic utilities .\n\nUsing a PCR-based TRAP assay , we investigated telomerase activity in 8 adenomatous polyps , 9 dysplastic polyps , and in 36 paired cancer-normal mucosa specimens , one liver and one spleen metastasis from patients resected for sporadic colorectal cancer .\n\nTelomerase was absent or very low in normal mucosa and in adenomatous polyps .\n\nDysplastic polyps and adenocarcinoma samples showed telomerase activity , with higher levels in cancer tissues compared to dysplastic lesions .\n\nA high telomerase activity was shown to be associated with late-staged cancers and metastasis , providing arguments supporting the role of telomerase not only in the development but also in the progression of colorectal carcinoma .\n\nMoreover , telomerase evaluation may help to confirm the malignant transformation in polypoid colorectal lesions with different levels of dysplastic alterations .", "output": "Activating invasion and metastasis" }, { "input": "To search for potential biomarkers used to monitor the process of immortalization , we investigated the relative level of telomerase activity and other immortal phenotypes in the SHEE esophageal epithelial cell line .\n\nThis human fetal esophageal epithelial cell line , induced by human papilloma virus ( HPV ) 18 E6E7 , was continually propagated over 100 passages .\n\nFourteenth passage cells ( SHEE14 ) were cultured in a flask with a serum-free medium and continually cultured to the 30th passage ( SHEE30 ) .\n\nCells of SHEE14 , SHEE20 and SHEE30 were examined according to cell morphology , cell cycle , apoptosis , contact-inhibition growth , anchorage- dependency , dose-dependency to epithelial growth factors ( EGF ) , telomerase activity and tumorigenicity .\n\nThe SHEE14 cells exhibited good differentiation with contact-inhibition and anchorage-dependent growth .\n\nThe SHEE20 cells exhibited increase of senescent and apoptotic cells , and difficulty in propagation .\n\nThe SHEE30 cells exhibited a higher proliferative index and some undifferentiated cells , with weakened contact-inhibition and anchorage-dependent growth .\n\nThe telomerase was activated in cells of SHEE30 , but not in SHEE14 and SHEE20 cells .\n\nThe different response to dose-dependency to EGF was not statistically different in SHEE14 and SHEE30 .\n\nThree groups of cells displayed lack of tumor formation in nude mice .\n\nCompared with SHEE14 and SHEE20 , SHEE30 cells were of immortalized status with immortal phenotype , which consisted of telomerase activity , increase of cell proliferation , weakened contact-inhibition and anchorage-dependent growth , dose dependency to EGF and lack of tumor formation .\n\nFrom passage 14 to 30th passage , SHEE cells went through cellular senescence , apoptosis and immortalization .\n\nWith a view toward diagnostic and biological aspects , telomerase activity is a crucial step and a cardinal requirement for immortalization .\n\nThe telomerase activity and other immortal phenotypes are potential markers for monitoring the process of immortalization .", "output": "Enabling replicative immortality, Evading growth suppressors, Resisting cell death" }, { "input": "Environmental carcinogen exposure may play an important role in the incidence of cancer in children .\n\nIn addition to environmental pollutants , maternal smoking during pregnancy may be a contributing factor .\n\nMajor carcinogenic components of cigarette smoke and other combustion by-products in the environment include polycyclic aromatic hydrocarbons ( PAH ) .\n\nMouse offspring exposed during midpregnancy to the PAH , benzo[a]pyrene ( B[a]P ) , show significant deficiencies in their immune functions , observed in late gestation which persist for at least 18 months .\n\nTumor incidences in these progeny are 8 to 10-fold higher than in controls .\n\nWe have demonstrated a significant reduction in thymocytes ( CD4+ CD8+ , CD4+ CD8+ Vbeta8+ , CD4+ CD8+ Vgamma2+ ) from newborn and splenocytes ( CD4+ CD8+ ) from 1-week-old mouse progeny exposed to B[a]P in utero .\n\nTo investigate possible causes of the observed T cell reduction , we analyzed the thymocytes and splenocytes from progeny and maternal tissues for the presence of B[a]P-DNA adducts .\n\nAdducts were detected in maternal , placental and offspring lymphoid tissues at day 19 of gestation , at birth and 1-wk after birth .\n\nThe presence of B[a]P-DNA adducts in immature T cells may , in part , explain the previously observed T cell immunosuppression and tumor susceptibility in mice exposed to B[a]P in utero .\n\nThe effects of DNA lesions on progeny T cells may include interference with normal T-cell development .\n\nThese results provide a possible explanation for the relationship between maternal smoking during pregnancy and childhood carcinogenesis .", "output": "Genomic instability and mutation, Avoiding immune destruction" }, { "input": "Nur77 is an orphan receptor .\n\nAlthough Nur77 affects cell proliferation and apoptosis through its capability of binding to a variety of response elements and regulating their transactivation activities , the intrinsic function of Nur77 is not yet fully understood ; in particular , its regulation of apoptosis and proliferation has been characterized as cell type-dependent and agent context-dependent .\n\nIn this study , Nur77 can be seen to regulate apoptosis via its expression and translocation , rather than its transactivation activity in gastric cancer cells .\n\nNur77 was constitutively expressed in BGC-823 cells .\n\nThe tetradecanoylphorbol-1,3-acetate ( TPA ) treatment not only resulted in up-regulation of the Nur77 mRNA level , but also led to translocation of Nur77 protein from the nucleus to the mitochondria , and caused the release of cytochrome c .\n\nThis TPA-induced translocation of Nur77 was in association with the initiation of apoptosis in gastric cancer cells .\n\nAlthough all-trans retinoic acid ( ATRA ) could not induce apoptosis in BGC-823 cells due to failure of stimulating Nur77 translocation , expression of Nur77 in the nucleus was required for cell growth inhibition by ATRA .\n\nTransfection of antisense Nur77 receptor into BGC-823 cells resulted in resistance of cell growth against ATRA inhibition , and the cells were still arrested in the S phase .\n\nFurthermore , the action of Nur77 in TPA-induced apoptosis was mediated through a protein kinase C signaling pathway , while mitogen-activated protein kinase and phosphatidylinositol 3-kinase signaling pathways were responsible for the regulation of Nur77 mRNA expression .\n\nTaken together , the data revealed the dual functioning mechanisms of Nur77 in gastric cancer cells in response to TPA and ATRA .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Intracystic papillary carcinoma ( IPC ) of the breast is a rare form of noninvasive breast cancer .\n\nAn appreciation of associated pathology with IPC may be critical in surgical decision-making .\n\nMETHODS The medical records of all patients with IPC treated between 1985 and 2001 were retrospectively reviewed .\n\nThree patient groups were identified according to the pathologic features of the primary tumor : IPC alone , IPC with associated ductal carcinoma in situ ( DCIS ) , and IPC with associated invasion with or without DCIS .\n\nTypes of treatment and outcomes were compared between groups .\n\nRESULTS Forty patients were treated for IPC during the study period .\n\nFourteen had pure IPC , 13 had IPC with DCIS , and 13 had IPC with invasion .\n\nThe incidence of recurrence and the likelihood of dying of IPC did not differ between the three groups regardless of the type of surgery ( mastectomy or segmental mastectomy ) performed and whether radiation therapy was administered .\n\nThe disease-specific survival rate was 100% .\n\nCONCLUSIONS When IPC is identified , it is frequently associated with DCIS and or invasion .\n\nStandard therapy should be based on associated pathology .\n\nThe role of radiation therapy in pure IPC remains to be determined .", "output": "Activating invasion and metastasis" }, { "input": "Retinoic acid ( RA ) and its derivatives inhibit the proliferation of normal human mammary epithelial cells ( HMEC ) and some breast carcinoma lines by mechanisms which are not fully understood .\n\nTo identify genes that mediate RA-induced cell growth arrest , an HMEC cDNA library was synthesized and subtractive screening was performed .\n\nWe identified the interleukin-1beta ( IL-1beta ) gene as an RA induced gene in HMEC .\n\nNorthern blot analyses showed that the IL-1beta gene was up-regulated as early as 2 h after RA treatment .\n\nResults from the treatment of HMEC with cycloheximide and actinomycin D indicated that the regulation of the IL-1beta gene by RA occurred at the transcriptional level and that the IL-1beta gene is a direct , downstream target gene of RA .\n\nTo evaluate the effects of IL-1beta on cell proliferation , the proliferation of HMEC was measured in the presence of RA or IL-1beta , or both .\n\nEither RA or IL-1beta could significantly inhibit the proliferation of HMEC .\n\nHowever , the addition of soluble IL-1 receptor antagonist ( sIL-1ra ) to the cell culture medium did not block RA-induced HMEC growth inhibition , whereas sIL-1ra did block the growth inhibition of HMEC by IL-1beta .\n\nIL-1beta expression was not observed in the three carcinoma cell lines , MCF-7 , MDA-MB-231 , and MDA-MB-468 , as compared to the HMEC .\n\nGrowth curves of the breast carcinoma cell lines showed strong inhibitory effects of RA and IL-1beta on the growth of the estrogen receptor ( ER ) positive MCF-7 cell line , but only a small effect on the ER negative MDA-MB-231 cells .\n\nThe expression of the IL-1beta gene was also transcriptionally activated by RA in normal epithelial cells of prostate and oral cavity .\n\nOur results suggest that : ( a ) the IL-1beta gene is a primary target of RA receptors in HMEC ; ( b ) the enhanced expression of the IL-1beta gene does not mediate the RA-induced growth arrest of HMEC ; and ( c ) the expression of the IL-1beta gene is low or absent in all three human breast carcinoma cell lines examined , but the defect in the IL-1beta signaling pathway may be different in ER positive versus ER negative carcinoma cells .", "output": "Sustaining proliferative signaling" }, { "input": "Genetic immunotherapy with tumor antigen gene-modified dendritic cells ( DC ) generates robust immunity , although antitumor protection is not complete in all models .\n\nPrevious experience in a model in which C57BL/6 mice immunized with DC transduced with adenoviral vectors expressing MART-1 demonstrated a 20-40% complete protection to a tumor challenge with B16 melanoma cells .\n\nTumors that did develop in immunized mice had slower growth kinetics compared to tumors implanted in na\ufffdve mice .\n\nIn the present study , we wished to determine if the supraphysiological production of the Th1-skewing cytokine interleukin-12 ( IL-12 ) could enhance immune activation and antitumor protection in this model .\n\nIn a series of experiments immunizing mice with DC cotransduced with MART-1 and IL-12 , antitumor protection and antigen-specific splenocyte cytotoxicity and interferon gamma production inversely correlated with the amount of IL-12 produced by DC .\n\nThis adverse effect of IL-12 could not be explained by a direct cytotoxic effect of natural killer cells directed towards DC , nor the production of nitric oxide leading to down-regulation of the immune response - the two mechanisms previously recognized to explain immune-suppressive effects of IL-12-based vaccine therapy .\n\nIn conclusion , in this animal model , IL-12 production by gene-modified DC leads to a cytokine-induced dose-dependent inhibition of antigen-specific antitumor protection .", "output": "Avoiding immune destruction" }, { "input": "2,3,7,8-tetrachlorodibenzo-p-dioxin ( TCDD ) is the most potent tumor promoter ever tested in rodents .\n\nAlthough it is known that most of TCDD actions are mediated by binding to the aryl hydrocarbon receptor ( AhR ) , the mechanisms leading to tumor promotion still remain to be elucidated .\n\nLoss of contact inhibition is one characteristic hallmark in tumorigenesis .\n\nIn rat liver epithelial WB-F344 cells , TCDD induces a release from contact inhibition , which is manifested by a twofold increase in cell number when TCDD ( 1 nM for 48 h ) is added to confluent cells in the presence of serum , but not when given to exponentially growing or subconfluent , serum-deprived WB-F344 cells .\n\nLoss of G1 arrest was also shown by flow cytometric analysis .\n\nWe demonstrate that TCDD treatment significantly increases cyclin D2 and cyclin A protein levels and show by immunofluorescence that these proteins accumulate in the nucleus .\n\nAlthough TCDD treatment leads to a strong increase in cyclin D2/cdk4 and cyclin A/cdk2 complex formation , we could only detect an elevation of cyclin A/cdk2 activity .\n\nIn accordance with a lack of elevated cdk4 activity , no decrease in the amount of hypophosphorylated retinoblastoma protein could be shown after TCDD treatment .\n\nThe importance of increased cyclin A/cdk2 activity for TCDD-dependent release from contact inhibition was shown by the fact that the cdk2/cdc2-specific inhibitor olomoucine ( 25 microM ) abolished TCDD response .\n\nThese data indicate cyclin A-dependent loss of G1 arrest after TCDD treatment mainly downstream of the retinoblastoma protein .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "A persistent immune response to hepatitis viruses is a well-recognized risk factor for hepatocellular carcinoma .\n\nHowever , the molecular and cellular basis for the procarcinogenic potential of the immune response is not well defined .\n\nHere , using a unique animal model of chronic hepatitis that induces hepatocellular carcinogenesis , we demonstrate that neutralization of the activity of Fas ligand prevented hepatocyte apoptosis , proliferation , liver inflammation , and the eventual development of hepatocellular carcinoma .\n\nThe results indicate that Fas ligand is involved not only in direct hepatocyte killing but also in the process of inflammation and hepatocellular carcinogenesis in chronic hepatitis .\n\nThis is the first demonstration that amelioration of chronic inflammation by some treatment actually caused reduction of cancer development .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Retinoblastoma ( Rb ) is the most common intraocular tumor of childhood .\n\nIn this study we examined primary Rb specimens and Rb cell lines for the expression of immunoglobulin superfamily ( IgSF ) antigens : MHC class I and II ( MHC-I and MHC-II ) , neural cell adhesion molecule ( NCAM ) , intercellular adhesion molecule-1 ( ICAM-1 ) , and Thy-1 , which play an important role in immune system and tumor cell interactions .\n\nMHC-I and-II , ICAM-1 ( CD54 ) , NCAM ( CD56 ) , and Thy-1 ( CDw90 ) immunoreactivity was studied in eight primary Rb biopsy specimens using immunohistochemistry , three using immunoelectron microscopy , and six Rb cell lines using flow cytometry ( FCM ) .\n\nParenchymal and vascular-associated cells , phenotypically similar to retinal microglia , strongly expressed MHC-II immunoreactivity and were distributed throughout primary Rb specimens .\n\nHowever , MHC-II expression on Rb cell lines was similar to nonspecific control levels .\n\nTumor cells in primary Rb specimens displayed high NCAM , moderate Thy-1 , and low MHC-I and ICAM-1 immunolabeling .\n\nTumor vasculature expressed low to moderate MHC-I and ICAM-1 immunoreactivity and moderate Thy-1 immunoreactivity .\n\nNCAM was not detected on the vasculature of primary Rb specimens .\n\nRb cell lines displayed variable expression of Thy-1 , ICAM-1 , and MHC-I .\n\nNCAM was highly expressed on five of six Rb cell lines .\n\nThe high levels of constitutive NCAM immunoreactivity on Rb tumor cells confirm the neuroectodermal origins of this tumor .\n\nAdditionally , the variable expression of Thy-1 may suggest separate neural lineages or differences in the maturational status ofsome Rb tumors .\n\nThe presence of a population of infiltrating MHC-II-positive cells in primary Rb tumors has implications for immunomodulation of Rb growth .", "output": "Avoiding immune destruction" }, { "input": "We have previously reported that stressed apoptotic tumor cells are more immunogenic in vivo than nonstressed ones .\n\nUsing confocal microscopy we have confirmed our previous observation that heat-stressed apoptotic 12B1-D1 leukemia cells ( BCR-ABL(+) ) express HSP60 and HSP72 on their surface .\n\nTo explore how the immune system distinguishes stressed from nonstressed apoptotic tumor cells , we analyzed the responses of dendritic cells to these 2 types of apoptotic cells .\n\nWe found that nonstressed and heat-stressed apoptotic 12B1-D1 cells were taken up by dendritic cells in a comparable fashion .\n\nHowever , when stressed apoptotic 12B1-D1 cells were coincubated with immature dendritic cells for 24 hours , this resulted in greater up-regulation of costimulatory molecules ( CD40 , CD80 , and CD86 ) on the surface of dendritic cells .\n\nMoreover , stressed apoptotic 12B1-D1 cells were more effective in stimulating dendritic cells to secrete interleukin-12 ( IL-12 ) and in enhancing their immunostimulatory functions in mixed leukocyte reactions .\n\nFurthermore , we demonstrated that immunization of mice with stressed apoptotic 12B1-D1 cells induced the secretion of T helper-1 ( T(H)1 ) profile of cytokines by spleen cells .\n\nSplenocytes from mice immunized with stressed apoptotic cells , but not nonstressed ones , were capable of lysing 12B1-D1 and the parental 12B1 line , but not a B-cell leukemia line , A20 .\n\nOur data indicate that stressed apoptotic tumor cells are capable of providing the necessary danger signals , likely through increased surface expression of heat shock proteins ( HSPs ) , resulting in activation/maturation of dendritic cells and , ultimately , the generation of potent antitumor T-cell responses .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "Binding of erythropoietin ( EPO ) to its receptor ( EPOR ) on erythroid cells induces the activation of numerous signal transduction pathways , including the mitogen-activated protein kinase Jun-N-terminal kinase ( JNK ) .\n\nIn an effort to understand the regulation of EPO-induced proliferation and JNK activation , we have examined the role of potential autocrine factors in the proliferation of the murine erythroleukemia cell line HCD57 .\n\nWe report here that treatment of these cells with EPO induced the expression and secretion of tumor necrosis factor alpha ( TNF-alpha ) .\n\nEPO-dependent proliferation was reduced by the addition of neutralizing antibodies to TNF-alpha , and exogenously added TNF-alpha induced proliferation of HCD57 cells .\n\nEPO also could induce TNF-alpha expression in BAF3 and DA3 myeloid cells ectopically expressing EPOR .\n\nAddition of TNF-alpha activated JNK in HCD57 cells , and the activity of JNK was partially inhibited by addition of a TNF-alpha neutralizing antibody .\n\nPrimary human and murine erythroid progenitors expressed TNF-alpha in either an EPO-dependent or constitutive manner .\n\nHowever , TNF-alpha had an inhibitory effect on both immature primary human and murine cells , suggestive that the proliferative effects of TNF-alpha may be limited to erythroleukemic cells .\n\nThis study suggests a novel role for autocrine TNF-alpha expression in the proliferation of erythroleukemia cells that is distinct from the effect of TNF-alpha in normal erythropoiesis .", "output": "Sustaining proliferative signaling" }, { "input": "Prognosis of patients with non small cell lung cancer ( NSCLC ) remains difficult to assess , even after adjustment for pathological stage .\n\nPrognostic value of numerous biological markers has been evaluated , with conflicting results .\n\nData of 86 patients with NSCLC treated by surgery were collected with clinical characteristics , histopathological data including tumor differentiation and status of blood and lymphatic vessel invasion and evaluation by immunohistochemistry of Rb , Bcl-2 and Ki-67 expression .\n\nPrognostic values for overall survival ( OS ) and event-free survival ( EFS ) were analyzed by the log tank test and the multivariable Cox model .\n\nUsing univariable analyses , pT , pN , poor differentiation or large cell subtype were associated with a poor OS , while lymphatic and/or blood vessel invasion were associated with a short EFS .\n\nNone of the molecular markers had a significant prognostic value for either outcome .\n\nIn multivariable analyses , only stage remained of prognostic value for OS .\n\nInterestingly , the presence of blood vascular invasion in the tumor was significantly predictive for subsequent metastatic occurrence in stages I and II .\n\nThis feature might , therefore , be relevant for administration of adjuvant therapy in completely resected NSCLC .", "output": "Activating invasion and metastasis" }, { "input": "Hormone replacement therapy , which is a common menopausal treatment , is contraindicated in women with breast cancers due to concerns regarding the potential for breast cell proliferation .\n\nAs such , there is a need for alternative methods for treating menopausal symptoms .\n\nTo determine the influence of one such alternative , black cohosh ( Cimicifuga racemosa [ CR] ) , on estrogen-dependent mammary cancers , we conducted an in vitro investigation of the effect of an isopropanolic CR-extract on the proliferation of estrogen receptor-positive breast cancer cells .\n\nThe experiments were performed using the human breast adenocarcinoma ( MCF-7 ) cell test system , an established in vitro model for estrogen-dependent tumors .\n\nThe influence of CR-extract on the proliferation of the MCF-7 cells was determined by measuring the incorporation of radioactively labeled thymidine .\n\nUnder estrogen-deprived conditions , the CR-extract ( 10(-3)-10(-5) dilutions ) significantly inhibited MCF-7 cell proliferation .\n\nAdditionally , application of the CR-extract inhibited estrogen-induced proliferation of MCF-7 cells .\n\nMoreover , the proliferation-inhibiting effect of tamoxifen was enhanced by the CR-extract .\n\nSuch data that suggest a non-estrogenic , or estrogen-antagonistic effect of CR on human breast cancer cells lead to the conclusion that CR treatment may be a safe , natural remedy for menopausal symptoms in breast cancer .", "output": "Sustaining proliferative signaling" }, { "input": "INTRODUCTION AND AIMS The survival of pancreatic cancer patients with portal vein resection is extremely poor due to the high incidence of liver metastasis .\n\nThe occurrence of liver metastasis is decreased by locoregional arterial infusion after pancreatic surgery .\n\nChemosensitivity tests can provide the basis for individualized chemotherapy in each patient and predict the clinical response .\n\nTherefore , the current study was designed to clarify whether locoregional chemotherapy based on the results of chemosensitivity tests has the clinical effects of preventing liver metastasis and improving survival for patients with portal vein resection .\n\nMETHODOLOGY The resected specimens from 40 of 47 patients with resection of pancreatic cancer were assessed for chemosensitivity to various anticancer drugs .\n\nFourteen patients underwent portal vein resection due to direct invasion , and nine of these patients received intra-arterial adjuvant chemotherapy on the basis of the results of MTT assay to prevent liver metastasis .\n\nThe remaining five patients received no chemotherapy .\n\nRESULTS None of the patients who received intra-arterial chemotherapy had liver metastasis , and this group of patients had improved survival .\n\nThe mean survival of patients with intra-arterial chemotherapy was significantly longer than that of patients without chemotherapy ( 25.6 months with chemotherapy versus 9.4 months without chemotherapy ) .\n\nCONCLUSION A pilot study of postoperative intra-arterial chemotherapy showed the reduction of liver metastasis and improvement of survival among pancreatic cancer patients with portal vein resection .", "output": "Activating invasion and metastasis" }, { "input": "beta-Defensins are small antimicrobial peptides of the innate immune system produced in response to microbial infection of mucosal tissue and skin .\n\nWe demonstrate that murine beta-defensin 2 ( mDF2beta ) acts directly on immature dendritic cells as an endogenous ligand for Toll-like receptor 4 ( TLR-4 ) , inducing up-regulation of costimulatory molecules and dendritic cell maturation .\n\nThese events , in turn , trigger robust , type 1 polarized adaptive immune responses in vivo , suggesting that mDF2beta may play an important role in immunosurveillance against pathogens and , possibly , self antigens or tumor antigens .", "output": "Avoiding immune destruction" }, { "input": "Tumor cells are elusive targets for immunotherapy due to their heterogeneity and genetic instability .\n\nHere we describe a novel , oral DNA vaccine that targets stable , proliferating endothelial cells in the tumor vasculature rather than tumor cells .\n\nTargeting occurs through upregulated vascular-endothelial growth factor receptor 2 ( FLK-1 ) of proliferating endothelial cells in the tumor vasculature .\n\nThis vaccine effectively protected mice from lethal challenges with melanoma , colon carcinoma and lung carcinoma cells and reduced growth of established metastases in a therapeutic setting .\n\nCTL-mediated killing of endothelial cells indicated breaking of peripheral immune tolerance against this self antigen , resulting in markedly reduced dissemination of spontaneous and experimental pulmonary metastases .\n\nAngiogenesis in the tumor vasculature was suppressed without impairment of fertility , neuromuscular performance or hematopoiesis , albeit with a slight delay in wound healing .\n\nOur strategy circumvents problems in targeting of genetically unstable tumor cells .\n\nThis approach may provide a new strategy for the rational design of cancer therapies .", "output": "Inducing angiogenesis, Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "BACKGROUND A retrospective study was performed to determine the indications for positron emission tomography ( PET ) using [ (18)F]fluorodeoxyglucose ( FDG ) in patients with esophageal cancer , including those with early cancer , and to investigate whether the tumor-to-normal ratio ( T/N ratio ) could be used as a substitute for the standardized uptake value ( SUV ) .\n\nMETHODS Thirty-six patients were included in the study .\n\nThirty-one patients who had 36 biopsy-proven lesions ( 35 squamous cell carcinomas and one small cell carcinoma ) underwent PET study prior to treatment .\n\nPET images were evaluated visually and the relationship between the depth of invasion and the PET findings were examined in 22 lesions of 19 patients from whom specimens were obtained from the primary tumor by surgery or endoscopic mucosal resection .\n\nPET results were also compared with computed tomography ( CT ) and endoscopic ultrasonography ( EUS ) for detection of regional lymph node metastases in 18 patients who underwent extended lymph node dissection .\n\nFive patients underwent PET studies for the detection of recurrence and the PET findings were compared with their CT findings .\n\nThe T/N ratio and the SUV were calculated for 20 primary tumors .\n\nRESULTS Among the 15 tumors that were pT1b or greater , all 15 were positive on PET and all seven of the lesions confined to the mucosa ( Tis or T1a ) were negative .\n\nThe sensitivity , specificity and accuracy of detecting nodal involvement were , respectively , 37.5 , 96.1 and 88.3% by CT , 30.8 , 88.5 and 81.0% by EUS and 41.7 , 100 and 92.2% by PET .\n\nMore sites of recurrence were detected by PET than by CT .\n\nThere was no statistically significant correlation between the SUV and the T/N ratio .\n\nCONCLUSIONS PET imaging can detect primary esophageal cancer with a depth of invasion of T1b or greater , but Tis and T1a tumors are undetectable .\n\nPET seems to be more accurate than CT or EUS for diagnosing lymph node metastasis .\n\nThe T/N ratio cannot be used as a substitute for the SUV .", "output": "Activating invasion and metastasis" }, { "input": "The INK4a/ARF tumor suppressor locus is implicated in the senescence-like growth arrest provoked by oncogenic Ras in primary cells .\n\nINK4a and ARF are distinct proteins encoded by transcripts in which a shared exon is decoded in alternative reading frames .\n\nHere we analyze dermal fibroblasts ( designated Q34 ) from an individual carrying independent missense mutations in each copy of the common exon .\n\nBoth mutations alter the amino acid sequence of INK4a and functionally impair the protein , although they do so to different degrees .\n\nOnly one of the mutations affects the sequence of ARF , causing an apparently innocuous change near its carboxy terminus .\n\nUnlike normal human fibroblasts , Q34 cells are not permanently arrested by Ras or its downstream effectors Ets1 and Ets2 .\n\nMoreover , ectopic Ras enables the cells to grow as anchorage-independent colonies , and in relatively young Q34 cells anchorage independence can be achieved without addition of telomerase or perturbation of the p53 pathway .\n\nWhereas ARF plays the principal role in Ras-induced arrest of mouse fibroblasts , our data imply that INK4a assumes this role in human fibroblasts .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE To investigate treatment of human pancreatic cancer cell lines and xenografts with combinations of Erbitux ( IMC-C225 ) anti-epidermal growth factor receptor ( EGFR ) antibody , gemcitabine , and radiation .\n\nMETHODS AND MATERIALS BxPC-3 and MiaPaCa-2 human pancreatic carcinoma cells were treated in vitro for 24 h with IMC-C225 ( 5 microg/mL ) , then exposed to epidermal growth factor ( EGF ) ( 10 mM ) for 5 min .\n\nImmunoblots were screened for EGFR expression and the ability of IMC-C225 to block EGF-induced tyrosine phosphorylation of EGFR .\n\nCells were treated with IMC-C225 ( 5 microg/mL ) on Day 0 , the IC(50) dose of gemcitabine on Day 1 for 24 h , followed by 3 Gy 60Co irradiation on Day 2 , or the combination of each agent .\n\nFor cell proliferation , cells were counted on Day 4 , and for apoptosis , cells were stained with annexin V-FITC and propidium iodide , then analyzed by FACS .\n\nCells were treated with the same single or multiple treatments and analyzed in a clonogenic cell survival assay .\n\nThe effect of IMC-C225 , gemcitabine , and radiation on the growth of BxPC-3 and MiaPaCa-2 tumor xenografts was determined .\n\nAthymic nude mice bearing established s.c. tumor xenografts of 6-8 mm diameter received 6 weeks of treatment with IMC-C225 ( 1 mg every 3 days x 6 ) alone or in combination with gemcitabine ( 120 mg/kg i.v. every 6 days x 6 ) , and 6 weekly fractions of 3 Gy radiation on the days after gemcitabine administration .\n\nTumor growth was measured with Vernier calipers .\n\nRESULTS BxPC-3 and MiaPaCa-2 cell lines expressed low levels of EGFR .\n\nIMC-C225 inhibited EGF-induced tyrosine phosphorylation of the EGF receptor on both cell lines .\n\nTreatment of cells with a combination of IMC-C225 + gemcitabine + radiation produced the highest induction of apoptosis and inhibition of proliferation in vitro .\n\nCombination treatment with IMC-C225 , gemcitabine , and radiation produced 100% complete regression of MiaPaCa-2 tumors for more than 250 days , and the greatest growth inhibition of BxPC-3 tumors compared to any single or dual treatments .\n\nCONCLUSIONS The IMC-C225 therapy in combination with gemcitabine chemotherapy and radiation therapy demonstrated statistically significantly greater efficacy over the single and double combination therapies .\n\nThis form of multimodality treatment shows potential clinical application in the treatment of pancreatic cancer in humans .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "During papillomavirus infection , the E5 protein localizes in the cell Golgi apparatus and other endomembrane compartments .\n\nCells transformed by E5 do not express major histocompatibility class I complex ( MHC I ) on the cell surface , while cells transformed by the other transforming proteins E6 and E7 do .\n\nIn addition , the total amount of both MHC I protein and mRNA is reduced in E5-transformed cells .\n\nHere we show that expression of bovine papillomavirus E5 causes the retention of MHC I in the Golgi apparatus , thus preventing its transport to the cell surface .\n\nWe ascribe this effect to a failure of acidification of the Golgi apparatus , as similar effects are observed in control cells treated with the ionophore monensin .\n\nTreatment of E5-transformed cells with either beta- or gamma-interferon increases the synthesis of MHC I , showing that inhibition of MHC I expression by E5 is not irreversible .\n\nHowever , even after interferon treatment , MHC I , although increased in quantity , is not transported to the cell surface .\n\nE5 therefore affects MHC I at several levels , but prevention of MHC I transport to the cell surface appears to be the dominant effect .\n\nLack of surface MHC I would have profound consequences for presentation of viral peptides to the immune system .", "output": "Avoiding immune destruction" }, { "input": "Using standard culture conditions , primary human mammary epithelial cells ( HMECs ) undergo a premature , transient growth arrest termed M0 ( mortality stage 0 ) after 10-15 population doublings in vitro .\n\nIt has been reported that emergence from this growth arrest by the abrogation of p16(INK4a) , a cyclin-dependent kinase inhibitor , and expression of the catalytic component of human telomerase ( hTERT ) are necessary for HMEC immortalization .\n\nHere we show that primary HMECs , grown on feeder layers , do not undergo this growth arrest and can be immortalized without abrogating p16 .\n\nThese findings support the concept that the so-called M0 stage represents a cell culture stress-induced growth arrest and that hTERT is sufficient to immortalize HMECs when cultured under adequate conditions .", "output": "Enabling replicative immortality" }, { "input": "Optimal lymphocyte activation requires the simultaneous engagement of stimulatory and costimulatory receptors .\n\nStimulatory immunoreceptors are usually composed of a ligand-binding transmembrane protein and noncovalently associated signal-transducing subunits .\n\nHere , we report that alternative splicing leads to two distinct NKG2D polypeptides that associate differentially with the DAP10 and KARAP ( also known as DAP12 ) signaling subunits .\n\nWe found that differential expression of these isoforms and of signaling proteins determined whether NKG2D functioned as a costimulatory receptor in the adaptive immune system ( CD8+ T cells ) or as both a primary recognition structure and a costimulatory receptor in the innate immune system ( natural killer cells and macrophages ) .\n\nThis strategy suggests a rationale for the multisubunit structure of stimulatory immunoreceptors .", "output": "Avoiding immune destruction" }, { "input": "PURPOSE Osteosarcoma is an aggressive primary bone cancer characterized by expression of platelet-derived growth factor ( PDGF ) and its cognate receptor .\n\nCoexpression of the growth factor and receptor suggests their role in autocrine or paracrine growth mechanisms .\n\nIt has been reported previously that STI571 has specific activity in inhibiting select tyrosine kinase receptors , including PDGF and c-Kit .\n\nOsteosarcomas express low levels of c-Kit but abundant levels of PDGF receptor ( PDGFR ) .\n\nEXPERIMENTAL DESIGN To investigate the potential of STI571 as therapy for osteosarcoma , we studied its effects on PDGF-mediated cell growth in vitro and in an in vivo mouse model .\n\nRESULTS PDGF acted as a potent mitogen in a dose-dependent manner in two osteosarcoma cell lines .\n\nSTI571 ( 1.0 micro M ) inhibited both PDGFR-alpha and PDGFR-beta phosphorylation and the downstream phosphorylation targets extracellular signal-regulated kinase and Akt .\n\nSTI571 also inhibited PDGF-mediated growth and induced apoptosis in vitro as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and terminal deoxynucleotidyl transferase-mediated nick end labeling staining .\n\nTo study the effect of STI571 alone or in combination with Taxol in an in vivo model , an osteosarcoma cell line ( KRIB ) was transplanted into the tibia of athymic nude mice .\n\nMice were treated with STI571 ( 50 mg/kg p.o. q M-F ) , Taxol ( 8 mg/kg i.p. weekly ) , or STI571 plus Taxol for 6 weeks .\n\nThere was no significant difference in tumor size between treatment and control mice .\n\nAberrant signaling pathways downstream of the PDGFR in the v-Ki-ras oncogene-transformed KRIB cell line may in part explain this finding .\n\nCONCLUSIONS Our data demonstrate that STI571 inhibits PDGF-mediated growth and leads to apoptosis of osteosarcoma cells in vitro by selective inhibition of the PDGFR tyrosine kinase .\n\nThe effectiveness of STI571 in our studies suggests targeting of PDGFRs as a novel treatment for osteosarcoma .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Onconase ( Onc ) is an amphibian ribonuclease of the pancreatic RNase family that is cytostatic and cytotoxic to several tumor lines .\n\nIt also shows anti-tumor activity in mouse tumor models and is currently in phase III clinical trials .\n\nIn animal tests and clinical trials Onc shows lesser toxicity and fewer side effects compared to most chemotherapeutic drugs .\n\nIntriguingly , repeated infusions of this protein do not cause apparent immunological reactions in patients .\n\nThe aim of the present study was to investigate sensitivity to Onc of human lymphocytes during their mitogenic stimulation in response to the polyvalent mitogen phytohemagglutinin ( PHA ) , and in mixed allogeneic lymphocyte cultures .\n\nUnexpectedly , we observed that frequency of cells undergoing activation-induced apoptosis was markedly increased in all cultures containing Onc .\n\nApoptosis was measured by flow cytometry using markers that detect activation of caspases , the in situ presence of DNA strand breaks , and loss of fragmented DNA ( 'sub-G1 ' cell subpopulation ) .\n\nThe enhancement of frequency of activation-induced apoptosis ( up to 244% ) was observed at 4.2-83 nM Onc concentration , which is at least an order magnitude lower than its minimal concentration reported to affect proliferation or induce apoptosis of leukemic and solid tumor cell lines .\n\nThe cell cycle progression of lymphocytes that responded to PHA mitogenically was not affected at 8.3 or 83 nM Onc concentration .\n\nBecause activation-induced apoptosis is the key mechanism regulating several in vivo immunological functions including induction of tolerance , the observed effects of Onc may explain the apparent lack of immune reactions to this protein in treated patients .\n\nThe propensity of Onc to potentiate the activation-induced apoptosis suggests that this drug may have clinical utility as immunomodulating agent , e.g. , to suppress transplant rejection or treat autoimmune diseases .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "We studied whether feeding pregnant female rats different high fat diets affects structural zones in the spleen and lymph nodes , involved in production of T and B cells , as well as cell kinetics and apoptosis in some offspring with mammary glands tumors .\n\nRat mothers were fed either a 7% or 15% corn-oil or a 7% or 15% olive-oil diet .\n\nAt four weeks of age , female offspring ( n=10-15 per group ) were transferred to 7% corn oil diet .\n\nFive-week old offspring were exposed twice to the carcinogen , dimethylbenz(a)antracene ( 10 mg/rat/week ) .\n\nThree months later , tumors were counted and sized , and samples from the spleen , axillary lymph nodes and tumors collected for immunohistochemical analyses .\n\nFeeding the mothers with both the 7% and 15% olive-oil diets significantly increased the number of tumor-free rats in offspring .\n\nTumors were characterized with active mitosis , intensive lymphoid infiltration inside a knot and high rates of apoptosis , particularly in tumors obtained from rats whose mothers were fed the 15% olive-oil diet .\n\nIn the spleen , the 15% olive-oil diet significantly increased the areas of the follicles and germinal centers but only in tumor-free rats .\n\nIn tumor-bearing rats , areas of germinal centers increased compared to the 7% olive-oil diet .\n\nThe 15% olive-oil diet increased all areas of the lymph nodes in tumor-free rats , while in tumor-bearing rats , this diet increased the areas of the cortex and mantle layer .\n\nWe conclude that exposure to various diets in utero and during lactation affects the immune system .\n\nIn addition , the promotion of apoptosis may play a key role in the mechanisms involved in the transplacental effects on mammary tumor development as seen using a 15% olive-oil diet , similar to the high fat diets of Mediterranean countries .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "Annexin II is secreted into the extracellular environment , where , via interactions with specific proteases and extracellular matrix proteins , it participates in plasminogen activation , cell adhesion , and tumor metastasis and invasion .\n\nHowever , mechanisms regulating annexin II transport across the cellular membrane are unknown .\n\nIn this study , we used coimmunoprecipitation to show that Annexin-II was bound to insulin and insulin-like growth factor-1 ( IGF-1 ) receptors in PC12 cells and NIH-3T3 cells overexpressing insulin ( NIH-3T3(IR) ) or IGF-1 receptor ( NIH-3T3(IGF-1R) ) .\n\nStimulation of insulin and IGF-1 receptors by insulin caused a temporary dissociation of annexin II from these receptors , which was accompanied by an increased amount of extracellular annexin II detected in the media of PC12 , NIH-3T3(IR) , and NIH-3T3(IGF-1R) cells but not in that of untransfected NIH-3T3 cells .\n\nActivation of a different growth factor receptor , the platelet-derived growth factor receptor , did not produce such results .\n\nTyrphostin AG1024 , a tyrosine kinase inhibitor of insulin and IGF-1 receptor , was shown to inhibit annexin II secretion along with reduced receptor phosphorylation .\n\nInhibitors of a few downstream signaling enzymes including phosphatidylinositol 3-kinase , pp60c-Src , and protein kinase C had no effect on insulin-induced annexin II secretion , suggesting a possible direct link between receptor activation and annexin II secretion .\n\nImmunocytochemistry revealed that insulin also induced transport of the membrane-bound form of annexin II to the outside layer of the cell membrane and appeared to promote cell aggregation .\n\nThese results suggest that the insulin receptor and its signaling pathways may participate in molecular mechanisms mediating annexin II secretion .", "output": "Sustaining proliferative signaling" }, { "input": "Dietary exposure to aflatoxins is one of the major risk factors for hepatocellular carcinoma .\n\nIndividual susceptibility to aflatoxin-induced hepatocarcinogenesis may be modulated by both genetic and environmental factors affecting metabolism .\n\nA cross-sectional study was performed to evaluate determinants of the formation of aflatoxin covalently bound to albumin ( AFB1-albumin adducts ) .\n\nA total of 474 subjects who were free of liver cancer and cirrhosis and were initially selected as controls for previous case-control studies of aflatoxin-induced hepatocarcinogenesis in Taiwan , were employed in this study .\n\nAflatoxin-albumin adducts were determined by competitive enzyme-linked immunosorbent assay , hepatitis B surface antigen and antibodies to hepatitis C virus by enzyme immunoassay , as well as genotypes of glutathione S-transferase M1-1 and T1-1 by polymerase chain reaction .\n\nThe detection rate of AFB1-albumin adducts was significantly higher in males ( 42.5% ) than in females ( 21.6% ) ( multivariate-adjusted odds ratio=2.6 , 95% confidence interval=1.4-5.0 ) .\n\nThe formation of detectable albumin adducts was moderately higher in hepatitis B surface antigen carriers ( 42.8% ) than in non-carriers ( 36.6% ) ( multivariate-adjusted odds ratio=1.4 , 95% confidence interval=1.0-2.1 ) .\n\nIn addition , the detection rate of AFB1-albumin adducts tended to increase with the increasing number of null genotypes of glutathione S-transferase M1-1 and glutathione S-transferase T1-1 .\n\nIn conclusion , this cross-sectional study has assessed the relative contributions of environmental exposure and host susceptibility factors in the formation of AFB1-albumin adducts in a well characterised Chinese adult population .\n\nThis study further emphasises the necessity to reduce aflatoxin exposure in people living in an area endemic for chronic hepatitis B virus infection .", "output": "Genomic instability and mutation" }, { "input": "A large number of human cancers display alterations in the Ink4a/cyclin D/Cdk4 genetic pathway , suggesting that activation of Cdk4 plays an important role in oncogenesis .\n\nHere we report that Cdk4-null mouse embryonic fibroblasts are resistant to transformation in response to Ras activation with dominant-negative ( DN ) p53 expression or in the Ink4a/Arf-null background , judged by foci formation , anchorage-independent growth , and tumorigenesis in athymic mice .\n\nCdk4-null fibroblasts proliferate at normal rates during early passages .\n\nWhereas Cdk4(+/+)Ink4a/Arf(-/-) cells are immortal in culture , Cdk4(-/-)Ink4a/Arf(-/-) cells undergo senescence during continuous culture , as do wild-type cells .\n\nActivated Ras also induces premature senescence in Cdk4(-/-)Ink4a/Arf(-/-) cells and Cdk4(-/-) cells with DNp53 expression .\n\nThus , Cdk4 deficiency causes senescence in a unique Arf/p53-independent manner , which accounts for the loss of transformation potential .\n\nCdk4-null cells express high levels of p21(Cip1/Waf1) with increased protein stability .\n\nSuppression of p21(Cip1/Waf1) by small interfering RNA ( siRNA ) , as well as expression of HPV-E7 oncoprotein , restores immortalization and Ras-mediated transformation in Cdk4(-/-)Ink4a/Arf(-/-) cells and Cdk4(-/-) cells with DNp53 expression .\n\nTherefore , Cdk4 is essential for immortalization , and suppression of Cdk4 could be a prospective strategy to recruit cells with inactive Arf/p53 pathway to senescence .", "output": "Enabling replicative immortality" }, { "input": "Alterations of the epidermal growth factor receptor ( EGFR ) gene are common in some forms of cancer and the most frequent is a deletion of exons 2-7 .\n\nWe have previously shown that this mutant receptor , called DeltaEGFR , confers enhanced tumorigenicity to glioblastoma cells through elevated proliferation and reduced apoptotic rates of the tumor cells in vivo .\n\nTo understand the molecular mechanisms that underlie DeltaEGFR-enhanced proliferation , we examined the gene products that control cell cycle progression .\n\nWe found that levels of the cyclin-dependent kinase ( CDK ) inhibitor , p27 , were lower in U87MG.DeltaEGFR tumors than in parental U87MG or control U87MG.DK tumors .\n\nConsequently , CDK2-cyclin A activity was also elevated , concomitant with the RB protein hyperphosphorylation .\n\nIn addition , activated phosphatidylinositol 3-kinase ( PI3-K ) and phosphorylated Akt levels were also elevated in the U87MG.DeltaEGFR tumors .\n\nU87MG.DeltaEGFR cells failed to arrest in G(1) in response to serum starvation in vitro and while maintaining high levels of PI3-K activity and hyperphosphorylated RB .\n\nTreatment of U87MG.DeltaEGFR cells with LY294002 , a PI3-K inhibitor , caused reduced levels of phosphorylated Akt and concomitantly up-regulated levels of p27 .\n\nExpression of a kinase dead dominant-negative Akt mutant in the U87MG.DeltaEGFR cells similarly resulted in up-regulation of p27 and down-regulation of tumorigenicity in vivo .\n\nThese results suggest that the constitutively active DeltaEGFR can enhance cell proliferation in part by down-regulation of p27 through activation of the PI3-K/Akt pathway .\n\nThis pathway may represent another therapeutic target for treatment of those aggressive glioblastomas expressing DeltaEGFR .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Whole body hyperthermia ( WBH ) has been used as an adjunct to radio-/chemotherapy in patients with various malignant diseases .\n\nAlthough clear evidence is still missing , it has been hypothesized that an activation of the immune system might contribute to the therapeutic effect of WBH .\n\nTo examine whether a treatment with 60-minute 41.8 degrees C WBH as an adjunct to chemotherapy ( WBH-CT ) induces an activation of T cells , blood samples were collected at numerous time points before and up to 48 h post-treatment .\n\nThe aim of this study was to examine the effect of WBH-CT on the expression of a broad range of activation markers on peripheral blood lymphocytes ( PBL ) , on serum cytokines and intracellular cytokine levels in T cells , and the capacity of these cells to proliferate .\n\nImmediately after 41.8 degrees C WBH-CT treatment , a drastic increase in peripheral natural killer ( NK ) cells ( P<0.05 ) and CD56+ cytotoxic T lymphocytes ( CTL ; P<0.01 ) in the patients ' peripheral blood was observed .\n\nAt 5 h post-treatment , the percentages of both effector cell types had returned to baseline levels .\n\nThis transient phenomenon was accompanied by a short period of reduced T cell activity , indicated by diminished serum levels of soluble interleukin-2 receptors ( sIL-2R ) at 3 h post-WBH-CT ( P<0.05 ) and decreased lymphocytic proliferation at the same point in time .\n\nThis first phase was followed by a marked but short-lived increase in the patients ' serum levels of interleukin-6 ( IL-6 ; P<0.01 ) during the first 5 h following treatment , with a subsequent decrease to baseline levels at 24 h and significantly increased serum levels of tumor necrosis factor-alpha ( TNF-alpha ) at 0 h ( P<0.01 ) , 3 h ( P<0.05 ) , 5 h ( P<0.05 ) and 24 h ( P<0.01 ) post-WBH-CT .\n\nThe third phase of the immunological consequences of WBH-CT consisted of an increase in the percentage of peripheral cytotoxic T lymphocytes ( CTL ) expressing CD56 , reaching a maximum at 48 h post-WBH ( P<0.01 ) .\n\nFurthermore , the percentage of CD4+ T cells expressing the T cell activation marker CD69 increased nearly two-fold over time , reaching its maximum at 48 h ( P<0.05 ) .\n\nAs an additional marker for T cell activation , serum levels of sIL-2R increased markedly ( P<0.01 ) , reaching maximum levels at the same point in time .\n\nElevated intracellular concentrations of interferon-gamma ( IFN-gamma ) and/or TNF-alpha in CD8+ T cells were found in 4 out of 5 patients at 24 h post-WBH-CT .\n\nSince similar changes were not observed in patients receiving chemotherapy alone , this is the first study to provide evidence for prolonged WBH-CT-induced activation of human T cells .", "output": "Avoiding immune destruction" }, { "input": "AIM To investigate the progressive transformation of immortal cells of human fetal esophageal epithelium induced by human papillomavirus , and to examine biological criteria of sequential passage of cells , including cellular phenotype , proliferative rate , telomerase , chromosome and tumorigenicity .\n\nMETHODS The SHEE cell series consisted of immortalized embryonic esophageal epithelium which was in malignant transformation when cultivated over sixty passages without co-carcinogens .\n\nCells of the 10th , 31st , 60th and 85th passages were present in progressive development after being transfected with HPV .\n\nCells were cultivated in a culture flask and 24-hole cultural plates .\n\nProgressive changes of morphology , cell growth , contact-inhibition , and anchorage-dependent growth characteristics were examined by phase contrast microscopy .\n\nThe cell proliferation rate was assayed by flow cytometry .\n\nThe modal number of chromosomes was analyzed .\n\nHPV18E(6)E(7) was detected by Western blot methods and activities of telomerase were analyzed by TRAP .\n\nTumorigenicity of cells was detected with soft agar plates cultivated and with tumor formation in SCID mice .\n\nRESULTS In morphological examination the 10th passage cells were in good differentiation , the 60th and 85th passages cells were in relatively poor differentiation , and the 31st passage cells had two distinct differentiations .\n\nThe characteristics of the 85th and 60th passage cells were weakened at contact-inhibition and anchorage-dependent growth .\n\nKaryotypes of four stages of cells belonged to hyperdiploid or hypotriploid , and bimodal distribution of chromosomes appeared in the 31st and 60th passage cells .\n\nAll of these characteristics combined with a increasing trend .\n\nThe activities of telomerase were expressed in the latter three passages .\n\nFour fourths of SCID mice in the 85th passage cells and one fourth of SCID mice in the 60th passage cells developed tumors , but the cells in the 10th and 31st passage displayed no tumor formation .\n\nCONCLUSION In continual cultivation of fetal esophageal epithelial cells with transduction of HPV18E(6)E(7) , cells from the 10th to the 85th passage were changed gradually from preimmortal , immortal , precancerous to malignantly transformed stages .\n\nAll of these changes were in a dynamic progressive process .\n\nThe establishment of a continuous line of esophageal epithelium may provide a in vitro model of carcinogenesis induced by HPV .", "output": "Enabling replicative immortality, Evading growth suppressors" }, { "input": "We report the formation , detection , quantitation and structural characterization of products resulting from the adduction of deoxynucleosides ( deoxyadenosine , deoxyguanosine , deoxycytidine and 5-methyldeoxycytidine ) to the catechol estrogens ( CE ) of estrone , estradiol-17beta and estradiol-17 alpha .\n\nThe crude products are obtained in a one-pot synthesis through oxidation of catechols to quinones and subsequent Michael-type reaction with the deoxynucleosides in acidic medium .\n\nIn all experiments , adducts are detected by electrospray ionization mass spectrometry analysis after HPLC separation ( LC/ESI/MS(n) ) .\n\nThe two pyrimidines deoxycytidine and 5-methyldeoxycytidine yield only CE adducts to deoxynucleosides , which correspond to stable adducts on DNA .\n\nFor purines , the results depend on the CE ( 2,3- or 3,4-catechols ) used , the function and configuration on carbon 17 ( ketone for estrone , alcohol for alpha and beta isomers of estradiol ) , and on the purine itself ( deoxyadenosine or deoxyguanosine ) .\n\nBoth stable adducts and deglycosylated adducts are formed , and therefore formation of stable adducts on DNA as well as the loss of purines from the DNA strands could be possible .\n\nMS(2) and MS(3) experiments prove to be relevant for further structural determinations , enabling in some cases the elucidation of the regiochemistry of adduction on the A and B rings of the steroid moiety .", "output": "Genomic instability and mutation" }, { "input": "Triggering tumor necrosis factor receptor-1 ( TNFR1 ) induces apoptosis in various cell lines .\n\nIn contrast , stimulation of TNFR1 in L929sA leads to necrosis .\n\nInhibition of HSP90 , a chaperone for many kinases , by geldanamycin or radicicol shifted the response of L929sA cells to TNF from necrosis to apoptosis .\n\nThis shift was blocked by CrmA but not by BCL-2 overexpression , suggesting that it occurred through activation of procaspase-8 .\n\nGeldanamycin pretreatment led to a proteasome-dependent decrease in the levels of several TNFR1-interacting proteins including the kinases receptor-interacting protein , inhibitor of kappa B kinase-alpha , inhibitor of kappa B kinase-beta , and to a lesser extent the adaptors NF-kappaB essential modulator and tumor necrosis factor receptor-associated factor 2 .\n\nAs a consequence , NF-kappa B , p38MAPK , and JNK activation were abolished .\n\nNo significant decrease in the levels of mitogen-activated protein kinases , adaptor proteins TNFR-associated death domain and Fas-associated death domain , or caspase-3 , -8 , and -9 could be detected .\n\nThese results suggest that HSP90 client proteins play a crucial role in necrotic signaling .\n\nWe conclude that inhibition of HSP90 may alter the composition of the TNFR1 complex , favoring the caspase-8-dependent apoptotic pathway .\n\nIn the absence of geldanamycin , certain HSP90 client proteins may be preferentially recruited to the TNFR1 complex , promoting necrosis .\n\nThus , the availability of proteins such as receptor-interacting protein , Fas-associated death domain , and caspase-8 can determine whether TNFR1 activation will lead to apoptosis or to necrosis .", "output": "Resisting cell death" }, { "input": "Genomic instability has long been recognized as the main feature of neoplasia and a factor modulating individual cancer susceptibility .\n\nThere are attempts to find effective assays of both individual DNA repair capacity and genetic instability , and their relation to the cancer risk .\n\nGenetic predisposition plays an important role in the etiology and development of head and neck squamous cell carcinoma ( HNSCC ) .\n\nThe aim of our study was to search for a correlation between chromosomal instability and DNA repair capacity in HNSCC patients and healthy controls .\n\nThe chromosomal instability was measured by the number of bleomycin ( BLM)-induced chromosomal aberrations and diepoxybutane ( DEB)-induced sister chromatid exchanges .\n\nThe DNA repair capacity was assessed using the DEB-induced adaptive response ( AR ) .\n\nThe HNSCC patients in our study showed a significant increase in chromosomal instability after a preterminal exposure of their lymphocytes to either BLM for the last 5 h or DEB for the last 24 h of incubation .\n\nHowever , the AR was higher in HNSCC patients than in the control group , suggesting an increase in the DNA repair capacity in the cancer patients as compared to the control .\n\nThere is no correlation between the DNA repair capacity estimated on the basis of preterminal exposures to BLM and DEB and the DNA repair capacity estimated on the basis of the adaptive response to DEB .\n\nThe preterminal exposure and the adaptive response test may activate different DNA repair mechanisms .", "output": "Genomic instability and mutation" }, { "input": "Deregulation of D-type cyclin-dependent kinases ( CDK4 and 6 ) is widely observed in various human cancers , illustrating their importance in cell cycle control .\n\nLike other cyclin-dependent kinases ( CDKs ) , assembly with cyclins is the most critical step for activation of CDK4/6 .\n\nAs previously reported elsewhere , we observed that the level of cyclinD1-CDK4 complex and its associated kinase activity were significantly low in asynchronously proliferating mouse embryo fibroblasts lacking both p21(Cip1) and p27(Kip1) ( p21/p27-null MEFs ) .\n\nThese evidences imply that p21(Cip1) and p27(Kip1) CDK inhibitors are ' essential activators ' of cyclin D-kinases .\n\nWe , however , discovered here that both the assembly and activation of cyclin D1-CDK4 complex occur when quiescent p21/p27-null MEFs were stimulated to re-enter the cell cycle .\n\nThis mitogen-induced cyclin D1-kinase activity was blocked by overexpression of p16(INK4a) and resulted in the inhibition of S phase entry in p21/p27-null MEFs .\n\nFurthermore , ectopic expression of p34(SEI-1) , a mitogen-induced CDK4 binding protein , increased the levels of active cyclinD1-CDK4 complex in asynchronously proliferating p21/p27-null MEFs .\n\nTogether , our results suggest that there are several independent ways to stimulate the assembly of cyclin D1-CDK4 kinases .\n\nAlthough p21(Cip1) and p27(Kip1) play a role in this process , our results demonstrate that additional mechanisms must occur in G0 to S phase transition .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The ultraviolet ( UV ) radiation present in sunlight is immune-suppressive .\n\nRecently we showed that solar-simulated UV radiation ( UVA + UVB ; 295-400 nm ) , applied after immunization , suppressed immunological memory and the elicitation of delayed-type hypersensitivity to the common opportunistic pathogen , Candida albicans .\n\nFurther , we found that wavelengths in the UVA region of the solar spectrum ( 320-400 nm ) , devoid of UVB , were equally effective in activating immune suppression as UVA + UVB radiation .\n\nHere we report on the mechanisms involved .\n\nNo immune suppression was found in UV-irradiated mice injected with monoclonal anti-interleukin ( IL)-10 antibody , or mice exposed to solar-simulated UV radiation and injected with recombinant IL-12 .\n\nAntigen-specific suppressor T cells were found in the spleens of mice exposed to UVA + UVB radiation .\n\nApplying liposomes containing bacteriophage T4N5 to the skin of mice exposed to solar-simulated UVA + UVB radiation or mice exposed to UVA radiation blocked immune suppression , demonstrating an essential role for UV-induced DNA damage in the suppression of established immune reactions .\n\nThese findings indicate that UV radiation activates similar immunological pathways to suppress the induction , or the elicitation , of the immune response .", "output": "Genomic instability and mutation, Avoiding immune destruction" }, { "input": "Rho GTPases are signaling molecules known to control cell motility .\n\nSeveral recent studies have suggested a role for Rho proteins in mediating tumor metastasis independent of their affects on cell proliferation .\n\nAs Rho proteins require post-translational modification with a geranlygeranyl moiety for full activity , we tested the affect of blocking geranylation on localization , downstream signaling , and stimulation of invasion .\n\nExpression of a constitutively active Rho construct in A375 melanoma cells dramatically stimulated invasion through Matrigel membranes ; however , a constitutively active RhoA mutated so that it cannot be geranylated , failed to stimulate invasion .\n\nMoreover , expression of epitope or GFP tagged modifications of this nongeranylatable constitutively active Rho demonstrated that geranylation is necessary for correct cellular localization of Rho .\n\nGeranylation was also found to be necessary for full downstream activation of serum response factor mediated transcription .\n\nPharmacologic inhibition of Rho geranylation produced similar inhibition of Rho localization , signaling , and invasion .\n\nOur results suggest that inhibition of Rho geranylation may be an attractive pharmacologic target for inhibiting melanoma metastasis .", "output": "Activating invasion and metastasis" }, { "input": "To assess the immune function of microglia and macrophages in brain tumors , the expression of MHC class II and B7 costimulatory molecules in three rodent glioma models was examined .\n\nMicroglia and macrophages , which accounted for 5-12% of total cells , expressed B7.1 and MHC class II molecules in the C6 and 9L tumors , but not RG2 gliomas .\n\nInterestingly , the expression of B7.1 and MHC class II molecules by microglia and macrophage was associated with an increase in the number of tumor-infiltrating lymphocytes in C6 and 9L tumors .\n\nB7.2 expression , which was present at low levels on microglia and macrophages in normal brain , did not significantly change in tumors .\n\nInterestingly , the expression of all three surface antigens increased after microglia were isolated from intracranial C6 tumors and cultured for a short period of time .\n\nWe conclude that microglia immune activity may be suppressed in gliomas and directly correlates to the immunogenecity of experimental brain tumors .", "output": "Avoiding immune destruction" }, { "input": "Peptide growth factors have been implicated in progression of prostate cancer ( PCa ) to the androgen-independent state ; however , much of the evidence linking diffusible mitogens and survival factors to this process remains circumstantial .\n\nHeparin-binding epidermal growth factor-like growth factor ( HB-EGF ) , a prostate stroma-derived factor , promotes survival , proliferation , and neuroendocrine differentiation of androgen-dependent LNCaP PCa cells in vitro .\n\nTo test whether sustained exposure to HB-EGF can confer an androgen-independent phenotype , we generated stable populations of LNCaP cells that express constitutively a secreted form of HB-EGF ( LNCaP/sHB ) .\n\nLNCaP/sHB cells proliferated more rapidly under androgen-depleted conditions in vitro and formed larger tumors with higher frequency in intact and castrated severe combined immunodeficient mice , in comparison to control cells .\n\nLNCaP/sHB tumors also expressed higher levels of the neuroendocrine marker , neuron-specific enolase , compared with control tumors .\n\nIn castrates , increased neuron-specific enolase expression in LNCaP/sHB tumors was associated with reduced androgen receptor ( AR ) levels .\n\nIn vitro , AR protein levels were reduced in LNCaP/sHB cells , and in transient transfection assays using an androgen-responsive promoter ( mouse mammary tumor virus-long terminal repeat ) , LNCaP/sHB cells showed reduced sensitivity to dihydrotestosterone compared with controls .\n\nThis is the first demonstration that continuous exposure of AR-positive PCa cells to a single growth factor can promote an androgen-independent phenotype in vivo .\n\nThese findings also emphasize the potential role of pathways other than the AR axis in acquisition of androgen independence .", "output": "Sustaining proliferative signaling" }, { "input": "Comparative mutagenesis studies of N-(2'-deoxyguanosin-8-yl)-2-acetylaminofluorene ( dG-AAF ) and N-(2'-deoxyguanosin-8-yl)-2-aminofluorene ( dG-AF ) adducts positioned in the Nar I restriction enzyme site were performed using Escherichia coli ( E. coli ) and simian kidney ( COS-7 ) cells .\n\nOligodeoxynucleotides ( (5)(')TCCTCG(1)G(2)CG(3)CCTCTC ) containing a recognition sequence for the Nar I restriction enzyme were modified site-specifically with dG-AAF or dG-AF .\n\nModified and unmodified oligomers inserted into single-stranded phagemid shuttle vectors were used to transform E. coli or to transfect COS-7 cells .\n\nFollowing replication in host cells , progeny plasmids were recovered and analyzed for mutations .\n\nIn SOS-induced E. coli , dG-AAF primarily induced one- and two-base deletions .\n\nThe mutational frequency varied , depending on the position modified in the Nar I site ; 91% two-base deletions were observed at G(3) , while 8.4% and 2.8% deletions were detected at G(2) and G(1) , respectively .\n\nIn contrast , dG-AF at any position in the Nar I site failed to produce deletions , generating primarily G --> T transversions ( mutational frequency , 7.6-8.4% ) .\n\nIn COS-7 cells , both dG-AAF and dG-AF primarily induced G --> T transversions .\n\nMutation frequencies for dG-AAF were 9.4-24% , the highest values being at G(1) and G(3) .\n\nMutation frequencies for dG-AF were 9.3-21% , the higher value at G(2) .\n\nWe conclude from this study that the mutation potential of dG-AAF and dG-AF depends on the structure of the adduct , the sequence context of the lesion , and the host cell used for the experiment .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND & OBJECTIVE Altered cell adhesion has a critical role in the development of epithelial cancers .\n\nE-cadherin act on the maintenance of cell-cell adhesion and its function was thought to be regulated by associated cytoplasmic proteins , such as alpha-catenin and beta-catenin .\n\nThis study was designed to examine the expression of beta-catenin in gastric carcinoma and to determine the relationship between tumor characteristics and survival .\n\nMETHODS Immunohistochemical staining of beta-catenin and E-cadherin was performed in 148 patients with gastric carcinoma .\n\nRESULTS Abnormal expression of beta-catenin and E-cadherin was performed in 148 patients with gastric carcinoma .\n\nRESULTS Abnormal expression of beta-catenin and E-cadherin was demonstrated in 43.2% and 44.6% of tumors respectively .\n\nUp to 63% of tumors stained abnormally for one or two components of the cadherin-catenin complex ( beta-catenin , E-cadherin ) .\n\nAbnormal beta-catenin and E-cadherin staining occurred more frequently in poor differentiated tumor than in good differentiated tumors ( P < 0.005 , respectively ) .\n\nThere was a significant correlation between abnormal beta-catenin expression and depth of invasion(P < 0.025 ) .\n\nMoreover , abnormal beta-catenin expression was more frequent in tumors with positive lymph node metastasis ( 45/84 , 53.6% ) and distance metastasis(21/31 , 67.7% ) than in tumors without lymph node metastasis ( 19/64 , 29.7% ) ( P < 0.005 ) and distance metastasis ( 43/117 , 36.8% ) ( P < 0.005 ) .\n\nA survival advantage was noted in tumors retaining normal membranous expression of beta-catenin , independent of type , grade , or stage ( P < 0.005 ) .\n\nCONCLUSIONS Abnormal expression of the E-cadherin-catenin complex occurs frequently in gastric carcinoma .\n\nThe close correlation with poor survival suggests that abnormal beta-catenin may be a useful prognostic marker .", "output": "Activating invasion and metastasis" }, { "input": "In an attempt to find a strategy to modulate the proliferation of vascular endothelial cells , we examined whether constitutive activation of proto-oncogen protein p21 ( Ras ) induced the reentry of confluent human umbilical vascular endothelial cells ( HUVECs ) into the S phase .\n\nWhen an adenovirus construct expressing a constitutively active Ras mutant ( Ad/RasG12V ) was infected into HUVECs , their morphology changed strikingly and they appeared to be transformed .\n\nHowever , Ad/RasG12V-infected HUVECs did not enter the S phase , as determined by assessing 3H-thymidine incorporation .\n\nIn accordance with the above results , the expression of cyclin A both at the transcript and protein levels did not increase in Ad/RasG12V-infected HUVECs relative to that in control cells , although the expression of cyclin D1 was induced in Ad/RasG12V-infected cells .\n\nInterestingly , the expression of the cyclin-dependent kinase ( CDK ) inhibitor p21cip1 was remarkably increased while that of p27kip1 did not decrease in Ad/RasG12V-infected HUVECs .\n\nFurthermore , CDK2 activity was not induced in Ad/RasG12V-infected HUVECs .\n\nThese results suggested that the constitutive activation of Ras promoted the reentry of confluent HUVECs in the G0 phase into the G1 phase , but not into the S phase .\n\nThe results also indicated that the constitutive activation of Ras might have induced the persistent expression of p21cip1 and p27kip1 , and that this induction of p21cip1 and p27kip1 expression possibly caused the cell cycle arrest at the G1 phase .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The influence of radiation-induced apoptosis on radiosensitivity was studied in a set of closely related human lymphoblastoid cell lines differing in TP53 status .\n\nThe clonogenic survival of irradiated TK6 cells ( expressing wild-type TP53 ) , WTK1 cells ( overexpressing mutant TP53 ) , and TK6E6 cells ( negative for TP53 owing to transfection with HPV16 E6 ) was assessed in relation to the induction of apoptosis and its suppression by caspase inhibition or treatment with PMA as well as after treatment with caffeine .\n\nMeasurements using the alkaline comet assay and pulsed-field electrophoresis of the induction and repair of DNA strand breaks showed similar kinetics of the processing of early DNA damage in these cell lines .\n\nThe cytochalasin B micronucleus assay revealed identical levels of residual damage in the first postirradiation mitosis of these cells .\n\nAbrogation of TP53-dependent apoptosis in TK6E6 cells resulted in a distinct increase in radioresistance .\n\nFurther suppression of apoptosis as observed in WTK1 cells overexpressing mutant TP53 apparently was not responsible for the high radioresistance of WTK1 cells , since other means of highly efficient suppression of apoptosis ( caspase inhibition or PMA treatment ) increased the clonogenic survival of irradiated TK6 cells only to levels similar to those of TK6E6 cells with abrogated TP53-dependent apoptosis .\n\nConsidering the similar levels of residual chromosomal damage in TK6E6 cells and WTK1 cells , a hitherto unknown mechanism of tolerance needs to be inferred for these TP53 mutant cells .\n\nThis residual damage tolerance , however , appears to require an intact G2/M-phase checkpoint function since the relative radioresistance of the WTK1 cells was completely lost upon caffeine treatment , which also resulted in a failure of the TK6 and TK6E6 cells to execute apoptosis .\n\nIn this situation , the cellular response seems to be dominated entirely by TP53-independent mitotic failure .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "AIM The study was designed to explore the effects of antisense human telomerase RNA ( ahTR ) on the malignant phenotype of gastric carcinoma cell line SGC-7901 , and its potential role in gene therapy for tumors .\n\nMETHODS An ahTR eukaryotic expression vector , including the sequence of template region of telomere repeats , was constructed by recombinant technology of molecules and then transfected into gastric carcinoma cell line SGC-7901 by liposome DOTAP .\n\nSubsequently , the expression of hTR RNA and ahTR RNA by reverse transcription-polymerase chain reaction , telomerase activity by telomeric repeat amplification protocol-ELISA ( TRAP-ELISA ) , telomere length by Southern blotting , cell morphology under light microscope , cellular proliferation capacity by 3-(4,5-dimethyl-2 thiazoyl)-2,5-diphenyl-2H-tetrazolium bromide assay , cell-cycle distribution by flow cytometry , efficiency of clone formation in soft agar , and tumorigenecity in nude mice were examined and evaluated in ahTR-transfected cells , control plasmid pCI-neo transfected cells and their parental cells .\n\nRESULTS An ahTR eukaryotic expression vector was constructed and successfully transfected into SGC-7901 cells .\n\nThe telomerase activity in ahTR-transfected SGC-7901 cells decreased from 100% to approximately 25% , and telomere length in the cells shortened to 3.35 from 4.08 Kb at 60 population doublings .\n\nCompared with the parental cells and pCI-neo transfected cells , ahTR-transfected cells displayed some morphological changes , such as decreased atypia , and recovery of contact inhibition and density inhibition under light microscope .\n\nFurthermore , ahTR-transfected cells displayed decreased invasive capacity in Borden's chamber invasive model , increased G0/G1 phase rate and apoptotic rate .\n\nSurprisingly , ahTR-transfected SGC-7901 cells lost their capacity for clone formation in soft agar and tumorigencity in nude mice .\n\nCONCLUSION Antisense-hTR transfection can inhibit the growth of SGC-7901 cells and partially reverse the malignant phenotypes .\n\nThis study provides an exciting approach for cancer therapy by inhibiting telomerase activity using an antisense gene .", "output": "Resisting cell death, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "We identified REDD1 as a novel transcriptional target of p53 induced following DNA damage .\n\nDuring embryogenesis , REDD1 expression mirrors the tissue-specific pattern of the p53 family member p63 , and TP63 null embryos show virtually no expression of REDD1 , which is restored in mouse embryo fibroblasts following p63 expression .\n\nIn differentiating primary keratinocytes , TP63 and REDD1 expression are coordinately downregulated , and ectopic expression of either gene inhibits in vitro differentiation .\n\nREDD1 appears to function in the regulation of reactive oxygen species ( ROS ) ; we show that TP63 null fibroblasts have decreased ROS levels and reduced sensitivity to oxidative stress , which are both increased following ectopic expression of either TP63 or REDD1 .\n\nThus , REDD1 encodes a shared transcriptional target that implicates ROS in the p53-dependent DNA damage response and in p63-mediated regulation of epithelial differentiation .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Estrogen affects apoptotic cell death in estrogen-responsive tissues .\n\nThe purpose of the present study was to examine dynamic changes in apoptotic cell death in the anterior pituitary gland during the estrous cycle and to investigate neuroendocrine regulation of these changes in cycling female rats .\n\nTerminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling ( TUNEL ) for in situ detection of DNA strand breaks revealed that the number of TUNEL-positive anterior pituitary cells was changing during the estrous cycle , with a maximum in the morning of proestrus and a minimum in the morning of estrus .\n\nA similar pattern was observed with Bax immunostaining ; however , no difference was observed in Bcl-2 immunostaining .\n\nMost of Bax-immunoreactive cells were identified as a subpopulation of gonadotropes .\n\nPentobarbital administered in the afternoon of proestrus attenuated the decrease in TUNEL-positive or Bax-immunoreactive cells in the morning of estrus , although estradiol treatment failed to affect it .\n\nThis action of pentobarbital was reduced by simultaneous treatment with an ovulation-inducing dose of gonadotropin-releasing hormone ( GnRH ) , but not with progesterone , an ovarian steroid also released after GnRH treatment .\n\nThese results suggest that anterior pituitary cells , mostly gonadotropes , undergo a cyclic change in apoptotic cell death during the estrous cycle and that the inhibition of apoptosis on estrus is due , at least in part , to the proestrous surge of GnRH secretion .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "PURPOSE The roles of terminal sialyl and fucosyl residues in cell surface glycans in the metastatic potential of H7721 cells , a human hepatocarcinoma cell line , were studied .\n\nMETHODS Neuraminidase and alpha-L-fucosidase were used to remove the sialyl and fucosyl residues , respectively .\n\nCell adhesion to fibronectin ( Fn ) , laminin ( Ln ) , and human umbilical vein epithelial cell ( HUVEC ) , as well as chemotactic cell migration and invasion , were selected as the parameters of metastatic potential ex vivo .\n\nRESULTS Sialyl residue is not essential for cell adhesion to Fn , but is important in cell adhesion to Ln and invasion , and is crucial in cell adhesion to HUVEC and migration .\n\nIn contrast , fucosyl residue contributes more than sialyl residue to cell adhesion to Fn and Ln , but less to adhesion to HUVEC , and is not essential in chemotactic cell migration and invasion .\n\nCell adhesion to HUVEC , migration , and invasion were inhibited by the monoclonal antibody of sialyl Lewis X , but not by the antibody of non-sialyl Lewis X. CONCLUSION Terminal sialyl residues on cell surface glycans are more important than fucosyl residues in mediating cell adhesion to HUVEC and cell migration/invasion , but the reverse is true in cell adhesion to Fn and Ln .", "output": "Activating invasion and metastasis" }, { "input": "Animal studies indicate that the immune system is one of the most sensitive targets of the toxic effects of 2,3,7,8-tetrachloro-p-dibenzodioxin ( TCDD ) .\n\nTCDD inhibits immunoglobulin secretion and decreases resistance to bacterial , viral , and parasitic infections in exposed animals .\n\nNearly 20 years after the Seveso , Italy , accident , we measured immunoglobulin and complement plasma levels in a random sample of the population in the most highly exposed zones ( n = 62 ) and in the surrounding noncontaminated area ( n = 58 ) .\n\nPlasma IgG levels decreased with increasing TCDD plasma concentration ( r = -0.35 , p = 0.0002 ) .\n\nMedian IgG concentration decreased from 1,526 mg/dL in the group with the lowest ( < 3.5 ppt ) TCDD levels to 1,163 mg/dL in the group with the highest ( 20.1-89.9 ppt ) TCDD levels ( p = 0.002 ) .\n\nThe association was significant ( p = 0.0004 ) after adjusting for age , sex , smoking , and consumption of domestic livestock and poultry in multiple regression analysis and persisted after exclusion of subjects with inflammatory diseases and those using antibiotics or nonsteroidal anti-inflammatory drugs .\n\nIgM , IgA , C3 , and C4 plasma concentrations did not exhibit any consistent association with TCDD levels .\n\nWe performed a systematic review of all the articles published between 1966 and 2001 on human subjects exposed to TCDD reporting information on circulating levels of immunoglobulins and/or complement components .\n\nThe literature indicates that the evidence for effects of TCDD on humoral immunity is sparse .\n\nMethodologic issues , results , and possible sources of variation between studies are discussed .\n\nThe possible long-term immunologic effects of TCDD exhibited by the participants of the present study , coupled with the increased incidence of lymphatic tumors in the area of the accident , warrant further investigation .", "output": "Avoiding immune destruction" }, { "input": "During mitotic exit , a small GTPase Tem1 needs to be activated .\n\nDuring most of the cell cycle , Tem1 activity is antagonized by a GTPase activating complex ( GAP ) composed of Bub2 and Bfa1 .\n\nBfa1 protein has cell cycle regulated phosphorylation depending upon the Polo-like kinase Cdc5 .\n\nThis phosphorylation dissociates Bfa1 from Tem1 and thus relieves the inhibition of Tem1 by the GAP complex .\n\nBub2 and Bfa1 are also required to prevent mitotic exit when there is DNA damage , spindle damage or spindle misorientation at G(2)/M phase .\n\nWhile Cdc5 inhibits Bfa1/Bub2 , mutating the Cdc5 phosphorylation sites on Bfa1 does not have a strong activating effect on Bub2/Bfa1 , suggesting there must be additional regulation in this pathway .\n\nHere we report that Bub2 protein also has cell cycle regulated phosphorylation .\n\nThis phosphorylation is partially dependent upon the Polo-like kinase Cdc5 and is consistent with negative regulation of the Bub2/Bfa1 GAP complex .\n\nSpindle damage or spindle misorientation prevents Bub2 phosphorylation .\n\nThe spindle damage effect is dependent upon the spindle assembly checkpoint components Mad2 and Mps1 .\n\nThus like Bfa1 , Bub2 protein is also controlled both during mitotic exit and in response to cell cycle checkpoints .\n\nBub2 phosphorylation is likely to be controlled by a novel kinase .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Liquid chromatography coupled to tandem mass spectrometry has been used for the detection and the structural characterization of T-rich model oligonucleotides covalently modified by estradiol-2,3-quinone .\n\nAfter separation by gradient elution , adducts were analyzed by negative electrospray mass spectrometry , enabling to evidence and localize the modifications in the oligonucleotide sequence .\n\nModifications by one molecule of estrogen were evidenced on purines ( A , G ) whereas no reaction was observed on pyrimidic bases ( T ) .\n\nIsomeric adducts were differentiated using tandem mass spectrometry , and energy resolved mass spectrometry allowed to underline differences in the behavior of the adducts towards collisional excitation into an ion trap device .", "output": "Genomic instability and mutation" }, { "input": "The capacity to repair DNA damage is an important factor that affects the therapeutic outcome in cancer treatment .\n\nTo clarify the cellular repair response , we investigated the kinetics of DNA excision repair initiated by 1,3-bis(2-chloroethyl)-1-nitrosourea ( BCNU ) in human leukemia CCRF-CEM cells at an exponential growth phase in vitro .\n\nUsing the alkaline single-cell gel electrophoresis ( comet ) assay , we quantitated the repair kinetics as the amount of DNA single-strand breaks that were generated from the incision and were diminished by the rejoining in the repair process .\n\nCEM cells could initiate DNA excision repair in response to BCNU by starting an incision reaction .\n\nHowever , the incision capacity came to a plateau at a concentration of 80 to 100 microM or after an incubation time of 90 to 120 minutes .\n\nWhen the cells were pulsed with 40 microM BCNU , the maximal incision occurred at the end of the incubation period , and the repair process was completed within 4 hours When cells were treated with 100 microM BCNU , the incised DNA was not rejoined at 4 hours , suggesting that the repair was not completed .\n\nHigher concentrations might surpass the cellular capacity for repair and would be associated with increased cell death .\n\nEvaluation of the repair process may provide a clue for therapeutic strategies to improve clinical efficacy if accelerated DNA repair is responsible for the drug resistance .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Prostaglandin E(2) ( PGE(2) ) receptor subtype EP(2) , which is coupled to cAMP metabolism , is known to mediate proliferation of primary human keratinocytes in vitro .\n\nThe effect of gain or loss of EP(2) receptors in immortalized human keratinocytes ( HaCat cells ) was examined .\n\nHaCat keratinocytes were transfected with sense or anti-sense constructs of the EP(2) receptor .\n\nLoss or gain of EP(2) expression was documented by immunoblot and associated changes in agonist-stimulated cAMP production .\n\nLoss or gain of EP(2) receptor expression correlated with alterations in plating efficiencies but with modest affects on growth .\n\nWhen cell lines were studied in an organ culture model , anti-sense clones were highly invasive compared with vector controls and sense transfectants .\n\nA marked increase in prostaglandin production is commonly seen in malignant lesions .\n\nBecause prostaglandin receptors are known to undergo ligand-induced receptor down-regulation , we sought to determine whether EP(2) receptor down-regulation results in increased invasiveness .\n\nIn vector controls , invasiveness was reproduced by ligand-dependent EP(2) receptor down-regulation as assessed by immunohistochemistry .\n\nIn addition , loss of EP(2) receptor expression was associated with decreased paxillin expression , a critical component of focal adhesion assembly .\n\nThus , down-regulation of EP(2) receptors represents a potential mechanism for neoplastic progression to an invasive phenotype .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "The goal of this study was to determine the prevalence of sequence variants in the class I beta-tubulin ( clone m40 ) gene and their occurrence in human tumors and cancer cell lines .\n\nDNA was isolated from 93 control individuals representing a wide variety of ethnicities , 49 paclitaxel-naive specimens ( 16 ovarian cancers , 17 non-small cell lung cancers , and 16 ovarian cancer cell lines ) , and 30 paclitaxel-resistant specimens ( 9 ovarian cancers , 9 ovarian cancer cell lines , and 12 ovarian cancer xenografts in nude mice ) .\n\nDenaturing high-performance liquid chromatography and direct sequence analysis detected two silent polymorphisms in exon 4 , Leu217Leu ( CTG/CTA ) and Gly400Gly ( GGC/GGT ) , with minor allele frequencies of 17 and 0.5% , respectively .\n\nFive nucleotide substitutions and one single-base deletion were detected in introns 1 , 2 , and 3 and in the 3 ' untranslated region .\n\nAnalysis of 49 paclitaxel-naive and 30 paclitaxel-resistant specimens revealed no additional polymorphisms in the coding region .\n\nIn addition , no amino acid replacements were found in chimpanzee , gorilla , and orangutan in comparison to human .\n\nOur data demonstrate a very high degree of sequence conservation in class I beta-tubulin , suggesting that all residues are important in tubulin structure and function .\n\nIndividual variation in response to treatment with paclitaxel is not likely to be caused by genetic variations in the beta-tubulin drug target .\n\nMoreover , acquired mutations in class I beta-tubulin are unlikely to be a clinically relevant cause of drug resistance .", "output": "Genomic instability and mutation" }, { "input": "Our previous studies have shown that genistein inhibits the growth of PC3 prostate cancer cells and induces apoptosis by inhibiting nuclear factor kappaB ( NF-kappaB ) and Akt signaling pathways .\n\nTo better understand the precise molecular mechanism(s) by which genistein exerts its effects on PC3 cells , we utilized cDNA microarray to interrogate 12,558 known genes to determine the gene expression profiles altered by genistein treatment .\n\nWe found a total of 832 genes that showed a greater than twofold change after genistein treatment from two independent experiments with a high degree of concordance .\n\nAmong these genes , 774 genes were down-regulated and 58 genes were up-regulated with genistein treatment .\n\nCluster analysis showed nine different types of expression alternations .\n\nThese genes were also subjected to cluster analysis according to their biological functions .\n\nWe found that genistein regulated the expression of genes that are critically involved in the regulation of cell growth , cell cycle , apoptosis , cell signaling transduction , angiogenesis , tumor cell invasion and metastasis .\n\nReverse transcription-polymerase chain reaction ( RT-PCR ) analysis was used to confirm the results of cDNA microarray , and the results of RT-PCR were consistent with the microarray data .\n\nWe conclude that genistein affected the expression of a large number of genes that are related to the control of cell survival and physiologic behaviors .\n\nThe gene expression profiles provide comprehensive molecular mechanism(s) by which genistein exerts its pleiotropic effects on cancer cells .\n\nGenistein-induced regulation of these genes may be further exploited for devising chemopreventive and/or therapeutic strategies for prostate cancer .", "output": "Inducing angiogenesis, Resisting cell death, Activating invasion and metastasis" }, { "input": "The signal transducer and activator of transcription ( STAT)1 is a cytoplasmic-transcription factor that is phosphorylated by Janus kinases ( Jak ) in response to interferon gamma(IFN-gamma) .\n\nThe phosphorylated STAT1 translocates to the nucleus , where it turns on specific sets of IFN-gamma-inducible genes , such as the interferon regulatory factor ( IRF)-1 .\n\nWe show here that gamma irradiation reduces the IFN-gamma mRNA expression .\n\nThe inhibition of the STAT1 phosphorylation and the IRF-1 expression by gamma irradiation was also observed .\n\nIn contrast , the mRNA levels of IL-5 and transcription factor GATA-3 were slightly induced by gamma irradiation when compared to the non-irradiated sample .\n\nFurthermore , we detected the inhibition of cell-mediated immunity by gamma irradiation in the allogenic-mixed lymphocytes ' reaction ( MLR ) .\n\nThese results postulate that gamma irradiation induces the polarized-Th2 response and interferes with STAT1 signals , thereby causing the immunosuppression of the Th1 response .", "output": "Avoiding immune destruction" }, { "input": "Properly regulated keratinocyte cell death is fundamentally important to maintain structural integrity and homeostatic function of epidermis .\n\nMoreover , from an oncological perspective , therapeutic approaches selectively targeting apoptosis of malignant cell types while sparing normal keratinocytes in surrounding skin is desirable .\n\nApo2Ligand/tumor necrosis factor-related apoptosis-inducing ligand ( Apo2L/TRAIL ) has been observed to preferentially induce cytopathic effects on transformed/malignant cell types compared with their non-neoplastic counterparts .\n\nIn this report , two different biologically active preparations of Apo2L/TRAIL , a non-tagged version , NT-Apo2L/TRAIL , and a leucine zipper fusion protein , LZ-Apo2L/TRAIL , were examined for their ability to trigger apoptosis in normal human keratinocytes , and in an immortalized cell line ( HaCaT cells ) .\n\nDifferences between these preparations were observed , including : NT-Apo2L/TRAIL induced less keratinocyte apoptosis compared with LZ-Apo2L/TRAIL ; NT-Apo2L/TRAIL also induced less apoptosis of HaCaT cells compared with LZ-Apo2L/TRAIL ; LZ-Apo2L/TRAIL but not NT-Apo2L/TRAIL induced cytotoxic effects when keratinocytes became growth arrested due to undergoing spontaneous replicative senescence--a biological state previously observed to be resistant to UV-light-induced apoptosis .\n\nSimilarities between preparations included : an enhanced ability for both Apo2L/TRAIL preparations to kill a greater relative percentage of HaCaT cells compared with keratinocytes ; enhanced cytotoxicity towards keratinocytes that had their NF-B activity inhibited ; a dependence of both Apo2L/TRAIL preparations on FADD and caspase activation ; triggering of the same caspase cascades including caspase 8 and 3 ; and an ability to induce apoptosis even when HaCaT cells and keratinocytes were transduced to overexpress either Bcl-2 or Bcl-x(L) ( survival factors that reduce susceptibility to UV-light-induced apoptosis ) .\n\nThese results indicate that while both preparations of Apo2L/TRAIL possess biological activity , there are important differences as regards their ability to induce apoptosis in normal and immortalized keratinocytes .\n\nMoreover , the death receptor pathway triggered by LZ-Apo2L/TRAIL can overcome the apoptotic resistance normally observed in response to UV-light mediated by Bcl-2/Bcl-x(L) , as well as by the state of cellular senescence .\n\nUnraveling the molecular basis for these differential biological effects may reveal a new strategic role for these death receptor/ligands linked to apoptosis in maintaining the dynamic balance of keratinocyte proliferation , differentiation , and cell death necessary to achieve a homeostatic thickness and function of normal skin .\n\nIn addition , it may be possible to utilize these Apo2L/TRAIL preparations for the treatment of various sun-induced skin cancers as they can differentially trigger apoptosis of transformed keratinocytes , or keratinocytes with abnormal NF-kappaB signaling , while sparing adjacent normal keratinocytes .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "4-Hydroxy-2-nonenal ( HNE ) , a major racemic product of lipid peroxidation , reacts with histidine to form a stable HNE-histidine Michael addition-type adduct possessing three chiral centers in the cyclic hemiacetal structure .\n\nIn the present study , we characterized configurational isomers of a HNE-N(alpha)-acetylhistidine adduct by NMR spectroscopy and by molecular orbital calculations .\n\nIn addition , we raised monoclonal antibodies against ( R)-HNE-histidine and ( S)-HNE-histidine adducts , characterized their specificities , and examined in vivo localizations of each adduct under oxidative stress .\n\nTo facilitate structural characterization of the configurational isomers of an HNE-histidine adduct , we prepared the ( R)-HNE-histidine and ( S)-HNE-histidine adducts by incubating N(alpha)-acetylhistidine with each HNE enantiomer , both of which provided two peaks ( Ra and Rb from ( R)-HNE-histidine and Sa and Sb from ( S)-HNE-histidine adducts ) in reversed-phase high-performance liquid chromatography .\n\nThe NMR analysis showed that each peak was a mixture of two diastereomers .\n\nIn addition , the analysis of the nuclear Overhauser effect enabled the determination of configurations of the eight isomers .\n\nThe relative amounts of these isomers in the NMR analysis correlated with the relative energies calculated by molecular orbital methods .\n\nOn the other hand , using ( R)-HNE-modified and ( S)-HNE-modified keyhole limpet hemocyanins as the antigens , we raised the monoclonal antibodies , mAbR310 and mAbS412 , which enantioselectively recognized the ( R)-HNE-histidine and ( S)-HNE-histidine adducts , respectively .\n\nAmong the mixtures ( Ra , Rb , Sa , and Sb ) of diastereomers , mAbR310 showed the highest immunoreactivity to Rb ( the mixture of 2R,4S,5R and 2S,4S,5R isomers ) , whereas mAbS412 preferentially recognized Sa ( the mixture of 2R,4S,5S and 2S,4S,5S isomers ) .\n\nThe presence of ( R)-HNE and ( S)-HNE epitopes in vivo was immunohistochemically examined in the kidney of rats exposed to the renal carcinogen , ferric nitrilotriacetate , by which nuclear and cytosolic stainings with mAbR310 and mAbS412 , respectively , were detected .", "output": "Genomic instability and mutation" }, { "input": "Transfected linear DNA molecules are substrates for double-strand break ( DSB ) repair in mammalian cells .\n\nThe DSB repair process can involve recombination between the transfected DNA molecules , between the transfected molecules and chromosomal DNA , or both .\n\nIn order to determine whether these different types of repair events are linked , we devised assays enabling us to follow the fate of linear extrachromosomal DNA molecules involved in both interplasmid and chromosome-plasmid recombination , in the presence or absence of a pre-defined chromosomal DSB .\n\nPlasmid-based vectors were designed that could either recombine via interplasmid recombination or chromosome-plasmid recombination to produce a functional beta-galactosidase ( betagal ) fusion gene .\n\nBy measuring the frequency of betagal+ cells at 36 h post-transfection versus the frequency of betagal+ clones after 14 days , we found that the number of cells containing extrachromosomal recombinant DNA molecules at 36 h ( i.e. , betagal+ ) , either through interplasmid or chromosome-plasmid recombination , was nearly the same as the number of cells integrating these recombinant molecules .\n\nFurthermore , when a predefined DSB was created at a chromosomal site , the extrachromosomal recombinant DNA molecules were shown to integrate preferentially at that site by Southern and fiber-FISH ( fluorescence in situ hybridization ) analysis .\n\nTogether these data indicate that the initial recombination event can potentiate or commit extrachromosomal DNA to integration in the genome at the site of a chromosomal DSB .\n\nThe efficiency at which extrachromosomal recombinant molecules are used as substrates in chromosomal DSB repair suggests extrachromosomal DSB repair can be coupled to the repair of chromosomal DSBs in mammalian cells .", "output": "Genomic instability and mutation" }, { "input": "The ability of p53 to alter , at the transcriptional level , the gene expression of downstream targets is critical for its role as a tumor suppressor .\n\nMost models of p53 activation postulate the stepwise recruitment by p53 of coactivators , histone acetyltransferases , and/or chromatin remodeling factors to a promoter region to facilitate the subsequent access of the general transcriptional machinery required for transcriptional induction .\n\nWe demonstrate here , however , that the promoter regions for the p53 target genes , p21 , 14-3-3sigma , and KARP-1 , exist in a constitutively open conformation that is readily accessible to DNase I. This conformation was not altered by DNA damage or by whether p53 was present or absent in the cell .\n\nIn contrast , p53 response elements , which resided outside the immediate promoter regions , existed within DNase I-resistant chromatin domains .\n\nThus , p53 activation of downstream target genes occurs without p53 inducing chromatin alterations detectable by DNase I accessibility at either the promoter or the response element .\n\nAs such , these data support models of p53 activation that do not require extensive chromatin alterations to support cognate gene expression .", "output": "Genomic instability and mutation" }, { "input": "The HT29 adenocarcinoma is a common model of epithelial cell differentiation and colorectal cancer and its death is an oft-analyzed response to TNF family receptor signaling .\n\nThe death event itself remains poorly characterized and here we have examined the involvement of caspases using pan-caspase inhibitors. zVAD-fmk did not block death of HT29 cells in response to activation of the Fas , TRAIL , TNF , TWEAK and LTbeta receptors .\n\nThe secondary induction of TNF or the other known bona fide death inducing ligands did not account for death following LTbeta receptor activation indicating that TNF family receptors can trigger a caspase-independent death pathway regardless of the presence of canonical death domains in the receptor .\n\nTo provide a frame of reference , the phenotype of HT29 death was compared to four other TNF family receptor triggered death events ; Fas induced Jurkat cell apoptosis , TNF/zVAD induced L929 fibroblast necrosis , TNF induced death of WEHI 164 fibroblastoid cells and TNF/zVAD induced U937 death .\n\nThe death of HT29 and U937 cells under these conditions is an intermediate form with both necrotic and apoptotic features .\n\nThe efficient coupling of TNF receptors to a caspase-independent death event in an epithelial cell suggests an alternative approach to cancer therapy .", "output": "Resisting cell death" }, { "input": "Tumor metastasis represents a complex multistep process that requires migration , invasion , and angiogenesis .\n\nIn this study , we examined the impact of molecular blockade of the epidermal growth factor receptor on the invasive and metastatic capacity of human squamous cell carcinoma ( SCC ) of the head and neck using in vitro and in vivo model systems .\n\nTreatment with the anti-epidermal growth factor receptor antibody C225 attenuated the migration of SCC-1 tumor cells through a chemotaxis chamber in a dose-dependent manner .\n\nIncubation of SCC cells with 10-100 nM C225 for 4 h resulted in 40-60% inhibition of cell migration .\n\nFurthermore , in the presence of C225 , the capacity of SCC-1 to invade across a layer of extracellular matrix ( Matrigel ) was significantly inhibited .\n\nUsing an in vivo orthotopic floor-of-mouth xenograft model , locoregional tumor invasion of SCC-1 into muscle , vessel , bone , and perineural tissues was inhibited in C225-treated mice .\n\nThis inhibition was additionally characterized by down-regulation in the expression of matrix metalloproteinase-9 .\n\nThese data suggest that inhibition of metastatic potential by C225 may be mediated via decreased migration and invasion of SCC cells .\n\nRegarding angiogenesis in vitro , we first studied human umbilical vascular endothelial cells , which established a capillary-like network structure ( tube formation ) in the presence of reconstituted Matrigel .\n\nTreatment with C225 reduced cell-to-cell interaction of human umbilical vascular endothelial cells , resulting in disruption of tube formation .\n\nThe effect of C225 was additionally examined using an in vivo tumor xenograft neovascularization model of angiogenesis .\n\nSystemic treatment with C225 not only reduced tumor growth and the number of blood capillaries but also hindered the growth of established vessels toward the tumor .\n\nTaken together , these results provide evidence that C225 can suppress tumor-induced neovascularization and metastasis in SCC of the head and neck .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "A number of cancer chemotherapeutic drugs designed to have cytotoxic actions on tumor cells have recently been shown to also have antiangiogenic activities .\n\nEndothelial cell migration and proliferation are key components of tumor angiogenesis , and agents that target the microtubule cytoskeleton can interfere with these processes .\n\nIn this study , the effect on endothelial cell functions of the microtubule-stabilizing drugs Taxotere and Taxol were evaluated in three in vitro assays : a chemokinetic migration assay , an angiogenesis factor-mediated chemotactic migration assay , and a three-dimensional Matrigel tubule formation assay , using rat fat pad endothelial cells ( RFPECs ) and/or human umbilical vein endothelial cells ( HUVECs ) .\n\nTaxotere was active in all three assays at concentrations that were not cytotoxic and did not inhibit endothelial cell proliferation .\n\nIn the RFPEC chemokinetic migration and in vitro tubule formation assays , the IC50 values were approximately 10(-9) M for both Taxotere and Taxol .\n\nHUVEC migration , however , was more sensitive to Taxotere , with an observed IC50 of 10(-12) M in a chemokinetic assay .\n\nIn a Boyden chamber assay , HUVEC chemotaxis stimulated by either of two angiogenic factors , thymidine phosphorylase or vascular endothelial growth factor , was inhibited by Taxotere with an IC50 of 10(-11) M and was ablated at 10(-9) M. Taxotere was also up to 1000-fold more potent than Taxol in inhibiting either chemokinetic or chemotactic migration .\n\nWhen the microtubule cytoskeleton was visualized using immunofluorescence staining of alpha-tubulin , there were no gross morphological changes observed in HUVECs or RFPECs treated with Taxotere at concentrations that inhibited endothelial cell migration but not proliferation .\n\nThe effects of Taxotere on migration were associated with a reduction in the reorientation of the cell's centrosome , at concentrations that did not affect gross microtubule morphology or proliferation .\n\nReorientation of the centrosome , which acts as the microtubule organizing center , in the intended direction of movement is a critical early step in the stabilization of directed cell migration .\n\nThese data indicate that endothelial cell migration correlates more closely with changes in microtubule plasticity than with microtubule gross structure .\n\nThe antiangiogenic activity of Taxotere in vivo was assessed in a Matrigel plug assay .\n\nIn this assay , the angiogenic response to fibroblast growth factor 2 was inhibited in vivo by Taxotere with an ID50 of 5.4 mg/kg when injected twice weekly over a 14-day period , and angiogenesis was completely blocked in mice that received 10 mg/kg Taxotere .\n\nThe in vivo data further suggested that Taxotere had selectivity for endothelial cell migration and/or microvessel formation because infiltration of inflammatory cells into the Matrigel plug was much less sensitive to inhibition by Taxotere .\n\nIn conclusion , Taxotere is a potent and potentially specific inhibitor of endothelial cell migration in vitro and angiogenesis in vitro and in vivo .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "Approximately 10% of the U.S. population ingests <50% of the current recommended daily allowance for zinc .\n\nWe investigate the effect of zinc deficiency on DNA damage , expression of DNA-repair enzymes , and downstream signaling events in a cell-culture model .\n\nLow zinc inhibited cell growth of rat glioma C6 cells and increased oxidative stress .\n\nLow intracellular zinc increased DNA single-strand breaks ( comet assay ) .\n\nZinc-deficient C6 cells also exhibited an increase in the expression of the zinc-containing DNA-repair proteins p53 and apurinic endonuclease ( APE ) .\n\nRepletion with zinc restored cell growth and reversed DNA damage .\n\nAPE is a multifunctional protein that not only repairs DNA but also controls DNA-binding activity of many transcription factors that may be involved in cancer progression .\n\nThe ability of the transcription factors p53 , nuclear factor kappaB , and activator protein 1 ( AP1 ) to bind to consensus DNA sequences was decreased markedly with zinc deficiency , as assayed by electrophoretic mobility-shift assays .\n\nThus , low intracellular zinc status causes oxidative DNA damage and induces DNA-repair protein expression , but binding of p53 and important downstream signals leading to proper DNA repair are lost without zinc .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Ovarian hormones have a pivotal role in the control of proliferation in the mammary gland , and cumulative life-time exposure to ovarian hormones is known to be a determinant of breast cancer risk .\n\nWe have shown previously that a p.o.-active , long-acting butyrate analogue , sodium phenylbutyrate ( PB ) , reduced proliferation in normal and malignant human breast cells in culture and reduced expression of ovarian hormone receptors , suggesting that PB had cellular effects consistent with decreasing breast cancer risk .\n\nThe aim of this study was to determine the in vivo effects of PB in the normal mammary gland on epithelial cell proliferation , estrogen receptor alpha ( ER alpha ) expression , and cyclin D1 expression .\n\nBALB/c mice were treated with PB , delivered by mini-osmotic pumps , for 7 days .\n\nModerate ( 250 mg/kg/day ) and high ( 500 mg/kg/day ) PB treatment resulted in a decrease in proliferation in mammary epithelial cells ( P < 0.001 ) , determined by bromodeoxyuridine incorporation .\n\nAnalysis of ER alpha immunostaining revealed a significant reduction in moderate- and high-treatment groups ( P = 0.01 and P = 0.02 ) , and expression of cyclin D1 was virtually ablated ( P < 0.001 ) .\n\nHistone deacetylase inhibition is a known mechanism of butyrate action , and consistent with this , PB increased levels of acetylated histone H3 in the mammary gland .\n\nIn summary , PB decreased proliferation in the mammary gland in vivo at clinically achievable doses .\n\nDecreased proliferation was accompanied by changes in the levels of ER alpha and cyclin D1 .\n\nThese data show that PB modulates parameters thought to be involved in the carcinogenic process in the normal mammary gland , and compounds in this class may therefore be useful candidates for breast cancer chemoprevention .", "output": "Sustaining proliferative signaling" }, { "input": "Although there have been numerous studies of site-specific mutagenesis by dGuo adducts of benzo[a]pyrene diol epoxides ( B[a]P DEs ) , the present study represents the first example of site-specific mutagenesis by dGuo adducts of the highly carcinogenic benzo[c]phenanthrene 3,4-diol 1,2-epoxides ( B[c]Ph DEs ) .\n\nThe eight adducts that would result from cis- and trans-opening at C-1 of four optically active isomers of B[c]Ph DEs by the N(2)-amino group of dGuo were incorporated into 5'-TTCGAATCCTTCCCCC ( context III ) and 5'-GGGGTTCCCGAGCGGC ( context IV ) at the underlined site .\n\nThese modified oligonucleotides along with unmodified controls were ligated into single-stranded M13mp7L2 , which were then used to transfect SOS-induced Escherichia coli .\n\nUpon replication of the lesions in each of the two sequence contexts , mutational analysis of the progeny was performed by differential hybridization .\n\nFor the 16 adducts , the mutation frequencies varied over 2 orders of magnitude with a reasonably even distribution ( 0.4-1% for three adducts , 1-2% for six adducts , 3-7.4% for five adducts , and one adduct each at 11 and 39% ) .\n\nFor all but this last adduct , the mutation frequency for a given B[c]Ph DE adduct was less than for its B[a]P analogue with the same stereochemistry in the same sequence .\n\nFor the vectors containing adducts with S configuration at the site of attachment of the hydrocarbon to the dGuo base , the main base substitution was G --> T followed by G --> A. In contrast , for the vectors containing adducts with R configuration , the main base substitution was G --> A. The most notable observation in the present study is the low frequency of mutations induced by the B[c]Ph DE-dGuo adducts relative to their B[a]P counterparts .\n\nA possible structural basis for this difference is proposed .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND/PURPOSE Although angiogenic factors may play an important role in the biology of neuroblastoma , which frequently spreads hematogenously , the mechanism remains unclear .\n\nThe authors studied tumor progression and invasion from the perspective of angiogenesis and sought to understand the features of this type of tumor .\n\nMETHODS Thirty-one specimens were resected from patients with neuroblastoma and the expression of vascular endothelial growth factor ( VEGF ) , and its receptor ( Flk-1 ) was examined using immunohistochemistry .\n\nThe authors looked for correlations among the expressions of VEGF and its receptor with various clinicopathologic factors .\n\nIn addition , they examined the expression and location of VEGF and Flk-1 mRNA in 10 primary neuroblastoma using in situ hybridization .\n\nRESULTS Both in situ hybridization and immunohistochemistry showed the presence of VEGF expression within the neuroblastoma cells .\n\nWe found VEGF mRNA in neuroblastoma cells but not vascular endothelial cells according to in situ hybridization .\n\nFurther , Flk-1 mRNA was present both in neuroblastoma cells and vascular endothelial cells .\n\nThe level of VEGF expression was higher in unfavorable histology , using the criteria of Shimada , than in favorable histology .\n\nCONCLUSION The authors suggest that paracrine and autocrine systems are involved in the angiogenesis of neuroblastoma , and the expression of VEGF correlates with the prognosis in neuroblastoma .", "output": "Inducing angiogenesis" }, { "input": "We investigated the effectiveness of continuous hyperthermic peritoneal perfusion ( CHPP ) for the peritoneal dissemination of gastric cancer .\n\nA total 124 patients with advanced gastric cancer were enrolled in this study .\n\nProphylactic CHPP ( P-CHPP ) was performed in 45 patients who had macroscopic serosal invasion without peritoneal dissemination , and 79 patients without CHPP were a control group .\n\nTherapeutic CHPP ( T-CHPP ) was performed in 21 patients with peritoneal dissemination , and 52 patients without CHPP were a control group .\n\nThere was no significant difference in 5 year survival between patients treated and not treated with P-CHPP .\n\nUnivariate analysis showed that location of tumor , tumor diameter , and lymph node metastasis influenced prognosis , but there was no prognostic factor in the Cox proportional regression hazard model .\n\nThere was no significant difference in 5-year survival between patients treated and not treated with T-CHPP .\n\nUnivariate analysis showed that degree of peritoneal dissemination and adjuvant chemotherapy influenced prognosis , and the Cox proportional regression hazard model showed that the macroscopic types and degree of peritoneal dissemination affected prognosis .\n\nIn the patients with CHPP , the incidences of respiratory failure and renal failure were each statistically greater than in the patients undergoing CHPP .", "output": "Activating invasion and metastasis" }, { "input": "Ionizing radiation is a potent inducer of DNA damage because it causes single- and double-strand breaks , alkali-labile sites , base damage , and crosslinks .\n\nThe interest in ionizing radiation is due to its environmental and clinical implications .\n\nSingle-strand breaks , which are the initial damage induced by a genotoxic agent , can be used as a biomarker of exposure , whereas the more biologically relevant double-strand breaks can be analyzed to quantify the extent of damage .\n\nIn the present study the effects of 137CS gamma-radiation at doses of 1 , 5 , and 10 Gray on DNA and subsequent repair by C3H10T1/2 cells ( mouse embryo fibroblasts ) were investigated .\n\nTwo versions of the comet assay , a sensitive method for evaluating DNA damage , were implemented : the alkaline one to detect single-strand breaks , and the neutral one to identify double-strand breaks .\n\nThe results show a good linear relation between DNA damage and radiation dose , for both single-strand and double-strand breaks .\n\nA statistically significant difference with respect to controls was found at the lowest dose of 1 Gy .\n\nHeterogeneity in DNA damage within the cell population was observed as a function of radiation dose .\n\nRepair kinetics showed that most of the damage was repaired within 2 h after irradiation , and that the highest rejoining rate occurred with the highest dose ( 10 Gy ) .\n\nSingle-strand breaks were completely repaired 24 h after irradiation , whereas residual double-strand breaks were still present .\n\nThis finding needs further investigation .", "output": "Genomic instability and mutation" }, { "input": "The RuvABC and RecBCD proteins promote rescue of stalled or broken DNA replication forks in Escherichia coli .\n\nStrains lacking these proteins cope poorly with DNA damage and have problems with chromosome segregation and cell division .\n\nWe show how these difficulties are overcome to varying degrees by a sub-class of RNA polymerase mutations selected for their stringent phenotype .\n\nThirty-five mutations were sequenced .\n\nAll but one change single amino acids in RpoB or RpoC that lie on or near the path taken by DNA through the enzyme , indicating they may affect the stability of transcription complexes .\n\nFour mutant enzymes are shown to form unstable open complexes at the lambdacro promoter .\n\nAt least one may also release stalled complexes or limit their formation , as it reduces the need for reactivation of transcription by GreA or GreB , and for transcription-coupled DNA repair of UV damage by Mfd .\n\nThe results shed light on the interplay between DNA replication and transcription and suggest ways in which conflicts between these two vital cellular processes are avoided or resolved .", "output": "Genomic instability and mutation" }, { "input": "The Escherichia coli AlkB protein protects against the cytotoxicity of methylating agents by repair of the DNA lesions 1-methyladenine and 3-methylcytosine , which are generated in single-stranded stretches of DNA .\n\nAlkB is an alpha-ketoglutarate- and Fe(II)-dependent dioxygenase that oxidizes the relevant methyl groups and releases them as formaldehyde .\n\nHere , we identify two human AlkB homologs , ABH2 and ABH3 , by sequence and fold similarity , functional assays , and complementation of the E. coli alkB mutant phenotype .\n\nThe levels of their mRNAs do not appear to correlate with cell proliferation but tissue distributions are different .\n\nBoth enzymes remove 1-methyladenine and 3-methylcytosine from methylated polynucleotides in an alpha-ketoglutarate-dependent reaction , and act by direct damage reversal with the regeneration of the unsubstituted bases .\n\nAlkB , ABH2 , and ABH3 can also repair 1-ethyladenine residues in DNA with the release of acetaldehyde .", "output": "Genomic instability and mutation" }, { "input": "The effect of peptides released during the fermentation of milk on the humoral immune system and on fibrosarcoma growth was studied .\n\nLactobacillus helveticus was able to release peptidic compounds during milk fermentation due to its high proteolytic activity , as was shown by the degree of proteolysis and size-exclusion HPLC elution profiles .\n\nThree fractions of these compounds were separated and fed to mice during different periods ( 2 , 5 , and 7 d ) .\n\nThe humoral immune response was assessed by following the number of IgA-secreting cells , and the antitumor activity was monitored by studying the regression of subcutaneously implanted fibrosarcomas .\n\nFeeding during 2 and 7 d with the medium-sized fraction ( Fraction II ) significantly increased the IgA-producing cells in the intestines , whereas feeding with the large compound fraction ( Fraction I ) during 5 d and the small compound fraction ( Fraction III ) during all three feeding periods provided similar increases .\n\nA double dose of Fraction II showed the highest IgA-producing cell count .\n\nThe increase by Fraction III was shown to be caused by the presence of L-Tryptophan .\n\nFraction II significantly decreased the size of fibrosarcoma when previously fed during 7 d , and feeding with Fraction I during 5 d decreased significantly its size after 35 d of growth .\n\nAlthough the mechanisms by which lactic acid bacteria enhance the immune system are not clear , this study clearly shows that bioactive compounds released in fermented milks contribute to the immunoenhancing and antitumor properties of these products .\n\nThe release of bioactive peptides by lactic acid bacteria can have important implications on the modulation of the cellular immune response .", "output": "Avoiding immune destruction" }, { "input": "Food-derived heterocyclic aromatic amines ( HCAs ) have proved to be carcinogenic in both rodents and nonhuman primates .\n\nTwo different metabolic pathways are suggested for the metabolic activation of HCA .\n\nThe hepatic pathway proceeds via a two-step process involving N-hydroxylation by cytochrome P4501A2 and subsequent O-acetylation by N-acetyltransferase-2 .\n\nAn alternative pathway may be of particular interest in extrahepatic tissues and proceeds via one-electron oxidation catalyzed by prostaglandin H synthase ( PHS ) , rendering free-radical metabolites .\n\nIn this study , we investigated the metabolic activation of two HCAs , 2-amino-3-methylimidazo[4,5-f]quinoline ( IQ ) and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine ( PhIP ) , by two different enzyme systems in vitro , generating different primary and secondary reactive metabolites .\n\nRat liver S9 mix and PHS were used as the activating system and represent the hepatic and extrahepatic pathways , respectively .\n\nElectron-spin resonance spectroscopy showed that both IQ and PhIP exerted inhibiting effects on PHS-mediated formation of hydroxyl radicals during the conversion of arachidonic acid to prostaglandins .\n\nEvidence for the formation of HCA free radicals was presented in an indirect way by the formation of glutathione-derived thiyl radicals , with purified PHS as the activating system .\n\nActivation by S9 mix did not result in the formation of detectable radical metabolites , showing that the two metabolic routes primarily led to the formation of different metabolites .\n\nIn all electron-spin resonance experiments , IQ appeared to be more effective than PhIP .\n\nIn contrasts , DNA adduct analysis by means of ( 32)P-postlabeling showed similar adduct patterns for S9 and PHS in single-stranded and double-stranded salmon testes DNA after incubation with PhIP , indicating the ultimate formation of a common reactive intermediate .\n\nFor IQ , activation by PHS led to an additional adduct spot that was not present after S9 activation .\n\nFurthermore , activation of IQ resulted in higher adduct levels compared with PhIP for both activation pathways .\n\nOverall , adduct levels were higher in single-stranded DNA than double-stranded DNA .\n\nOur results showed that the hepatic and extrahepatic pathways resulted in different primary metabolites , while the ultimate formation of a similar reactive intermediate for PhIP , possibly an arylnitrenium ion , suggested that both pathways could play an important role in the onset of carcinogenesis .", "output": "Genomic instability and mutation" }, { "input": "The Msh2 DNA mismatch repair gene is one of five genes implicated in the pathogenesis of hereditary nonpolyposis colorectal cancer ( HNPCC ) .\n\nTo address the possible mechanisms of the site-specific occurrence of HNPCC , the effect of Msh2 deficiency on mutations in different parts of the colon was investigated using the BC-1(lacI)/Msh2 double transgenic mouse .\n\nCompared to the Msh2(+/+) mice , Msh2(-/-) mice had an 8-9-fold increase of mutation frequency ( MF ) in the lacI gene from the cecum and the proximal and distal colon .\n\nThe mutational spectra were also significantly different between Msh2(+/+) and Msh2(-/-) mice , with a significant increase in the frequency of -1 frameshifts and G:C-->A:T base substitutions in the repair-deficient mice .\n\nHowever , in spite of the site-specific predisposition of HNPCC in humans , we found no significant difference in the MF or mutation spectrum between the three parts of the colon in Msh2(+/+) , Msh2(+/-) , or Msh2(-/-) mice .\n\nIn addition , 11 independent mutants harboring complex mutations within the lacI gene were recovered in the Msh2(-/-) mice .\n\nInterestingly , while the Msh2(+/-) mice displayed an overall MF similar to that observed in the wild-type mice , sequencing revealed a significantly different mutational spectrum between Msh2(+/+) and Msh2(+/-) mice , mainly characterized by an increase in -1 frameshifts .\n\nDue to the prevalence of frameshift mutations in HNPCC patients , this haploinsufficiency effect of the Msh2 gene in safeguarding genomic integrity may have important implications for human carrier status .", "output": "Genomic instability and mutation" }, { "input": "Cis-diamminedichloroplatinum ( II ) ( cisplatin ) is a well characterized antitumor drug used for the treatment of a variety of human cancers .\n\nThe cytotoxicity of cisplatin is mainly mediated through the formation of DNA adducts , which are also believed to be responsible for the secondary malignancies produced by the drug .\n\nThe aim of this study was to determine the in vivo mutagenic activity of cisplatin in the lacZ plasmid-based transgenic mouse model .\n\nThe mutant frequency ( MF ) and the spectrum of mutations induced by cisplatin in the mouse liver were analyzed and compared to controls .\n\nThe mean MF in the lacZ gene was increased 2-fold in mice treated with a single 6 mg/kg body weight dose of cisplatin and sacrificed after 17 and 28 days ( P = 0.001 and P < 0.0001 ) .\n\nRestriction analysis and sequencing of mutant DNA showed that cisplatin was able to induce both large deletions and point mutations .\n\nA specific profile of base substitution and frameshift mutations was identified in treated mice , consisting primarily of G:C-->A:T transitions at GpG and ApG sites , the preferential DNA binding sites of cisplatin , and single basepair deletions/insertions .\n\nThe present results provide the first evidence that cisplatin has mutagenic activity in vivo and induces a characteristic pattern of mutations in the mouse liver .\n\nThis mutagenicity may be responsible for its tumorigenic activity .", "output": "Genomic instability and mutation" }, { "input": "Epidemiological data and animal models have provided evidence that nonsteroidal antiinflammatory drugs ( NSAIDs ) have an anticancer effect .\n\nHowever , the molecular mechanisms underlying these antineoplastic effects are not well understood .\n\nWe described previously that expression levels of the chemokine receptor , CCR5 , and the beta2-integrin , Mac-1 , were down-regulated on primary monocytes after incubation in supernatants from human carcinoma cell lines , and that this down-regulation resulted in impaired monocyte function with respect to migration and adhesion .\n\nWe now demonstrate that these impairments are also present in vivo .\n\nMonocytes from cancer patients displayed significantly reduced CCR5 levels and migration capacities in comparison to cells from healthy donors .\n\nBecause migration is necessary for the antitumor activity of monocytes/macrophages , these deficits may contribute to the suppressed immune system seen in cancer patients .\n\nIn a clinical study , we analyzed the effect of a selective COX-2 inhibitor , Rofecoxib , on the migration of monocytes derived from cancer patients .\n\nThe results revealed significant improvement in migration equal to those levels seen in healthy donors .\n\nWe conclude that in patients with cancer , the intake of Rofecoxib for 3 wk leads to significant restoration of monocyte function .\n\nThese data may , at least in part , help explain the anticancer effects of NSAIDs .", "output": "Avoiding immune destruction" }, { "input": "A6 is an eight amino acid peptide derived from the non-receptor binding region of urokinase plasminogen activator ( uPA ) , which interferes with the uPA/uPA receptor system .\n\nA6 has been synthesized as a potential anti-angiogenic , anti-cancer agent .\n\nThe current study has investigated the potential therapeutic activity of A6 in the Lewis lung carcinoma ( 3LL ) model of pulmonary metastasis .\n\nA6 was found to have direct anti-tumor activity against established 3LL pulmonary metastases at a low tumor burden ( 10-20 colonies per lung ) and was therapeutic in combination with cyclophosphamide at high tumor burdens ( > 100 colonies per lung ) .\n\nMechanistic studies have revealed that A6 directly inhibits the invasion of 3LL cells through a Matrigel model basement membrane by 40-45% .\n\nMoreover , treatment with either A6 or doxorubicin resulted in thicker tubes in endothelial tube formation studies .\n\nOur results suggest that A6 , by virtue of its anti-invasive and anti-angiogenic properties , might work additively or synergistically with chemotherapeutic agents and thereby contribute to enhanced therapy of established 3LL cancer metastases .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Tumor-associated macrophages ( TAM ) have been shown to play an important role in tumor angiogenesis .\n\nThe purpose of this study was to determine whether monocyte recruitment , activation and differentiation mediated by monocyte chemotactic protein-1 ( MCP-1 ) and macrophage colony stimulating factor ( M-CSF ) modulate the expression of the angiogenic factor , Interleukin ( IL)-8 .\n\nIsolated human peripheral blood monocytes secreted low basal levels of IL-8 .\n\nIncubation of monocytes with M-CSF or MCP-1 resulted in an up-regulation of IL-8 mRNA and protein expression .\n\nThe differential expression of IL-8 by monocytes following MCP-1 and M-CSF treatments involved activation of the NFkB transcription factor .\n\nFurther activation with lipopolysaccharide ( LPS ) caused an increase in IL-8 secretion in monocytes but not in monocyte-derived macrophages ( MDM ) .\n\nMDM-conditioned media significantly up-regulated IL-8 expression in human malignant melanoma cells in vitro .\n\nIn summary , we demonstrated that MCP-1 and M-CSF , critical for monocyte recruitment , activation and differentiation , differentially regulate IL-8 expression and may play an important role in monocyte/macrophage-mediated tumor angiogenesis .", "output": "Inducing angiogenesis" }, { "input": "Recent studies have shown that the antiestrogen tamoxifen ( TAM ) can be used in the treatment of malignant neoplasms other than breast cancer .\n\nIn the present study , we investigated the expression of estrogen receptor ( ER ) in six malignant rhabdoid tumor ( MRT ) cell lines .\n\nAlterations in MRT cell growth in response to estrogen or antiestrogens ( 4-hydroxytamoxifen ( 4-OHT ) , TAM , and ICI 182 780 ) were also investigated .\n\nRT-PCR and western blotting showed that ER-alpha was expressed in three of the six MRT cell lines .\n\nWhile 17-beta-estradiol ( E2 ) did not significantly alter MRT cell line proliferation , the hydroxylated tamoxifen metabolite 4-OHT significantly inhibited the growth of all 6 MRT cell lines .\n\nHowever , the steroidal antiestrogen ICI 182 780 did not alter the proliferation of any of the MRT cell lines. 4-OHT induced apoptosis in both ER-alpha-negative and ER-alpha-positive MRT cell lines , as assessed by nuclear morphology and DNA fragmentation .\n\nNeither growth inhibition nor induction of apoptosis due to 4-OHT was blocked by the addition of excess E2 .\n\nOur data suggested that 4-OHT induced cytotoxic effects against MRT cells , and that these effects were independent of ER expression .", "output": "Sustaining proliferative signaling" }, { "input": "The colon carcinoma cell line CC531 is metastatic to liver after splenic injection in syngeneic rats .\n\nAfter repeated in vivo passages , a subline was selected that produced liver metastases at a considerably higher rate than the original cell line .\n\nThese cells were characterized by increased intracellular glutathione , proliferation and ability to restore glutathione after exposure to oxidative stress , thus indicating an elevated resistance to oxidative stress .\n\nFurthermore , the increased metastatic ability was also accompanied by increased proliferation rate , adhesion to extracellular matrix proteins and endothelial cells , and secretion of a 60 kD matrix metalloproteinase .\n\nWhen cultured in vitro for a prolonged time ( more than 30 trypsinizations ) , the cells showed a reduced in vivo metastatic ability , reduced secretion of three metalloproteinases including the 60 kD proteinase , and reduced intracellular glutathione .\n\nThese results indicate that metastatic ability can be influenced through several adaptive mechanisms , and that the cell's ability to resist oxidative stress and maintain intracellular glutathione are of central importance .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "The Sonic hedgehog ( SHH ) pathway is implicated in the etiology of the most common human cancer in Caucasians , the basal cell carcinoma ( BCC ) .\n\nMutations in the receptor of SHH , the patched gene , have been characterized in sporadic BCCs as well as those from patients with the rare genetic syndromes nevoid BCC and xeroderma pigmentosum ( XP ) .\n\nTo elucidate the role of UV in the deregulation of the SHH pathway , we analyzed for alterations of smoothened , a transmembrane signaling component regulated by patched , in BCCs and squamous cell carcinomas from UV hypersensitive XP patients .\n\nWe find UV-specific smoothened mutations in 30% of XP BCCs , three times higher than those in sporadic Caucasian BCCs , confirming the high rate of UV-induced mutations in DNA repair-deficient XP patients .\n\nNo alteration was found in XP squamous cell carcinomas , indicating the involvement of smoothened specifically in the development of BCC .", "output": "Genomic instability and mutation" }, { "input": "Neuroblastoma ( NB ) , a common pediatric neoplasm , consists of two main cell populations : neuroblastic/ganglionic cells and Schwann cells .\n\nNB tumors with abundant Schwannian stroma display a more benign clinical behavior than stroma-poor tumors .\n\nRecent studies suggest that Schwann cells influence NB tumor growth via secreted factors that induce differentiation , suppress proliferation , and inhibit angiogenesis .\n\nTwo angiogenesis inhibitors , pigment epithelium-derived factor and tissue inhibitor of metalloproteinase-2 , have been detected in Schwann cell secretions .\n\nHere , we isolated another Schwann cell-derived secreted inhibitor of angiogenesis , a 43-kDa protein identified as SPARC ( secreted protein acidic and rich in cysteine ) , an extracellular matrix protein .\n\nWe found SPARC to be critical for the antiangiogenic phenotype of cultured Schwann cells .\n\nWe also show that purified SPARC potently inhibits angiogenesis and significantly impairs NB tumor growth in vivo .\n\nSPARC may be an effective candidate for the treatment of children with clinically aggressive , Schwannian stroma-poor NB tumors .", "output": "Inducing angiogenesis" }, { "input": "OBJECTIVE To design and develop a novel , sensitive and versatile method for in vivo foot printing and studies of DNA damage , such as DNA adducts and strand breaks .\n\nMETHODS Starting with mammalian genomic DNA , single-stranded products were made by repeated primer extension , these products were ligated to a double-stranded linker having a randomized 3 ' overhang , and used for PCR .\n\nDNA breaks in p53 gene produced by restriction endonuclease AfaI were detected by using this new method followed by Southern hybridization with DIG-labeled probe .\n\nRESULTS This randomized terminal linker-dependent PCR ( RDPCR ) method could generate band signals many-fold stronger than conventional ligation-mediated PCR ( LMPCR ) , and it was more rapid , convenient and accurate than the terminal transferase-dependent PCR ( TDPCR ) .\n\nCONCLUSION DNA strand breakage can be detected sensitively in the gene level by RDPCR .\n\nAny lesion that blocks primer extension should be detectable .", "output": "Genomic instability and mutation" }, { "input": "DNA-protein cross-links ( DPCs ) are formed upon exposure to a variety of chemical and physical agents and pose a threat to genomic integrity .\n\nIn particular , acrolein and related aldehydes produce DPCs , although the chemical linkages for such cross-links have not been identified .\n\nHere , we report that oligodeoxynucleotides containing 1,N(2)-deoxyguanosine adducts of acrolein , crotonaldehyde , and trans-4-hydroxynonenal can form cross-links with the tetrapeptide Lys-Trp-Lys-Lys .\n\nWe concluded that complex formation is mediated by a Schiff base linkage because DNA-peptide complexes were covalently trapped following reduction with sodium cyanoborohydride , and pre-reduction of adducted DNAs inhibited complex formation .\n\nA previous NMR study demonstrated that duplex DNA catalyzes ring opening for the acrolein-derived gamma-hydroxy-1,N(2)-propanodeoxyguanosine adduct to yield an aldehydic function ( de los Santos , C. , Zaliznyak , T. , and Johnson , F .\n\n( 2001 ) J. Biol .\n\nChem. 276 , 9077-9082 ) .\n\nConsistent with this earlier observation , the adducts under investigation were more reactive in duplex DNA than in single-stranded DNA , and we concluded that the ring-open aldehydic moiety is the induced tautomer in duplex DNA for adducts exhibiting high relative reactivity .\n\nAdducted DNA cross-linked to Arg-Trp-Arg-Arg and Lys-Trp-Lys-Lys with comparable efficiency , and N(alpha)-acetylation of peptides dramatically inhibited trapping ; thus , the reactive nucleophile is located at the N-terminal alpha-amine of the peptide .\n\nThese data suggest that Schiff base chemistry can mediate DPC formation in vivo following the formation of stable aldehyde-derived DNA adducts .", "output": "Genomic instability and mutation" }, { "input": "Recent studies indicate that cyclooxygenase-2 ( COX-2 ) is overexpressed in pancreatic adenocarcinoma and may play a critical role in this rapidly progressing form of cancer .\n\nA human pancreatic adenocarcinoma cell line , Mia PaCa-2 , was incubated for 18 hours with 5 micromol/L of rofecoxib ( Vioxx ) , a selective COX-2 inhibitor .\n\nTotal RNA was isolated and gene expression analyzed by DNA microarray chips .\n\nIn a separate experiment , athymic mice were orthotopically injected with 7.5 x 10(5) Mia PaCa-2 cells through a minilaparotomy .\n\nAfter 1 month , laparotomy was repeated to measure tumor size , and mice were randomized to receive reformulated rodent chow containing either 12.5 mg/kg/day of rofecoxib or no drug for 21 days .\n\nTumor growth was assessed by comparing volume before and after treatment .\n\nIn vitro , rofecoxib decreased gene expression of cyclin D1/PRAD1 , a key component of cell cycle progression , while increasing expression of several cell cycle arrest genes , including p21/WAF1 , p33/ING , GADD34 , and GADD45 ( P < 0.05 ) .\n\nIn vivo , tumor growth was significantly reduced in treated vs. control mice ( P < 0.05 ) .\n\nNo systemic toxicity was observed in mice receiving rofecoxib .\n\nThese data suggest that rofecoxib slows the growth of human pancreatic cancer through changes in gene expression that favor cell cycle arrest .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The thyroid hormone ( T3 ) blocks proliferation and induces differentiation of neuroblastoma N2a-beta cells that overexpress the beta 1 isoform of the T3 receptor .\n\nAn element in the region responsible for premature termination of transcription mediates a rapid repression of c-myc gene expression by T3 .\n\nThe hormone also causes a decrease of cyclin D1 gene transcription , and is able to antagonize the activation of the cyclin D1 promoter by Ras .\n\nIn addition , a strong and sustained increase of the levels of the cyclin kinase inhibitor ( CKI ) p27(Kip1) are found in T3-treated cells .\n\nThe increased levels of p27(Kip1) lead to a marked inhibition of the kinase activity of the cyclin-CDK2 complexes .\n\nAs a consequence of these changes , retinoblastoma proteins are hypophosphorylated in T3-treated N2a-beta cells , and progression through the restriction point in the cell cycle is blocked .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Several members of the phosphatidylinositol 3-kinase family play key roles in recognising and responding to damage in DNA , induced by a variety of chemicals and other agents .\n\nOne of these , ATM , the product of the gene mutated in the human genetic disorder ataxia-telangiectasia ( A-T ) , recognises double strand breaks in DNA caused by ionizing radiation and radiomimetic chemicals .\n\nIn order to study DNA damage recognition and the abnormalities of genome instability and cancer predisposition that occur in A-T patients , we generated a mouse model expressing a mutant form of Atm corresponding to a common human mutation .\n\nIn this model , a 9 nucleotide in-frame deletion was introduced into the Atm gene and has been designated Atm-Delta SRI .\n\nThese animals had a longer lifespan than Atm gene disrupted mice ( Atm(-/-) ) and they developed less thymic lymphomas .\n\nA characteristic of the lymphomas appearing in Atm-Delta SRI mice was an increased rate of apoptosis compared to the corresponding tumours in Atm(-/-) mice .\n\nIncreased expression of FasL in these tumours may account for the higher levels of apoptosis .\n\nThese results demonstrate that expression of mutant Atm in mice gives rise to phenotypic differences compared to Atm(-/-) mice and has implications for heterogeneity described in the human syndrome .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Little is known about the requirements for human T-cell leukemia virus type I ( HTLV-I ) entry , including the identity of the cellular receptor(s) .\n\nRecently , we have generated an HTLV-I surface glycoprotein ( SU ) immunoadhesin , HTSU-IgG , which binds specifically to cell-surface protein(s) critical for HTLV-I-mediated entry in cell lines .\n\nHere , expression of the HTLV-I SU binding protein on primary cells of the immune system was examined .\n\nThe immunoadhesin specifically bound to adult T cells , B cells , NK cells , and macrophages .\n\nCell stimulation dramatically increased the amount of binding , with the highest levels of binding on CD4(+) and CD8(+) T cells .\n\nNaive ( CD45RA(high) , CD62L(high) ) CD4(+) T cells derived from cord blood cells , in contrast to other primary cells and all cell lines examined , bound no detectable HTLV-I SU .\n\nHowever , following stimulation , the level of HTSU-IgG binding was rapidly induced ( fewer than 6 hours ) , reaching the level of binding seen on adult CD4(+) T cells by 72 hours .\n\nIn contrast to HTLV-I virions , the soluble HTSU-IgG did not effect T-cell activation or proliferation .\n\nWhen incubated with human peripheral blood mononuclear cells in a mixed leukocyte reaction , HTSU-IgG inhibited proliferation at less than 1 ng/mL .\n\nThese results indicate that cell-surface expression of the HTLV SU binding protein is up-regulated during in vitro activation and suggest a role for the HTLV-I SU binding proteins in the immunobiology of CD4(+) T cells .", "output": "Avoiding immune destruction" }, { "input": "Human esophageal adenocarcinoma ( EAC ) develops in a sequence from gastroesophageal reflux disease ( GERD ) , columnar-lined esophagus ( CLE ) , dysplasia , and eventually to EAC .\n\nWe established a rat surgical EAC model with esophagogastroduodenal anastomosis ( EGDA ) to mimic the staged process of esophageal adenocarcinogenesis .\n\nProfiling of the AA metabolites with mass spectrometry showed that prostaglandin E2 ( PGE2 ) , leukotriene B4 ( LTB4 ) , 15-hydroeicosatetraenoic acid ( HETE ) , 12-HETE , 8-HETE and 5-HETE all increased at the esophagoduodenal junction after EGDA as compared with the proximal esophagus , with PGE2 as the major metabolite .\n\nConsistent with this profile , cyclooxygenase 2 ( Cox2 ) was overexpressed in the basal cell layer of esophageal squamous epithelium , CLE cells and EAC tumor cells of the EGDA rats , as compared with the normal esophageal epithelium .\n\nSulindac ( a Cox inhibitor ) , nordihydroguaiaretic acid ( NDGA , a lipoxygenase inhibitor ) and alpha-difluoromethylornithine ( DFMO , an ornithine decarboxylase inhibitor ) were tested for their possible inhibitory actions against the formation of EAC in the rat EGDA model .\n\nIn a short-term study ( for 4 weeks after surgery ) , dietary administration of both sulindac ( 300 and 600 p.p.m. ) and NDGA ( 100 p.p.m. ) effectively reduced the EGDA-induced inflammation .\n\nIn a long-term chemoprevention study ( for 40 weeks after surgery ) , 300 p.p.m. sulindac , alone or in combination with 100 p.p.m .\n\nNDGA or 0.5% DFMO , decreased the tumor incidence from 57.7 to 26.9% , or 16.7 or 20% , respectively ( P < 0.05 ) .\n\nNDGA alone ( 100 and 200 p.p.m. ) slightly decreased the tumor incidence to 52.4 and 37% , respectively , although the difference was not statistically significant .\n\nDFMO alone did not show significant effects on tumor incidence .\n\nInhibition of tumor formation by sulindac was correlated with lowered levels of PGE2 .\n\nIn conclusion , sulindac exerted its chemopreventive effect against the formation of EAC in the rat EGDA model possibly through its inhibition of Cox .", "output": "Tumor promoting inflammation" }, { "input": "To examine the association of cell cycle regulatory gene inactivation with human cell immortalization , we determined the expression status of INK4a , Rb , and WAF1/ CIP1 , in eleven in vitro immortalized human cell lines , including fibroblasts and keratinocytes .\n\nTwo human papillomavirus type 16 E6 expressing cell lines with telomerase activity , including a fibroblast cell line and a keratinocyte cell line , expressed no detectable p16(INK4a) .\n\nThese cell lines had a hyperphosphorylated pRb and reduced expression of p21(WAF1/CIP1) .\n\nAll of seven fibroblast cell lines immortalized either spontaneously or by ( 60)Co , X-rays , 4-nitroquinoline 1-oxide or aflatoxin B(1) , maintaining their telomeres by the ALT ( alternative lengthening of telomeres ) pathway , displayed loss of expression of p16(INK4a) and hyperphosphorylation of pRb .\n\nLevels of p21(WAF1/CIP1) expression varied among the cell lines .\n\nTwo fibroblast cell lines that became immortalized following infection with a retrovirus vector encoding human telomerase catalytic subunit ( hTERT ) cDNA were also accompanied by inactivation of p16(INK4a) and pRb pathways .\n\nAcquisition of telomerase activity alone was not sufficient for immortalization of these cell lines .\n\nTaken together , all the cell lines including fibroblasts and keratinocytes , with either telomerase activity or the ALT pathway for telomere maintenance showed loss of expression of p16(INK4a) and hyperphosphorylation of pRb .\n\nThese demonstrate the association of inactivation of both p16(INK4a) and pRb with immortalization of human cells including fibroblasts and epithelial cells and telomerase-positive cells and ALT-positive cells .", "output": "Enabling replicative immortality" }, { "input": "BACKGROUND & OBJECTIVE There is little ideal predictor available on evaluating the lymph node metastatic potential of breast carcinoma .\n\nThis study was designed to determine the expression of gene products of E-cadherin ( epithelial ) , N-cadherin ( nerve ) , and matrix metalloproteinase-9 ( MMP-9 ) in breast carcinoma tissue and investigate their association with the invasion and metastasis of breast carcinoma .\n\nMETHODS The authors examined the expressions of E-cadherin , N-cadherin , and MMP-9 in 72 cases of breast carcinoma(39 cases with lymph node metastasis and 33 cases without lymph node metastasis ) by immunohistochemistry .\n\nMultivariable Cox proportional hazards model was used to analyze the patients ' prognosis .\n\nRESULTS The average ranks of E-cadherin in lymph node metastasis group and no lymph node metastasis group were 29.19 and 45.14 , respectively , with significant difference ( P < 0.001 ) .\n\nThe expression of E-cadherin was correlated inversely with the metastasis of breast carcinoma .\n\nThe average ranks of N-cadherin and MMP-9 were 40.04 and 42.97 in lymph node metastasis group , and 32.32 and 28.85 in no lymph node metastasis group , both with significant difference ( P < 0.05 ) , and these expressions were positively correlated with the lymph node metastasis of breast carcinoma .\n\nThe patients who had high expression of E-cadherin had a longer survival time .\n\nCONCLUSION Expression of E-cadherin , N-cadherin , and MMP-9 are associated strongly with lymph node metastasis of breast carcinoma .\n\nThese proteins are indicators of metastasis potential and prognosis of breast carcinoma .", "output": "Activating invasion and metastasis" }, { "input": "It was proved that nuclear factor-kappa B play an important role in the activation of immune cell , anti-virus , various stress reaction , and regulation of apoptosis , etc .\n\nThe objective of this study was to investigate the effect of NF-kappa B signal-transduction pathway on apoptosis of hepatic carcinoma cell line-7721 induced by TNF-alpha .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "BACKGROUND & OBJECTIVE Usually pituitary adenomas are histological benign and grow slowly , but a proportion of them will become locally aggressive , and develop into invasive pituitary adenomas .\n\nThe reasons for these differences in tumor behavior are poorly understood .\n\nPituitary adenomas are abounding blood vessels .\n\nAngiogenesis and tumor invasion both require degradation of the extracellular matrix components to allow cell migration .\n\nThe matrix metalloproteinases ( MMPs ) and their nature inhibitors-the tissue inhibitors of metalloproteinases ( TIMPs ) may play a central role in these processes .\n\nThe aggressive mechanism of pituitary adenomas was studied through investigating the expression of MMP-9 , MMP-2 , TIMP-1 , and TIMP-2 in both invasive and non-invasive adenomas .\n\nMETHODS Sixty-one surgical removed pituitary adenomas ( forty-nine cases invasive and twelve non-invasive adenomas ) were investigated .\n\nImmunohistochemistry staining ( SP method ) was used to detect the expression of MMP-9 , MMP-2 , TIMP-1 , and TIMP-2 in two groups .\n\nThe results were treated with semi-quantitative method and analyzed by using non-parameter rank sum test .\n\nRESULTS Immunohistochemical staining of tumor cells for MMP-9 , TIMP-1 , MMP-2 , and TIMP-2 were noted 95.9% ( 47/49 ) , 57.1% ( 28/49 ) , 75.5% ( 37/49 ) and 89.8% ( 44/49 ) in invasive adenomas , and 100% ( 12/12 ) , 91.7% ( 11/12 ) , 66.7% ( 8/12 ) , and 91.7% ( 11/12 ) in non-invasive adenomas , respectively .\n\nInvasive tumors were significantly less expressing TIMP-1 and TIMP-2 ( P < 0.05 ) .\n\nThere was no significant difference for MMP-9 or MMP-2 between invasive and non-invasive groups ( P > 0.05 ) .\n\nCONCLUSIONS TIMP-1 and TIMP-2 may play a key role in invasive pituitary adenomas to biological behavior .", "output": "Activating invasion and metastasis" }, { "input": "The xeroderma pigmentosum group C ( XPC ) protein specifically involved in genome-wide damage recognition for nucleotide excision repair ( NER ) was purified as a tight complex with HR23B , one of the two mammalian homologs of RAD23 in budding yeast .\n\nThis XPC-HR23B complex exhibits strong binding affinity for single-stranded DNA , as well as preferential binding to various types of damaged DNA .\n\nTo examine the structure-function relationship of XPC , a series of truncated mutant proteins were generated and assayed for various binding activities .\n\nThe two domains participating in binding to HR23B and damaged DNA , respectively , were mapped within the carboxy-terminal half of XPC , which also contains an evolutionary conserved amino acid sequence homologous to the yeast RAD4 protein .\n\nWe established that the carboxy-terminal 125 amino acids are dispensable for both HR23B and damaged DNA binding , while interactions with transcription factor IIH ( TFIIH ) are significantly impaired by truncation of this domain .\n\nFurthermore , deletion of the extreme carboxy-terminal domain totally abolished XPC activity in the cell-free NER reaction .\n\nThese results suggest that following initial damage recognition , the carboxy terminus of XPC may be essential for the recruitment of TFIIH , and that most truncation mutations identified in XP-C patients result in non-functional proteins .", "output": "Genomic instability and mutation" }, { "input": "Cancer-prone diseases ataxia-telangiectasia ( AT ) , Nijmegen breakage syndrome ( NBS ) and ataxia-telangiectasia-like disorder ( ATLD ) are defective in the repair of DNA double-stranded break ( DSB ) .\n\nOn the other hand , arsenic ( As ) has been reported to cause DSB and to be involved in the occurrence of skin , lung and bladder cancers .\n\nTo dissect the repair mechanism of As-induced DSB , wild type , AT and NBS cells were treated with sodium arsenite to study the complex formation and post-translational modification of Rad50/NBS1/Mre11 repair proteins .\n\nOur results showed that Mre11 went through cell cycle-dependent phosphorylation upon sodium arsenite treatment and this post-translational modification required NBS1 but not ATM .\n\nDefective As-induced Mre11 phosphorylation was rescued by reconstitution with full length NBS1 in NBS cells .\n\nAlthough As-induced Mre11 phosphorylation was not required for Rad50/NBS1/Mre11 complex formation , it might be required for the formation of Rad50/NBS1/Mre11 nuclear foci upon DNA damage .", "output": "Evading growth suppressors" }, { "input": "This study was set up to investigate the relationships between the formation and removal of DNA damage in form of 8-oxodeoxyguanosine ( 8-oxodG ) in neonatal ( day 16 of gestation ) as compared to adult rats .\n\nThe hypothesis addressed was whether the rapidly dividing foetal tissue has an enhanced requirement of DNA repair providing protection against potentially mutagenic DNA damages such as 8-oxodG .\n\nThe activity of the primary 8-oxodG-repair protein OGG1 was measured by a DNA incision assay and the expression of OGG1 mRNA was measured by Real-Time PCR normalised to 18S rRNA .\n\nThe tissue level of 8-oxodG was measured by HPLC-ECD .\n\nWe found a 2-3-fold increased incision activity in the foetal control tissue , together with a 3-15-fold increase in mRNA of OGG1 as compared to liver tissue from adult rats .\n\nThe levels of 8-oxodG in the foetal tissue were unaltered as compared to the adult groups .\n\nTo increase the levels of 8-oxodG , the rats received an injection ( i.p. ) of the hepatotoxin 2-nitropropane .\n\nThe compound induced significant levels of 8-oxodG in male rat livers 5h after the injection and in the foetuses 24h after the injection , while the female rats showed no increase in 8-oxodG .\n\nThe incision activity was slightly depressed in both male and female liver tissue and in the foetal tissue 5h after the injection , but significantly increased from 5 to 24h after the injection .\n\nHowever , it did not reach levels significantly above the control levels .\n\nIn conclusion , this study confirms that foetal tissue has increased levels of OGG1 mRNA and correspondingly an enhanced incision activity on an 8-oxodG substrate in a crude tissue extract .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "In both mitotic and meiotic processes , cellular surveillance of the integrity of genetic information transmission from parental cells to their subsequent generations is carried out by a network of proteins primarily involved in cell-cycle regulation , DNA replication , DNA repair , and chromosome segregation .\n\nWithin this context , the mammalian MRE11 represents an essential multifunctional protein that promotes repair of DNA double-strand breaks and plays a role in the signaling of DNA damage response .\n\nMutations in human hMRE11 gene could contribute to the rare \" AT-like \" disorder .\n\nHowever , at present time the functional roles of hMRE11 in these cellular processes are elusive .\n\nIn the current study , we provide evidence that hMRE11 interacts physically with the mismatch repair protein hMLH1 through yeast two-hybrid analysis .\n\nIn addition , we show that recombinant hMRE11 and hMLH1 proteins interact when these two proteins are coexpressed in bacterial cells , and both proteins can be co-immunoprecipitated from human cell extracts .\n\nFurthermore , hMRE11 and hMLH1 display similar expression patterns when examined with a human normal/tumor DNA array .\n\nTogether , these data suggest that hMRE11 and hMLH1 might act in a co-operative fashion during DNA damage detection , signaling , and repair .", "output": "Genomic instability and mutation" }, { "input": "Many different cellular pathways have evolved to protect the genome from the deleterious effects of DNA damage that result from exposure to chemical and physical agents .\n\nAmong these is a process called transcription-coupled repair ( TCR ) that catalyzes the removal of DNA lesions from the transcribed strand of expressed genes , often resulting in a preferential bias of damage clearance from this strand relative to its non-transcribed counterpart .\n\nLesions subject to this type of repair include cyclobutane pyrimidine dimers that are normally repaired by nucleotide excision repair ( NER ) and thymine glycols ( TGs ) that are removed primarily by base excision repair ( BER ) .\n\nWhile the mechanism underlying TCR is not completely clear , it is known that its facilitation requires proteins used by other repair pathways like NER .\n\nIt is also believed that the signal for TCR is the stalled RNA polymerase that results when DNA damage prevents its translocation during transcription elongation .\n\nWhile there is a clear role for some NER proteins in TCR , the involvement of BER proteins is less clear .\n\nTo explore this further , we studied the removal of 7-methylguanine ( 7MeG ) and 3-methyladenine ( 3MeA ) from the dihydrofolate reductase ( dhfr ) gene of murine cell lines that vary in their repair phenotypes. 7MeG and 3MeA constitute the two principal N-methylpurines formed in DNA following exposure to methylating agents .\n\nIn mammalian cells , alkyladenine DNA alkyladenine glycosylase ( Aag ) is the major enzyme required for the repair of these lesions via BER , and their removal from the total genome is quite rapid .\n\nThere is no observable TCR of these lesions in specific genes in DNA repair proficient cells ; however , it is possible that the rapid repair of these adducts by BER masks any TCR .\n\nThe repair of 3MeA and 7MeG was examined in cells lacking Aag , NER , or both Aag and NER to determine if rapid overall repair masks TCR .\n\nThe results show that both 3MeA and 7MeG are removed without strand bias from the dhfr gene of BER deficient ( Aag deficient ) and NER deficient murine cell lines .\n\nFurthermore , repair of 3MeA in this region is highly dependent on Aag , but repair of 7MeG is equally efficient in the repair proficient , BER deficient , and NER deficient cell lines .\n\nStrikingly , in the absence of both BER and NER , neither 7MeG nor 3MeA is repaired .\n\nThese results demonstrate that NER , but not TCR , contributes to the repair of 7MeG , and to a lesser extent 3MeA .", "output": "Genomic instability and mutation" }, { "input": "In Saccharomyces cerevisiae , Mre11p , Rad50p , and Xrs2p function as a multiprotein complex that has a central role in several DNA repair mechanisms .\n\nThough Mre11p has both single-stranded and double-stranded 3'-5 ' exonuclease activity in vitro , null mutants of MRE11 , RAD50 , and XRS2 exhibit reduced 5'-3 ' resection of HO-induced double-strand breaks ( DSBs ) in vivo .\n\nIn this study , we analyzed four mre11 mutants harboring changes in the N-terminus of Mre11p where the four phosphoesterase motifs specify the in vitro nuclease activities of Mre11p and its homologues .\n\nWe find that the 5'-3 ' resection defects in vivo do not correlate with several mitotic phenotypes : non-homologous end-joining ( NHEJ ) , telomere length maintenance , and adaptation to the DNA damage-inducible G2/M checkpoint .\n\nOverexpression of the 5'-3 ' exonuclease Exo1p in a mre11Delta strain partially increased 5'-3 ' resection and partially suppressed both methyl methanesulfonate ( MMS ) hypersensitivity and adaptation phenotypes , but did not affect telomere length or NHEJ .\n\nSurprisingly , the co-expression of two alleles , mre11-58S and mre11-N113S , each of which confers MMS hypersensitivity and short telomeres , can fully complement the MMS sensitivity and shortened telomere length of mre11Delta cells .\n\nWe propose that at least two separate activities associated with the N-terminus of Mre11p are required for its mitotic function .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Photodynamic therapy ( PDT ) may trigger apoptosis or necrosis in cancer cells .\n\nSeveral steps in the induction and execution of apoptosis require high amounts of adenosine-5'-triphosphate ( ATP ) .\n\nBecause the mitochondrial membrane potential ( delta psi ) decreases early in apoptosis , we raised the question about the mechanisms of maintaining a sufficiently high ATP level .\n\nWe therefore monitored delta psi and the intracellular ATP level of apoptotic human epidermoid carcinoma cells ( A431 ) after photodynamic treatment with aluminum ( III ) phthalocyanine tetrasulfonate .\n\nA maximum of caspase-3-like activity and nuclear fragmentation was found at fluences of about 4 J cm(-2) .\n\nUnder these conditions apoptotic cells reduced delta psi rapidly , while the ATP level remained high for 4-6 h after treatment for cells supplied with glucose .\n\nTo analyze the contribution of glycolysis to the energy supply during apoptosis , experiments were carried out with cells deprived of glucose .\n\nThese cells showed a rapid drop of ATP content and neither caspase activation nor nuclear fragmentation could be detected .\n\nWe conclude that the use of glucose as a source of ATP is obligatory for the execution of PDT-induced apoptosis .", "output": "Cellular energetics, Resisting cell death" }, { "input": "To explore the possibility of a new therapeutic strategy for leukemia by intervening in the DNA methylation to re-express p15 suppressor gene , methylation inhibitors , 5-Aza-2'-deoxycytidine ( 5-Aza-CdR ) and cell differentiation agent ( CDAII ) were used to treat myelogenous leukemia cell line KG1a in which p15 gene expression was suppressed due to DNA hypermethylation .\n\nThe biological characteristics of KG1a cells untreated or treated with the agents were investigated and analyzed using morphology , methylation specific-PCR ( MSP ) , ( 3)H-labeled microassay technique , restriction endonuclease reaction , flow cytometry and immunofluorescence methods .\n\nThe results indicated that both agents showed concentration-dependent and time-dependent inhibition of cell proliferation. 5-Aza-CdR and CDAII induced apoptosis and cell differentiation with G(2) and G(0)/G(1) arrest respectively .\n\nFurthermore , DNA methyltransferase activity and level of methylation in genomic DNA were decreased and p15 protein was re-expressed partially .\n\nIt is concluded that it is possible to treat leukemia by intervening in the DNA methylation using methyltransferase inhibitors and it is worth to make a thorough study on mechanism of the new strategy .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "The purpose of this investigation was to study the variation of p73 gene expression in the apoptotic process of acute myeloid leukemia ( AML ) cell line U937 induced by methotrexate ( MTX ) .\n\nMorphological changes of apoptotic cells were observed with microscopy and Wright's + Giemsa staining .\n\nDNA ladder and cell cycle were examined by agarose gel electrophoresis and flow cytometry respectively .\n\nUsing semi-quantitive reverse transcription-polymerase chain reaction ( RT-PCR ) , the expression of p73 mRNA was examined .\n\nResults showed that MTX could induce U937 cell apoptosis effectively .\n\nCondensed nuclei , fragmentation of chromosome and DNA ladder were seen after 6 hour following treatment of MTX 5 micro mol/L .\n\nSub-G(1) peak and S + G(2)/M arrest were also determined by FCM , but the quantity of p73 expression was generally constant .\n\nIn conclusion , U937 cell apoptosis induced by MTX did not change p73 mRNA level .", "output": "Resisting cell death" }, { "input": "Recent studies have shown that the transcription factor , nuclear factor kappaB ( NF-kappaB ) , regulates critical survival pathways in a variety of different cell types , including human pancreatic cancer cells .\n\nThe activation of NF-kappaB is controlled by proteasome-mediated degradation of its endogenous polypeptide inhibitor , inhibitor of nuclear factor kappaBalpha .\n\nWe investigated the effects of PS-341 , a peptide boronate inhibitor of the proteasome in human pancreatic cancer cells in vitro and in vivo .\n\nComparison of PS-341's effects on the growth of eight different human pancreatic cancer cell lines revealed marked heterogeneity in drug responsiveness , ranging from highly resistant ( IC50 > 10 microM ; Panc-48 , HS766T , and Mia-PaCa-2 ) to extremely sensitive ( IC50 < 40 nM ; L3.6pl , Hpaf2 , and BxPC3 ) .\n\nHowever , these effects did not correlate with differential inhibition of NF-kappaB activation .\n\nDirect quantification of apoptosis revealed that PS-341's effects on cell growth largely correlated with sensitivity to programmed cell death .\n\nEvaluation of PS-341's effects on established orthotopic tumor xenografts demonstrated that biweekly intravenous administration of the maximum-tolerated dose of the drug ( 1 mg/kg ) led to significant reductions in the volumes of L3.6pl tumors but not Mia-PaCa-2 tumors .\n\nLaser scanning cytometer-mediated quantification of drug-induced apoptosis in the xenografts confirmed that PS-341 induced DNA fragmentation and activation of caspase-3 in L3.6pl tumors but not in Mia-PaCa-2 tumors .\n\nHowever , histological examination of drug-treated tumors revealed extensive central necrosis and reductions in microvessel density and VEGF expression in both tumor types .\n\nTaken together , our results demonstrate that PS-341 inhibits the growth of human pancreatic tumors via direct effects on tumor cells and indirect effects on the tumor vasculature .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "Free radical-induced cellular stress contributes to cancer during chronic inflammation .\n\nHere , we investigated mechanisms of p53 activation by the free radical , NO .\n\nNO from donor drugs induced both ataxia-telangiectasia mutated ( ATM)- and ataxia-telangiectasia mutated and Rad3-related-dependent p53 posttranslational modifications , leading to an increase in p53 transcriptional targets and a G(2)M cell cycle checkpoint .\n\nSuch modifications were also identified in cells cocultured with NO-releasing macrophages .\n\nIn noncancerous colon tissues from patients with ulcerative colitis ( a cancer-prone chronic inflammatory disease ) , inducible NO synthase protein levels were positively correlated with p53 serine 15 phosphorylation levels .\n\nImmunostaining of HDM-2 and p21(WAF1) was consistent with transcriptionally active p53 .\n\nOur study highlights a pivotal role of NO in the induction of cellular stress and the activation of a p53 response pathway during chronic inflammation .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Evading growth suppressors" }, { "input": "Adenoid cystic carcinoma ( ACC ) of the salivary glands is a highly infiltrative malignant tumour with a tendency for lung metastasis .\n\nGene therapy could be a potentially effective therapy for ACC and its metastasis .\n\nThe aims of the study were : To transduce interleukin-2 ( IL-2 ) gene into an ACC cell line with predisposition for lung metastasis ( ACC-M ) ; to compare the bioactivity of the gene-transduced cells and the parent cell line in vitro and in vivo .\n\nThe IL-2 gene was transduced via a bicistronic retroviral vector into the ACC-M cells .\n\nThe growth rate and DNA cell cycles of the parent ACC-M , the control viral vector AmGCEN , and the gene transduced AmIL-2 cell cultures were compared quantitatively and by flow cytometry , respectively .\n\nThe tumourigenic ability of the three cell lines was verified by inoculation in athymic nude mice .\n\nThe tumours developed were extracted and compared quantitatively and histologically .\n\nThere was no difference in the growth rate and the DNA count between the ACC-M , AmGCEN , and AmIL-2 cell cultures .\n\nIn the animal experiment , both the ACC-M and AmGCEN cells stimulated lung metastasis in all the mice , whereas there was no tumour found in the 1 x 10(6) AmIL-2 cells inoculation .\n\nOn 3 x 10(6) AmIL-2 cells stimulation , three out of six mice developed tumours but the mass and volume of the tumours were smaller than the other two groups .\n\nUnder light microscopy , the ACC-M tumours were mainly poorly differentiated with minimal cellular matrix , whereas the AmIL-2 tumours were well differentiated with ample matrix .\n\nThe transduction of IL-2 gene can reduce the tumourigenicity of ACC-M cells and induces tumour cell differentiation in mice .\n\nThe IL-2 gene can be a potential effective gene for the treatment of adenoid cystic carcinoma of salivary glands and its lung metastasis .", "output": "Activating invasion and metastasis" }, { "input": "DNA mismatch repair ( MMR ) is the process by which incorrectly paired DNA nucleotides are recognized and repaired .\n\nA germline mutation in one of the genes involved in the process may be responsible for a dominantly inherited cancer syndrome , hereditary nonpolyposis colon cancer .\n\nCancer progression in predisposed individuals results from the somatic inactivation of the normal copy of the MMR gene , leading to a mutator phenotype affecting preferentially repeat sequences ( microsatellite instability , MSI ) .\n\nRecently , we identified children with a constitutional deficiency of MMR activity attributable to a mutation in the h MLH1 gene .\n\nThese children exhibited a constitutional genetic instability associated with clinical features of de novo neurofibromatosis type 1 ( NF1 ) and early onset of extracolonic cancer .\n\nBased on these observations , we hypothesized that somatic NF1 gene mutation was a frequent and possibly early event in MMR-deficient cells .\n\nTo test this hypothesis , we screened for NF1 mutations in cancer cells .\n\nGenetic alterations were identified in five out of ten tumor cell lines with MSI , whereas five MMR-proficient tumor cell lines expressed a wild-type NF1 gene .\n\nSomatic NF1 mutations were also detected in two primary tumors exhibiting an MSI phenotype .\n\nFinally , a 35-bp deletion in the murine Nf1 coding region was identified in mlh1-/- mouse embryonic fibroblasts .\n\nThese observations demonstrate that the NF1 gene is a mutational target of MMR deficiency and suggest that its inactivation is an important step of the malignant progression of MMR-deficient cells .", "output": "Genomic instability and mutation" }, { "input": "IL-16 is a ligand and chemotactic factor for CD4+ T cells .\n\nIL-16 inhibits the CD3 mediated lymphocyte activation and proliferation .\n\nThe effects of IL-16 on the target cells are dependent on the cell type , the presence of co-activators etc .\n\nTo understand the regulation function and mechanism of IL-16 on target cells , we used a 130 a.a. recombinant IL-16 to study its effects on the growth of Jurkat T leukemia cells in vitro .\n\nWe found that the rIL-16 stimulated the proliferation of Jurkat cells at low dose ( 10(-9)M ) , but inhibited the growth of the cells at higher concentration ( 10(-5)M ) .\n\nResults showed that 10(-5) M of rIL-16 treatment induced an enhanced apoptosis in Jurkat cells .\n\nThe treatment blocked the expression of FasL , but up-regulated the c-myc and Bid expression in the cells .\n\nPre-treatment of PKC inhibitor or MEK1 inhibitor markedly increased or decreased the rIL-16 induced growth-inhibiting effects on Jurkat cells , respectively .\n\nThe results suggested that the rIL-16 might be a regulator for the growth or apoptosis of Jurkat cells at a dose-dependent manner .\n\nThe growth-inhibiting effects of rIL-16 might be Fas/FasL independent , but , associated with the activation of PKC , up-regulated expression of c-Myc and Bid , and the participation of the ERK signal pathway in Jurkat cells .", "output": "Resisting cell death" }, { "input": "Like all cancers , breast cancer is considered to result in part from the accumulation of multiple genetic alterations leading to oncogene overexpression and tumor suppressor loss .\n\nMore recently , CpG island hypermethylation is known to be associated with gene silencing in cancer , and these silenced genes can be reactivated by 5-aza-2'-deoxycytidine ( 5-Aza-CdR ) .\n\nRetionoic acid receptor beta 2 gene is a tumor suppressor gene and the chemopreventive effects of retinoids are due to induction of RAR beta 2 .\n\nIn this study , the effect of 5-Aza-CdR RAR beta 2 restoration was investigated in the MRK-nu-1 human female breast cancer cell line .\n\nChanges of the RAR beta 2 methylation status were assessed by methylation-specific PCR .\n\nReverse transcription PCR was used to evaluate RARb beta 2 restoration .\n\nCell cycling and growth inhibition were studied using flow cytometric analysis of DNA content and CellTiter 96 AQueous non-radioactive cell proliferation assay , respectively. 5-Aza-CdR treatment resulted in complete demethylation of the RAR beta 2 gene .\n\nRAR beta 2 restoration was accompanied by cell cycle arrest ( increase in the G0/G1- and decrease in the S- and G2/M-phases ) and time-dependent growth inhibition .\n\nIn conclusion , RAR beta 2 can be activated in vitro by 5-Aza-CdR , which may be one of the mechanisms for the tumor cell growth inhibition by 5-Aza-CdR .", "output": "Evading growth suppressors" }, { "input": "The vinorelbine sensitivity of eight recently established head and neck squamous cell carcinoma ( SCC ) cell lines was tested using the 96-well plate clonogenic assay .\n\nThe chemosensitivity of these head and neck SCC cell lines to vinorelbine expressed as IC50 , corresponding to the drug concentration causing 50% inhibition in clonogenic survival , varied between 0.6 and 1.0 nM .\n\nThe dose-dependent growth inhibition caused by vinorelbine was measured in three of these cell lines .\n\nA clear growth inhibition was observed at a concentration of 3 nM .\n\nThe same cell lines were studied with flow cytometry .\n\nWhen exposed to 3 nM and 5 nM vinorelbine , an accumulation of the cells in the G2/M-phase was observed in all cultures after 12 hours .\n\nThe morphological changes induced by 3 nM and 5 nM vinorelbine to the UT-SCC-33 cell line were analysed with time-lapse video microscopy .\n\nIn the cultures treated with 5 nM vinorelbine , the cells stayed mitotically arrested for 2-32 hours and thereafter died morphologically by apoptosis .\n\nThese results indicate that , in vitro , the head and neck SCC is consistently sensitive to vinorelbine , which blocks the cell cycle in G2/M , the most radiosensitive phase .\n\nThese encouraging results suggest that vinorelbine may potentially be used in conjunction with radiotherapy in the treatment of head and neck SCC .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "We have recently identified ICBP90 as being a protein able to bind in vitro a CCAAT box of the topoisomerase II alpha gene promoter .\n\nThe aim of the present work was to check whether ICBP90 is able to regulate in vivo topoisomerase II alpha expression in human lung fibroblasts under various proliferating conditions .\n\nTransient transfection experiments performed on moderately growing human lung fibroblasts ( 50% of confluence ) showed that overexpression of ICBP90 is associated with an elevation of topoisomerase II alpha expression and an increase of the cell proliferation rate .\n\nIn highly proliferating human lung fibroblasts ( 20% confluence ) overexpression of ICBP90 had no effect .\n\nIn contrast , in non-proliferating fibroblasts ( 100% confluence ) overexpression of ICBP90 allowed recovery of topoisomerase II alpha expression levels with a concomitant overgrowth of confluent cell cultures .\n\nOur results show that ICBP90 regulates topoisomerase II alpha expression and is able to overcome cell contact inhibition signaling , suggesting that increased ICBP90 expression may be involved in carcinogenesis .", "output": "Evading growth suppressors" }, { "input": "The triterpenoid fraction ( 100 and 200 mg/kg ) of the fruit bodies of Ganoderma lucidum inhibited primary solid-tumor growth in the spleen , liver metastasis and secondary metastatic tumor growth in the liver in intrasplenic Lewis lung carcinoma ( LLC)-implanted mice .\n\nIn addition , the triterpenoid fraction ( 800 micrograms/mL ) inhibited angiogenesis induced by Matrigel ( a soluble basement membrane extract of the Engelbreth-Holm-Swam ( EHS ) tumor ) supplemented with vascular endothelial growth factor ( VEGF ) and heparin in an in vivo model .\n\nThis suggested that the antitumor and antimetastatic activities of the triterpenoid fraction of G. lucidum might be due to the inhibition of tumor-induced angiogenesis .\n\nNext , we attempted to isolate the active substance(s) using the in vivo assay system of Matrigel-induced angiogenesis .\n\nThe acidic fraction of the triterpenoid fraction inhibited the Matrigel-induced angiogenesis .\n\nCompound I was isolated from the acidic fraction as an active substance that inhibited the Martigel-induced angiogenesis .\n\nCompound I was identified as ganoderic acid F based on the data of IR , 1H- and 13C-NMR and MS analyses .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Despite experimental evidence that sulforaphane can exert chemopreventive effects , whether these effects are specific for neoplastic cells is not known .\n\nFollowing our previous demonstration that sulforaphane induces cell cycle arrest and apoptosis in human T lymphoblastoid Jurkat leukemia cells and increases p53 and bax protein expression , we tested sulforaphane on non-transformed phytohemagglutinin-stimulated human lymphocytes .\n\nHere , we demonstrate that sulforaphane arrested cell cycle progression in G , phase , through a decrease in the protein expression of cyclin D3 .\n\nMoreover , sulforaphane induced apoptosis ( and also necrosis ) , mediated by an increase in the expression of p53 .\n\nThese findings suggest that sulforaphane is a growth modulator for T cells .\n\nOur in vitro evidence that sulforaphane is active and even cytotoxic in normal as well as transformed lymphocytes raises important questions regarding its suitability for cancer chemoprevention .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Glycol ethers are known reproductive and developmental toxins in laboratory animals , but little is known about their genotoxic effects in humans .\n\nIn the current article , the authors tested the hypothesis that human in utero exposure to ethylene glycol monomethyl ether ( EGME ) is associated with the development of specific congenital anomalies and elevated levels of chromosome aberrations .\n\nThe authors conducted a clinical and cytogenetic evaluation of 41 offspring of 28 females occupationally exposed to EGME for an average duration of 4.6 yr .\n\nSix offspring of 5 women who were occupationally exposed to EGME during pregnancy exhibited characteristic dysmorphic features that were not observed in 35 offspring of 23 women who worked in the same facility , but who were not pregnant at the time of exposure .\n\nPersistent cytogenetic damage was observed exclusively in all 6 in-utero-exposed offspring , but not in their 12 match non-in-utero-exposed controls .\n\nThe study characterizes EGME as a human teratogen , as indicated by the prevalence of characteristic dysmorphic features and persistent cytogenetic damage in individuals exposed in utero to this chemical .", "output": "Genomic instability and mutation" }, { "input": "DNA polymerases beta ( pol beta ) and eta ( pol eta ) are the only two eukaryotic polymerases known to efficiently bypass cisplatin and oxaliplatin adducts in vitro .\n\nFrameshift errors are an important aspect of mutagenesis .\n\nWe have compared the types of frameshifts that occur during translesion synthesis past cisplatin and oxaliplatin adducts in vitro by pol beta and pol eta on a template containing multiple runs of nucleotides flanking a single platinum-GG adduct .\n\nTranslesion synthesis past platinum adducts by pol beta resulted in approximately 50% replication products containing single-base deletions .\n\nFor both adducts the majority of -1 frameshifts occurred in a TTT sequence 3-5 bp upstream of the DNA lesion .\n\nFor pol eta , all of the bypass products for both cisplatin and oxaliplatin adducts contained -1 frameshifts in the upstream TTT sequence and most of the products of replication on oxaliplatin-damaged templates had multiple replication errors , both frameshifts and misinsertions .\n\nIn addition , on platinated templates both polymerases generated replication products 4-8 bp shorter than the full-length products .\n\nThe majority of short cisplatin-induced products contained an internal deletion which included the adduct .\n\nIn contrast , the majority of oxaliplatin-induced short products contained a 3 ' terminal deletion .\n\nThe implications of these in vitro results for in vivo mutagenesis are discussed .", "output": "Genomic instability and mutation" }, { "input": "Mice defective in the mismatch repair ( MMR ) gene Msh2 manifest an enhanced predisposition to skin cancer associated with exposure to UVB radiation .\n\nThis predisposition is further heightened if the mice are additionally defective for the nucleotide excision repair gene Xpc .\n\nTo test the hypothesis that the predisposition of Msh2 mutant mice to skin cancer reflects a mutator phenotype associated with increased proliferation of skin cells following exposure to UV radiation , Msh2 mutant mice were exposed to the tumor promoter TPA .\n\nSuch mice showed a robust proliferative response in the skin , but did not manifest evidence of dysplasia or neoplasia .\n\nWe conclude that the predisposition of Msh2 mice to UVB radiation-induced skin cancer reflects an interaction between the processes of mismatch repair and some other excision repair mode , the exact nature of which remains to be established .", "output": "Genomic instability and mutation" }, { "input": "We have made xeroderma pigmentosum group A gene ( XPA)-knockout mice ( XPA(-/-) mice ) .\n\nThe XPA(-/-) mice had no detectable activity for nucleotide excision repair ( NER ) and showed a high incidence of UVB-induced skin tumorigenesis .\n\nWe have also found that cell lines derived from skin cancers in UVB-irradiated XPA(-/-) mice become tolerant to UV-irradiation and showed abnormal UV-induced cell cycle checkpoints and decreased mismatch repair ( MMR ) activity .\n\nThese results suggested that the MMR-downregulation may help cells escape killing by UV-irradiation and thus MMR-deficient clones are selected for during the tumorigenic transformation of XPA(-/-) cells .\n\nIn this report , we examined whether the incidence of UVB-induced skin tumorigenesis is enhanced in XPA(-/-)MSH2(-/-) , XPA(-/-) and MSH2(-/-) mice when compared with that in wild-type mice .\n\nOur results indicate that the MSH2-deficiency caused a high incidence of spontaneous and UVB-induced skin tumorigenesis and the XPA and MSH2 genes have additive roles in the UV-induced skin tumorigenesis .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "Arsenic trioxide ( ATO ) is a novel agent to treat acute promyelocytic leukemia ( APL ) .\n\nATO can degrade chimeric PML-RAR proteins and induce apoptosis in various cancer cells .\n\nHowever , its effects on primary hematopoietic CD34+ have not been examined .\n\nIn this study , we compared the effects of ATO on HL60 leukemic cells and primary umbilical cord blood ( UCB ) CD34+ cells .\n\nHL60 cells and UCB CD34+ cells were cultured with different concentrations of ATO for up to three weeks and examined for changes of cell cycle .\n\nWe found that ATO ( < or = 5 microM ) caused prolongation of G1/S and G2/M phase in a dose-dependent manner .\n\nThe percentage of cells in G2/M increased significantly ( from 8.6 to 53.8% ) .\n\nHigh-dose ATO ( > or = 25 microM ) caused non-specific cell death in HL60 cells without any changes in cell cycle .\n\nIn contrast to HL60 cells , UCB CD34+ cells were more resistant to high-dose ATO and most ATO-resistant CD34+ cells remained in G0/G1 phase .\n\nPrimary cells that were resistant to ATO were rich in CD34+ cells .\n\nWe further show that the ATO resistance was not related to the expression of P-glycoprotein ( MDR-1 ) .\n\nOur results suggest that the resistance to ATO in primitive UCB CD34+ cells is most likely related to its cell-cycle status .\n\nThese results could be useful to design treatments for non-APL malignancies and to enrich hematopoietic stem cells in clinically applicable settings .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Mast cells ( MC ) are critical for a number of pathological conditions , including acute and chronic inflammation and tumor angiogenesis .\n\nWe have previously demonstrated that in B-cell non-Hodgkin's lymphoma ( B-NHL ) angiogenesis is correlated with total methachromatic and tryptase-positive MC and that both counts increase in step with the increase in malignancy , whereas the role of MC in malignant lymph nodes is not fully clear .\n\nAn extensive ultrastructural study has been made of representative samples of 30 B-NHL and 10 benign lymphadenopathies .\n\nA heterogeneous population of MC characterized by the presence of granules with a semilunar aspect and containing scrolls was observed .\n\nThe former are the expression of a slow but progressive release of angiogenic factors due to chronic , progressive stimulation of MC degranulation , while the latter contain tryptase , an angiogenic factor .\n\nThese two ultrastructural data confirm the important role played by MC in the angiogenesis associated with progression in B-NHL .", "output": "Inducing angiogenesis" }, { "input": "Vanadium pentoxide , commercially the most important compound of vanadium , presents a potential occupational hazard during the cleaning of oil-fired boilers and furnaces , the handling of catalysts , and during the refining , processing , or burning of vanadium-rich mineral ores or fossil fuels .\n\nVanadium pentoxide was nominated for study by the National Cancer Institute as a representative of the metals class study .\n\nMale and female F344/N rats and B6C3F1 mice were exposed to vanadium pentoxide ( 99% pure ) by inhalation for 16 days , 14 weeks , or 2 years .\n\nGenetic toxicology studies were conducted in Salmonella typhimurium and mouse peripheral blood. 16-DAY STUDY IN RATS : Groups of five male and five female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0 , 2 , 4 , 8 , 16 , or 32 mg/m(3) by inhalation , 6 hours per day , 5 days per week for 16 days .\n\nThree males in the 32 mg/m(3) group died before the end of the study .\n\nMean body weights of males and females exposed to 8 mg/m(3) or greater were less than those of the chamber controls .\n\nClinical findings included rapid respiration and hypoactivity in rats exposed to 16 or 32 mg/m(3) .\n\nRelative lung weights of 4 mg/m(3) or greater males and 2 mg/m(3) or greater females were significantly greater than those of the chamber controls .\n\nLavage fluid analysis indicated an inflammatory response in the lung that was either directly mediated by vanadium pentoxide or was secondary to lung damage induced by vanadium pentoxide exposure. 16-DAY STUDY IN MICE : Groups of five male and five female mice were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0 , 2 , 4 , 8 , 16 , or 32 mg/m(3) by inhalation , 6 hours per day , 5 days per week for 16 days .\n\nAll males exposed to 32 mg/m(3) and one 8 mg/m(3) male died or were killed moribund before the end of the study .\n\nMean body weights of 16 mg/m(3) males and 8 mg/m(3) or greater females were significantly less than those of the chamber controls , and the 32 mg/m(3) females lost weight during the study .\n\nAbsolute and relative lung weights of 4 mg/m(3) or greater males and all exposed groups of females and liver weights of 16 mg/m(3) males were significantly greater than those of the chamber controls .\n\nThe mediastinal lymph nodes were enlarged in 4 , 8 , and 16 mg/m(3) males and females , and lymphoid hyperplasia was confirmed histologically .\n\nLavage fluid analysis indicated an inflammatory response in the lung that was either directly mediated by vanadium pentoxide or was secondary to lung damage induced by vanadium pentoxide exposure. 3-MONTH STUDY IN RATS : Groups of 10 male and 10 female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0 , 1 , 2 , 4 , 8 , or 16 mg/m(3) by inhalation , 6 hours per day , 5 days per week for 3 months .\n\nSeven males and three females exposed to 16 mg/m(3) died during the study .\n\nMean body weights were significantly less in males exposed to 4 mg/m(3) or greater and in females exposed to 16 mg/m(3) .\n\nAbnormal breathing , thinness , lethargy , abnormal posture , and ruffled fur were observed in rats exposed to 16 mg/m(3) .\n\nHematology results indicated that exposure of rats to vanadium pentoxide induced a microcytic erythrocytosis in males and females .\n\nAbsolute and relative lung weights were significantly greater for 4 mg/m(3) or greater males and females than for the chamber controls as were the relative lung weights of 2 mg/m(3) males .\n\nThe estrous cycle of females exposed to 8 mg/m(3) was significantly longer than that of the chamber control group , and the number of cycling females in the 16 mg/m(3) group was reduced .\n\nThe incidences of several nonneoplastic lesions of the lung and nose were significantly increased in males and females exposed to 2 mg/m(3) or greater .\n\nData from pulmonary function analyses indicated that a restrictive lung disease was present in male and female rats exposed to 4 mg/m(3) or greater , while an obstructive lung disease was present only in the 16 mg/m(3) groups. 3-MONTH STUDY IN MICE : Groups of 10 male and 10 female mice were exposed to par", "output": "Tumor promoting inflammation" }, { "input": "An efficent antitumor and antiviral cellular immune response requires optimal interferon-gamma ( IFN-gamma ) secretion and perforin expression in CD8(+) T cells .\n\nThe aim of this study was to define whether CD4(+) and CD8(+) T cells from patients with undifferentiated carcinoma of nasopharyngeal type ( UCNT ) , a tumor regularly associated with the Epstein-Barr virus ( EBV ) , have abnormal phenotype profiles , cytokine production , perforin and CD3-zeta expressions .\n\nOur data showed that CD4 and CD8 subset distribution was not grossly altered in the peripheral blood of UCNT patients , while tumor biopsies contained an increased proportion of CD8(+) T cells .\n\nThe analysis of the CD4(+) subset showed a defect in interleukin-2 ( IL-2 ) production and a moderate increase of IL-10 production , a situation consistent with a Th1/Th2 imbalance .\n\nWe have also demonstrated that CD8(+) lymphocytes from UCNT patients had a marked impairment of IFN-gamma secretion and perforin expression .\n\nThis impairment was not related to the presence of detectable EBV DNA in the plasma .\n\nIn UCNT patients , the blockade of the perforin pathway and of IFN-gamma production may constitute important mechanisms for immune escape by the tumor and for impaired control of EBV replication .", "output": "Avoiding immune destruction" }, { "input": "Natural killer ( NK ) and CD56(+) T cells are thought to play a central role in antitumour immunity .\n\nTheir cytolytic activities are controlled by a variety of receptors including CD94 and killer immunoglobulin-like receptors ( KIR ) , which bind to major histocompatibility complex ( MHC ) class I molecules on target cells and mediate cell activation or inhibition .\n\nWe have examined the numbers , phenotypes and antitumour cytotoxic functions of hepatic NK and CD56(+) T cells isolated from 22 patients with hepatic malignancy and 19 healthy donors .\n\nFlow cytometry revealed that NK cell numbers were increased among hepatic mononuclear cells in malignancy compared to histologically normal livers ( mean : 38% vs 27% ; P=0.03 ) , but CD56(+) T cell numbers were not ( 28% vs 27% ) .\n\nNK cells and CD56(+) T cells from tumour-bearing livers exhibited lymphokine-activated killing of K562 targets and T cell receptor-mediated lysis of P815 cells .\n\nThe expression of CD94 and the KIR isotypes CD158a , CD158b and KIR3DL1 by CD56(+) T cells and NK cells was significantly and consistently reduced in tumour-bearing livers compared to healthy livers ( P<0.05 in all cases ) .\n\nSimultaneous ligation of CD158a , CD158b and KIR3DL1 caused an overall partial inhibition of CD56(+) T cell cytotoxic activity , suggesting that the observed reductions in KIR(+) cell numbers in malignancy are likely to lead to enhanced cytotoxicity .\n\nOur results suggest that , while hepatic CD56(+) T cells are not expanded in malignancy , downregulation of KIR and CD94 expression may be a mechanism by which the hepatic immune system can be activated to facilitate tumour rejection .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND Hepatocellular carcinoma ( HCC ) is a leading cause of death worldwide .\n\nFrequent cytogenetic abnormalities that occur in HCC suggest that tumor-modifying genes ( oncogenes or tumor suppressors ) may be driving selection for amplification or deletion of these particular genetic regions .\n\nIn many cases , however , the gene(s) that drive the selection are unknown .\n\nAlthough techniques such as comparative genomic hybridization ( CGH ) have traditionally been used to identify cytogenetic aberrations , it might also be possible to identify them indirectly from gene-expression studies .\n\nA technique we have called comparative genomic microarray analysis ( CGMA ) predicts regions of cytogenetic change by searching for regional gene-expression biases .\n\nCGMA was applied to HCC gene-expression profiles to identify regions of frequent cytogenetic change and to identify genes whose expression is misregulated within these regions .\n\nRESULTS Using CGMA , 104 HCC gene-expression microarray profiles were analyzed .\n\nCGMA identified 13 regions of frequent cytogenetic change in the HCC samples .\n\nTen of these regions have been detected in previous CGH studies ( +lq , -4q , +6p , -8p , +8q , -13q , -16q , -17p , +17q , +20q ) .\n\nCGMA identified three additional regions that have not been previously identified by CGH ( +5q , +12q , +19p ) .\n\nGenes located in regions of frequent cytogenetic change were examined for changed expression in the HCC samples .\n\nCONCLUSIONS Our results suggest that CGMA predictions using gene-expression microarray datasets are a practical alternative to CGH profiling .\n\nIn addition , CGMA might be useful for identifying candidate genes within cytogenetically abnormal regions .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE Although the adrenal gland is a common site of extrahepatic metastasis from hepatocellular carcinoma ( HCC ) , there are no definitive guidelines for the treatment of adrenal metastasis .\n\nThis study examines the effectiveness of various treatments for this disease .\n\nMETHODS We retrospectively analyzed 20 patients treated for adrenal metastasis of HCC by adrenalectomy ( n = 13 ) , transarterial chemoembolization ( TACE ) , or percutaneous ethanol injection therapy ( PEIT ) ( n = 7 ) .\n\nRESULTS There were no significant differences in cumulative survival rates between patients given adrenalectomy and those given TACE or PEIT , either after completing treatment for primary HCC or after the first treatment for adrenal metastasis .\n\nSix of seven patients with tumor thrombi in the inferior vena cava ( IVC ) from adrenal metastasis underwent adrenalectomy combined with intracaval thrombectomy , five of whom survived for more than 1 year after surgery , and two of whom are still alive without any recurrence more than 3 years after surgery .\n\nPEIT showed good results for small adrenal metastasis .\n\nCONCLUSION These findings suggest that therapeutic modalities should be chosen according to the clinical features of each individual , including the size of the metastatic tumor , whether there is invasion into the IVC , the function of the remaining liver , and the existence of intra- and/or nonadrenal extrahepatic lesions .\n\nFurthermore , intracaval tumor thrombectomy could be indicated for patients with IVC thrombus if they are suitable candidates for surgery .", "output": "Activating invasion and metastasis" }, { "input": "Polo-like kinase 3 ( Plk3 , alternatively termed Prk ) is involved in the regulation of DNA damage checkpoint as well as in M-phase function .\n\nPlk3 physically interacts with p53 and phosphorylates this tumor suppressor protein on serine-20 , suggesting that the role of Plk3 in cell cycle progression is mediated , at least in part , through direct regulation of p53 .\n\nHere we show that Plk3 is rapidly activated by reactive oxygen species in normal diploid fibroblast cells ( WI-38 ) , correlating with a subsequent increase in p53 protein level .\n\nPlk3 physically interacts with Chk2 and the interaction is enhanced upon DNA damage .\n\nIn addition , Chk2 immunoprecipitated from cell lysates of Daudi ( which expressed little Plk3 ) is capable of stimulating the kinase activity of purified recombinant Plk3 in vitro , and this stimulation is more pronounced when Plk3 is supplemented with Chk2 immunoprecipitated from Daudi after DNA damage .\n\nFurthermore , ectopic expression Chk2 activates cellular Plk3 .\n\nTogether , our studies suggest Chk2 may mediate direct activation of Plk3 in response to genotoxic stresses .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "An in vitro angiogenesis system was designed for screening angiogenic agonists and antagonists .\n\nIn order to obtain large quantities of cells and reproducibility , human endothelial cells with extended life spans were developed by retroviral transfection .\n\nThe resulting cells grown in a serum-free medium containing endothelial cell growth supplement ( ECGS ) have a telomerase activity , extended life spans of at least 21 passages , and an endothelial cell phenotype ( diI-acetylated-LDL upake , factor VIII-related antigen , VEGFR-1 and R-2 , and tissue-type plasminogen activator ( tPA) ) that resembled that of unaltered primary endothelial cells .\n\nExceptions were ( i ) a higher expression of tPA , and ( ii ) a non-significant growth response to FGF-2 or VEGF stimulation .\n\nWithin three-dimensional fibrin gels , specific cell clones rapidly formed tubular structures in a more reproducible manner than those observed with low-passage primary cells .\n\nTube formation by primary endothelial cells and those with extended life spans was dependent upon FGF-2 and ECGS , respectively .\n\nBoth cell types produced FGF-2 and VEGF cytokines .\n\nIncreasing doses of suramin significantly decreased the size of microvessels formed by both cell lines .\n\nThese functional results indicate that a vascular matrix system containing human cells with extended life spans can be successfully utilized as an in vitro assay for antiangiogenic compounds .", "output": "Inducing angiogenesis" }, { "input": "Epidemiological studies suggest that dietary polyunsaturated fatty acids ( PUFA ) may influence breast cancer progression and prognosis .\n\nIn order to study potential mechanisms of action of fatty acid modulation of tumor growth , we studied , in vitro , the influence of n-3 and n-6 fatty acids on proliferation , cell cycle , differentiation and apoptosis of MCF-7 human breast cancer cells .\n\nBoth eicosapentaenoic acid ( EPA ) and docosahexaenoic acid ( DHA ) inhibited the MCF-7 cell growth by 30% and 54% , respectively , while linoleic acid ( LA ) had no effect and arachidonic acid ( AA ) inhibited the cell growth by 30% ( p < 0.05 ) .\n\nThe addition of vitamin E ( 10uM ) to cancer cells slightly restored cell growth .\n\nThe incubation of MCF-7 cells with PUFAs did not alter the cell cycle parameters or induce cell apoptosis .\n\nHowever , the growth inhibitory effects of EPA , DHA and AA were associated with cell differentiation as indicated by positive Oil-Red-O staining of the cells .\n\nLipid droplet accumulation was increased by 65% , 30% and 15% in the presence of DHA , EPA and AA , respectively ; ( p < 0.05 ) .\n\nThese observations suggest that fatty acids may influence cellular processes at a molecular level , capable of modulating breast cancer cell growth .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Chinese medicinal herbs are traditionally used to prevent and treat a variety of diseases , including cancer .\n\nThese herbal preparations are purported to have many biological effects including direct antiproliferative effects on cancer cells , anti-mutagenic activity , and stimulatory or suppressive effects on immune responses .\n\nThe present study investigates the effects of aqueous extracts from seventy-one Chinese medicinal herbs on the growth of five breast cancer cell lines ( SK-BR-3 , MCF7 , MDA-MB-231 , BT-474 and MCNeuA ) .\n\nTwenty-one percent ( 15 out of 71 ) of the extracts demonstrated greater than 50% growth inhibition on at least 4 of the 5 cell lines .\n\nDose-response curves were obtained for several of the most potent crude extracts and demonstrated IC50 values ranging from < 10 micrograms/ml to > 1 mg/ml .\n\nSix of seven herbs tested induced high molecular weight DNA fragmentation , an early marker of apoptosis , while one of these also induced low molecular weight DNA fragmentation .\n\nFlow cytometric analysis of breast cancer cells exposed to one of these herbs ( Rheum palmatum ) suggested that it arrests cells in the G2/M phase of the cell cycle .\n\nThese results indicate that many of the herbs used in traditional Chinese medicine for the treatment of cancer have significant growth inhibitory effects on breast cancer cells in vitro .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "The effects of glucocorticoid ( GC ) on the proliferation of Dunn Osteosarcoma ( OS ) cells were examined under in vitro culture conditions .\n\nDexamethasone ( Dex ) inhibited the proliferation of Dunn OS cells in a dose-dependent manner , while the addition of anti-GC , RU486 , to the culture medium in part recovered Dex-induced growth inhibition .\n\nThe number of maximum binding sites ( Bmax ) and the dissociation constant ( Kd ) value of glucocorticoid receptor ( GR ) in Dunn OS cells were 19,560 sites/cell and 5.2 +/- 0.8 nM , respectively .\n\nRU486 competed with labeled Dex against GR at a concentration of 10(-6) M. Western blot analysis of [ 3H]Dex-mesylate-labeled cell homogenate and immunohistochemical staining against GR further confirmed the presence of GR .\n\nDex treatment of Dunn OS cells resulted in apoptosis with the characteristic internucleosomal DNA cleavage shown by the DNA ladder pattern in agarose gel electrophoresis .\n\nThese data demonstrate that GC inhibits the proliferation of Dunn OS cells via GR , for which one possible mechanism in vitro is induction of apoptosis .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "We examined the inducibility of drug resistance ( MDR1 , MRP1 , LRP ) and protein kinase C ( PKC ) isozyme ( alpha , epsilon , eta , theta , tau , zeta ) corresponding genes in A2780 ovarian cancer cells after a 24-hour treatment with adriamycin ( ADR ) , camptothecin ( CAM ) , etoposide ( ETO ) or vincristine ( VCR ) .\n\nSublethal concentrations of drugs were used to exclude short-term effects caused by selection .\n\nCell cycle analysis was performed to identify possible correlation between resistance factors , PKC isozymes and proliferation .\n\nWe found a mostly combined induction of MDR1 , LRP , PKC tau and PKC zeta by CAM , ETO and VCR .\n\nPKC alpha , epsilon , eta and theta gene expression altered variably .\n\nCell cycle analysis showed that A2780 cells responded with a marked G2/M arrest after a 24-hour treatment with CAM , ETO and VCR but an association between the induction of PKC isozymes corresponding genes and proliferation was not seen .\n\nOur analysis points to a possible link between atypical PKC tau/PKC zeta and MDR1/LRP in cytostatic stress response of cancer cells .", "output": "Evading growth suppressors" }, { "input": "Recent reports provide evidence that some growth factors behave as inhibitors of the apoptosis of the endothelial cells , bringing forward the concept of vascular survival as a post-angiogenesis process .\n\nAt least two different vasculature development processes occur within a tumor : the angiogenic ( formation of new vessels ) and the vascular survival pathway , which is devoted to the preservation of the newly-formed vessels in layers that lose contact with the adjacent normal tissue .\n\nWe developed a method to assess these processes in tissue samples .\n\nWe noted that differences among tumors may exist not only in the tumor angiogenic activity ( TAA ) but also in the vascular survival ability ( VSA ) .\n\nOne third of the highly angiogenic breast cancer cases examined had a poor ability to maintain high vessel density in inner tumor areas .\n\nBoth parameters are independently related to prognosis , while VSA was directly related to tumor dimensions and node involvement .\n\nPatients with high TAA and VSA had a particularly poor prognosis .\n\nIt is suggested that although cancer angiogenic activity is important for the local invasion and dissemination into vessels and lymphatics , the VSA may be important for the effective formation of viable tumor foci in lymph nodes or distant organs .\n\nRecognition and quantification of the vascular survival ability in human tumors may significantly improve the prognostic value of the assessment of tumor vasculature , and may help to stratify patients for clinical trials with novel anti-angiogenic or angiotoxic drugs .\n\nElucidation of the pathways may provide additional targets for antiangiogenic therapy .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Steroid hormone receptors , including estrogen receptor-alpha ( ERalpha ) , are ligand-activated transcription factors , and hormone binding leads to depletion of receptor levels via preteasome-mediated degradation .\n\nNEDD8 ( neural precursor cell-expressed developmentally down-regulated ) is an ubiquitin-like protein essential for protein processing and cell cycle progression .\n\nWe recently demonstrated that ubiquitin-activating enzyme ( Uba)3 , the catalytic subunit of the NEDD8-activating enzyme , inhibits ERalpha transcriptional activity .\n\nHere we report that Uba3-mediated inhibition of ERalpha transactivation function is due to increased receptor protein turnover .\n\nCoexpression of Uba3 with ERalpha increased receptor degradation by the 26S proteasome .\n\nInhibition of NEDD8 activation and conjugation diminished polyubiquitination of ERalpha and blocked proteasome-mediated degradation of receptor protein .\n\nThe antiestrogen ICI 182,780 is known to induce ER degradation .\n\nIn human MCF7 breast cancer cells modified to contain a disrupted NEDD8 pathway , ICI 182,780 degradation of ERalpha was impaired , and the antiestrogen was ineffective at inhibiting cell proliferation .\n\nThis study provides the first evidence linking nuclear receptor degradation with the NEDD8 pathway and the ubiquitin-proteasome system , suggesting that the two pathways can act together to modulate ERalpha turnover and cellular responses to estrogens .\n\nBased on our observation that an intact NEDD8 pathway is essential for the antiproliferation activity of the ICI 182,780 in ERalpha positive breast cancer cells , we propose that disruptions in the NEDD8 pathway provide a mechanism by which breast cancer cells acquire antiestrogen resistance while retaining expression of ERalpha .", "output": "Sustaining proliferative signaling" }, { "input": "PURPOSE The difference in radiosensitivity between two isogenic tumour cell lines was evaluated to determine whether factors such as sublethal and potentially damage repair , DNA double-strand break repair and fidelity of repair can be related to differences in radiosensitivity .\n\nMATERIALS AND METHODS The cell lines used were the ovarian carcinoma A2780s and a radiation-resistant derivative A2780cp .\n\nRadiation response was measured in terms of cell survival , recovery of sublethal ( SLD ) and potentially lethal damage ( PLD ) , induction of and recovery of DNA strand breaks , and fidelity of DNA repair using a cell-free plasmid assay .\n\nRESULTS While A2780cp was more resistant to radiation than A2780s , it showed less ability for recovery of SLD and PLD .\n\nDNA strand-break induction was the same for both cell lines , while only at very high doses did A2780cp show greater DNA strand-break recovery than A2780s .\n\nFidelity of rejoining DNA was significantly higher in the A2780cp cell line .\n\nCONCLUSION The difference in radiosensitivity between these two cell lines was not related to recovery of PLD or SLD or to the induction of DNA damage .\n\nIt appears that fidelity of DNA rejoining , which was significantly higher in the resistant cell line , may be related to the difference in radiosensitivity .", "output": "Genomic instability and mutation" }, { "input": "We studied the effect of oxidative stress induced by hyperoxia , hydrogen peroxide , or menadione on mouse leukemia P388 cells at early ( 4 days ) and late ( 7 days ) stages of tumor growth .\n\nOxidative stress proved to inhibit cell division and to induce apoptosis .\n\nSeven-day leukemia cells feature lower proliferative potential and higher sensitivity to oxidative stress and platidiam .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "The effects of tea polyphenols and tea pigments on cell cycle of hepatic cancer cells were studied .\n\nHepG2 cells were incubated with 50 and 100 mg/L tea polyphenols and tea pigments for 48 h respectively .\n\nFlow cytometry , Western blot and RT-PCR analysis were used .\n\nFlow cytometry analysis showed that tea polyphenols and tea pigments induced G1 arrest .\n\nWestern blot analysis showed tea polyphenols and tea pigments significantly inhibited the expression of cyclin D1 protein and induced higher expression of P21WAFI/CIPI protein .\n\nThe result of RT-PCR analysis demonstrated that Cdk4 was significantly inhibited by tea polyphenols and tea pigments .\n\nIt is concluded that the induction of cell cycle arrest may be an important mechanism of tea on cancer prevention .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "A clinicopathological study of 41 cases of pituitary apoplexy in a series of 324 surgically treated pituitary adenomas is presented .\n\nIn 23 patients , the predominant operative finding was hemorrhage with or without necrosis .\n\nHowever , there were 15 ( 37.7% ) cases where pale , necrotic tissue with no evidence of hemorrhage was found at surgery .\n\nPale , necrotic material was particularly found when there was a long interval between the acute clinical event and surgery .\n\nIt is concluded that the pale , necrotic debris represents one stage in the resorption process of blood after hemorrhagic necrosis of pituitary adenomas .\n\nThis entity needs to be kept in mind especially since the material closely resemble the pultaceous material seen in craniopharyngiomas and epidermoid cysts .", "output": "Resisting cell death" }, { "input": "The effects of dose per fraction on the ability of amifostine exposure to elevate angiostatin levels in the serum of mice and to inhibit spontaneous metastases formation using the well-characterized murine Sa-NH sarcoma were investigated .\n\nAmifostine was administered intraperitoneally at doses of 50 , 100 , or 200 mg/kg every other day for 6 days to C3Hf/Kam mice until tumors reached an average size of 8 mm in diameter .\n\nAmifostine was again administered immediately following surgical removal of the tumor-bearing limbs by amputation , and then once more 2 days later .\n\nNontumor-bearing control animals were treated using the same dosing and surgery schedules .\n\nThe average number of pulmonary metastases per animal was determined for each experimental group .\n\nA significant reduction ( P <.05 ) in the average number of pulmonary metastases was observed only in the group of animals exposed to a dose per fraction of 50 mg/kg .\n\nA dose of 100 mg/kg was less effective while 200 mg/kg had no effect on metastases formation in this study .\n\nThe effects of amifostine exposure on serum levels of the angiogenesis inhibitor angiostatin were also determined using Western analysis .\n\nCorrelating with the antimetastatic effect measured , exposure of animals to 50 mg/kg of amifostine resulted in a four-fold enhanced serum level of angiostatin above control levels .\n\nThis phenomenon occurred in both tumor-bearing as well as nontumor-bearing animals .\n\nIn contrast , a dose of 200-mg/kg amifostine administered intraperitoneally under these conditions had no measurable effect on angiostatin serum levels in this animal system .\n\nThe enhanced ability of relatively low doses of amifostine to inhibit spontaneous metastases formation suggests that effective antimetastatic therapies with amifostine can be designed with minimal toxic side effects .\n\nWhile the dose responses for angiostatin production and metastases inhibition by amifostine are well correlated , the precise mechanism of action underlying these phenomena is unclear but is suggestive of a redox driven process(es) .", "output": "Activating invasion and metastasis" }, { "input": "During the period of 1996-1998 ninety-four gastrectomy specimens with gastric carcinoma referred to Erciyes University , Medical Faculty , Department of Pathology , were examined histopathologically , histochemically and immunohistochemically .\n\nGeneral characteristics of gastric carcinomas and prognostic factors were studied .\n\nAccording the Lauren classification , of the 94 cases of gastric carcinomas , 56 were intestinal type , 21 were diffuse type and 17 were mixed type carcinoma .\n\nThe association rates of Helicobacter pylori , chronic atrophic gastritis and intestinal metaplasia with gastric carcinomas were high .\n\nThere was strong immunorectivity with HSP70 in 62,5% of the intestinal type carcinomas .\n\nThis ratios were lower in diffuse and mixed type carcinomas ( p<0.05 ) .\n\nThe more tumor size and invasion depth increased , the more HSP70 immunoreactivity was obtained ( p<0.05 ) .\n\nHSP70 immunorectivity was considerably higher in the patients having lymph node metastasis and vascular invasion ( p<0.05 ) .\n\nIt was found that the NK cell number was low in the tumor but higher around the tumor in early gastric carcinomas , compared with advanced carcinomas ( p>0.05 ) .\n\nIn the tumors larger than 10 cm with vascular invasion , NK cell number was lower around the tumor ( p>0.05 ) .\n\nDefining prognostic factors of gastric carcinomas is of importance to clinicians .\n\nIt is thought that HSP70 immunoreactivity , besides invasion depth , lymph node metastasis , vascular invasion , tumor size and inflammatory reaction against the tumor , is important in prognosis and associated with advanced stage .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "AIM To develop a simple , fast and inexpensive approach as well as an instrument for detection of gene mutation .\n\nMETHODS Pyrosequencing based on bioluminometry assay was employed to detect gene mutation .\n\nPyrosequencing is a method of sequencing by synthesis step-by-step using four enzymes , DNA-polymerase , ATP sulfurylase , luciferase and apyrase .\n\nThe signal was produced by detecting pyrophosphate released during a dNTP incorporation .\n\nFor mutation detection , a DNA fragment was amplified by PCR at first , followed by a single-stranded DNA preparation .\n\nIn the second step , a short primer was annealed to the position just before the mutation point .\n\nFinally , specific dNTPs were added in terms of the template sequence .\n\nThe mutation species can be readily determined by the sequence .\n\nRESULTS A new instrument was developed for gene mutation detection by pyrosequencing .\n\nTo iteratively inject small amount of each dNTP for the sequencing reaction , capillaries were used to connect dNTP reservoirs and the reaction chamber .\n\nEach dNTP was delivered by adding a gas pressure on the top of a corresponding dNTP reservoir , by which 0.2 microL of dNTP can be exactly added each time .\n\nIt was theoretically proved that undesired liquid seep through the capillary did not affect the sequencing reactions in pyrosequencing .\n\nIn addition , the three possible variants ( wildtype , mutant and heterozygote ) of a mutant point Cys275Ser in P53 gene exon 8 were determined by pyrosequencing using the instrument .\n\nA simple method was also described for rapidly distinguishing the type of a variant .\n\nCONCLUSION The developed method is very simple , and the corresponding instrument is inexpensive and easy to operate , which can be used to detect many types of mutation .", "output": "Genomic instability and mutation" }, { "input": "This article describes the leading steps to develop an assay of DNA damage for the marine amphipod Gammarus locusta , using agarose gel electrophoresis ( AGE ) .\n\nTo test the sensitivity and feasibility of the AGE technique , X-ray assays were performed with naked DNA and with live amphipods .\n\nThese positive controls demonstrated the effectiveness of the AGE technique to not only discriminate distinct levels of DNA strand breakage in a dose-dependent manner , but also to identify and quantify the type of strand breakage induced .\n\nIt was also shown that it is possible to detect DNA damage using whole-body DNA extracts from amphipods .\n\nTo explore the potential of this technique for use in ecotoxicological studies with amphipods , a 96-h waterborne-copper toxicity test was performed .\n\nCopper-induced DNA strand breakage was first observed after 24 h of exposure , and was recorded again at 96 h , at a copper concentration of 20 microg l(-1) .\n\nThe absence of strand breakage after 48 h of exposure is discussed in the light of the underlying mechanisms of copper toxicity and DNA repair .\n\nThese studies demonstrated the feasibility of including DNA damage as a biomarker in ecotoxicological studies with amphipods .\n\nInformation gained from the use of this biomarker would help with the interpretation of chronic toxicity tests and would contribute to our understanding of the impact of genotoxic insult in marine invertebrates , particularly crustaceans .", "output": "Genomic instability and mutation" }, { "input": "Dietary antioxidants , such as the carotenoids , may protect DNA from oxidative damage .\n\nThis has been proposed to explain the epidemiological association between higher consumption of fruits and vegetables , which are rich in antioxidants , and lower incidence of cancer .\n\nHowever , this remains to be demonstrated conclusively .\n\nThe effects of carotenoid supplementation on 1 ) baseline DNA damage , 2 ) susceptibility of cellular DNA to oxidative attack , and 3 ) DNA repair were measured in the human lymphocyte cell line Molt-17 .\n\nBaseline DNA damage , susceptibility to oxidant attack ( 100 mumol/l H2O2 for 5 min at 4 degrees C ) , and disappearance of DNA single-strand breaks ( SSB ) after oxidative challenge were monitored by single-cell gel electrophoresis .\n\nDNA repair patch synthesis activity in cell extracts was determined using assays that measure nucleotide incorporation during repair of oxidative lesions in template DNA .\n\nUnlike single-cell gel electrophoresis , the parameters measured with these assays are not dependent on strand break religation .\n\nThere was no evidence that beta-carotene , lutein , or beta-cryptoxanthin supplementation protected cellular DNA from oxidation under basal conditions or after oxidative challenge .\n\nHowever , only carotenoid-supplemented cells exhibited a significant decrease in numbers of SSB over a 2-h period after treatment with H2O2 .\n\nCarotenoid supplementation did not provoke any detectable change in repair patch synthesis activity .\n\nWe conclude that supplementation with carotenoids at 8 mumol/l does not provide significant antioxidant protection for DNA in Molt-17 lymphocytes but may enhance recovery of cells from oxidative challenge , as measured by loss of SSB .\n\nWe argue that these data are most consistent with carotenoids acting to enhance DNA strand break repair .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "OBJECTIVE To investigate the expression and mutation of MTA1 , nm23H1 and E-cadherin(E-cad) genes in ovarian carcinoma ( OC ) in relation to lymph node ( LN ) metastasis .\n\nMETHODS A panel of normal ovarian tissues , primary OC specimens and corresponding LNS was examined for mRNA expression and mutation of MTA1 and nm23H1 and protin expression of E-cad genes by using RT-PCR , RT-PCR-SSCP and immunohistochemistry .\n\nRESULTS The frequency of MTA1 over expression was 100%(7/7) in primary OC with metastasis but only 38.5%(5/13) in those without metastasis ( P = 0.0103 ) .\n\nOverexpression of MTA1 was observed in 87.5%(6/7) of LNS with metastasis but in only 23%(3/13) of LNS without metastasis ( P = 0.0118 ) .\n\nIn contrast with MTA1 , low expression of nm23H1 mRNA was seen in 7 of 7 OC with metastasis but only in 4 of 13(30%) of those without metastasis ( P = 0.0043 ) .\n\nLow nm23H1 expression was also seen in 7 of 7 LNS with metastasis but only in 5 of 13 ( 38.5% ) nonmetastatic LNS ( P = 0.0102 ) .\n\nMeantime , no expression of E-cad protein was observed in 7 of 7 OC with metastasis but in 6 of 13(46.2%) of those without metastasis ( P = 0.044 ) .\n\nIn correlation analysis of the three genes , MTA1 reversely correlated with nm23H1 and E-cad respectively ( r = -0.903 , -0.803 ) , and positive correlation existed between nm23H1 and E-cad ( r = 0.724 ) .\n\nNo mutation of MTA1 , nm23H1 and was found by SSCP analysis .\n\nCONCLUSION The mRNA expression of MTA1 , nm23H1 and E-cad is positively and negatively correlated with LN metastasis .\n\nThe expression abnormalities but not the mutations of the three genes are frequent events related to LN metastasis of ovarian cancer .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "Angiogenesis is increased in hematologic malignancies , including non-Hodgkin lymphoma ( NHL ) .\n\nElevated serum levels of two important angiogenic factors , vascular endothelial growth factor ( VEGF ) and basic fibroblast growth factor ( bFGF ) , are associated with a poor prognosis .\n\nImmunohistochemistry was used to evaluate 27 patients with NHL and bone marrow involvement ( 17 with low-grade B-cell NHL , including 7 with higher grade transformation ; 6 with intermediate-grade B-cell NHL ; and 4 with T-cell lymphoma ) .\n\nAmong the 17 patients with low-grade B-cell NHL , results for 7 were positive for VEGF stain ( 41.2% ) , and results were negative for all other stains for VEGF receptors , bFGF , and bFGF receptors .\n\nIn the 10 patients with intermediate-grade B-cell NHL and T-cell lymphoma , all VEGF staining was positive ( 100% ) , but bFGF staining was only weakly positive in 2 .\n\nStaining results for seven patients who had low-grade B-cell NHL with higher grade transformation showed that VEGF staining was positive in large lymphoid cells of 5 patients and in small lymphoid cells of one patient .\n\nStaining for the receptors VEGFR-1 and VEGFR-2 was positive in large lymphoid cells in four and two cases , respectively .\n\nStaining for bFGF was positive in two cases of large lymphoid cells .\n\nWe concluded that VEGF , but not bFGF , was associated with higher tumor grading of NHL and high-grade transformation of low-grade lymphoma .", "output": "Inducing angiogenesis" }, { "input": "The role of T cells in eradicating leukemic cells has been well demonstrated for chronic myeloid leukemia ( CML ) .\n\nType 1 ( T1 ) T-cell cytokines play a major role in this antileukemic immune effect .\n\nStudies in cancer patients have demonstrated a decreased T1 cytokine production , measured by enzyme-linked immunosorbent assay ( ELISA ) , in cultures of peripheral blood mononuclear cells .\n\nThis observation of malignancy-related suppressed T1 cytokines also occurs in untreated chronic-phase ( CP ) CML , raising the question of the influence of different CML treatment regimens on this immunosuppression .\n\nIntracellular flow cytometry ( ICF ) has facilitated the evaluation of cytokines on a single-cell level .\n\nThis study analyzed T1 ( interferon-gamma ) cytokine production in purified peripheral blood T cells by ICF , comparing different therapy approaches for CML .\n\nTwenty-one newly diagnosed CP CML patients were compared with 24 patients treated with interferon-alpha ( IFN-alpha ) and to 30 allogeneic bone marrow transplant ( BMT ) recipients ( BCR-ABL negative by reverse-transcriptase polymerase chain reaction , and free of , or having only limited graft-versus-host disease at the time of study ) .\n\nThirty-seven healthy controls were included .\n\nOur results showed a significantly decreased T-cell IFN-gamma synthesis in CP CML patients in relation to healthy controls ( P = 0.0007 ) .\n\nTreatment with IFN-alpha resulted in a shift from immunosuppression--documented for the group of untreated patients--to immunopotentiation , with an increase of T-cell IFN-gamma production ( P = 0.0266 ) .\n\nNotably , BMT enhanced IFN-gamma production of T cells to a level not only exceeding untreated patients ( P < 0.0001 ) but also healthy volunteers ( P < 0.0001 ) .\n\nThe observation of T1 cytokine up-regulation with IFN-alpha therapy indicates that enhanced T-cell function may be achievable in patients with CML , even in the absence of an allo-response .", "output": "Avoiding immune destruction" }, { "input": "Modulation of the balance between pro- and antiangiogenic factors holds great promise for the treatment of a broad spectrum of human disease ranging from ischemic heart disease to cancer .\n\nThis requires both the identification of angiogenic regulators and their efficient delivery to target organs .\n\nHere , we demonstrate the use of a noncatalytic fragment of matrix metalloproteinase 2 ( termed PEX ) delivered by lentiviral vectors in different angiogenesis models .\n\nTransduction of human endothelial cells with PEX virus suppressed endothelial invasion and formation of capillary-like structures without affecting chemotaxis in vitro .\n\nLentiviral delivery of PEX blocked basic fibroblast growth factor-induced matrix metalloproteinase 2 activation and angiogenesis on chicken chorioallantoic membranes .\n\nPEX expression also inhibited tumor-induced angiogenesis and tumor growth in a nude mouse model .\n\nThus , our study shows that lentiviral vectors can deliver sufficient quantities of antiangiogenic substances to achieve therapeutic effects in vivo .", "output": "Inducing angiogenesis" }, { "input": "ZBP-89 ( ZNF148 ) is a Zinc finger Binding Protein of 89 kDa that binds GC-rich DNA elements .\n\nOriginally , it was expression cloned using a DNA element mediating EGF regulation of the gastrin promoter .\n\nZBP-89 functions as both a transcriptional activator and repressor .\n\nA variety of extracellular regulators including TGFbeta , retinoic acid and butyrate stimulate ZBP-89 gene expression .\n\nButyrate activation of p21WAF1 is potentiated by ZBP-89 through the recruitment of the co-activator p300 , while chronic stimulation by butyrate increases ZBP-89 gene expression correlating with cell differentiation .\n\nZBP-89 stimulates growth arrest and apoptosis through its ability to bind the p21WAF1 promoter or its ability to form protein-protein interactions with p53 .\n\nZBP-89 protein is elevated in a variety of gastrointestinal cancers as well as the pancreas .\n\nIn particular , ZBP-89 is normally expressed in pancreatic islets and ducts and in about 30% of pancreatic adenocarcinomas .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "The purpose of this study was to use the proteomics approach , which is based on high resolution two-dimensional electrophoresis coupled with multivariate correspondence analysis and mass spectrometry , to classify objectively the biochemical basis of the anti-cancer activity of the synthetic cyclin-dependent kinase inhibitor , bohemine ( BOH ) .\n\nThe changes in the cell cycle and corresponding protein composition of the A549 human lung adenocarcinoma cell line after treatment with BOH were evaluated and proteins differentially expressed in the BOH treated A549 cells , compared to the untreated A549 counterparts , were selected .\n\nThirteen of these candidate proteins associated with the drug effects in vitro were identified by mass spectrometry .\n\nMany of these proteins fall into one of three functional categories : i ) metabolic pathways ( glycolysis , nucleic acid synthesis and NADPH production ) , ii ) stress response and protein folding , and iii ) cytoskeleton and exocytosis .\n\nChanges in protein expression patterns corresponded to a higher resistance of A549 lung carcinoma cells to BOH when compared to the CEM leukaemia cell line .\n\nThese protein changes reflect a fine balance of the resistant versus the susceptible phenotype in response to the drug .\n\nSince BOH is a selective cyclin-dependent kinase inhibitor , changes in the protein expression pattern can be more generally associated with cell cycle regulation as evidenced by inhibition of cell cycling in A549 cells .\n\nOur conclusions further underline the importance of cell cycle control in both the cellular signalling and metabolic pathways .", "output": "Evading growth suppressors" }, { "input": "To investigate the significance of sialylation and sulfation of lactosylceramide in transformed cells , we established ganglioside GM3- and lactosylsulfatide ( SM3)-reconstituted cells by transfecting cDNAs of GM3 synthase and cerebroside sulfotransferase into the J5 subclone of 3LL Lewis lung carcinoma cells .\n\nThe J5 clone was selected for the transfection of these genes because it lacks GM3 and SM3 but accumulates lactosylceramide .\n\nThe anchorage-dependent growth of both GM3- and SM3-reconstituted cells was similar .\n\nHowever , anchorage-independent growth ( as measured by colony-forming ability in soft agar ) of the SM3- reconstituted cells was almost completely lost , which supports our previous observation showing the suppression of tumorigenic potential in vivo and beta1 integrin gene expression induced by the introduction of cerebroside sulfotransferase gene ( Kabayama et al. [ 2001 ] J. Biol .\n\nChem. , 276 , 26777-26783 ) .\n\nThe GM3-reconstituted cells formed a significantly higher number of colonies in soft agar compared to mock-transfected cells and began to proliferate and become resistant to apoptosis when serum was depleted , indicating that endogenous GM3 is essential for maintaining these fundamental properties of malignant cells .\n\nWe also found that serum-induced ERK1/2 activation was suppressed in the GM3-reconstituted cells , suggesting that anchorage-independent cell cycle initiation by endogenous GM3 is elicited through pathway(s) independent of ERK1/2 activation .\n\nThe selective down-regulation of platelet-derived growth factor ( PDGF)-dependent ERK1/2 activation in the GM3-reconstituted cells was due to the substantial decreases of PDGF alpha receptor mRNA and protein , but in the SM3-reconstituted cells PDGF alpha receptor expression was similar to mock cells .\n\nThus , endogenously produced GM3 and SM3 differentially and distinctly regulate tumor-progression ability , that is , GM3 leads the transformed phenotype of J5 cells to promotion and SM3 to abrogation .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The anti-breast cancer drug tamoxifen has recently been shown to cause an increase in intracellular free-Ca(2+) concentrations ( [ Ca(2+)](i) ) in renal tubular cells , breast cells and bladder cells .\n\nBecause tamoxifen is known to alter ovary function in human patients and in rats , the present study was aimed at exploring whether tamoxifen could alter Ca(2+) movement in Chinese hamster ovary ( CHO-K1 ) cells .\n\nCytosolic free-Ca(2+) levels in populations of cells have been explored by using fura-2 as a fluorescent Ca(2+) indicator .\n\nTamoxifen at concentrations above 1 micro M increased [ Ca(2+)](i) in a concentration-dependent manner with an EC(50) value of 8 micro M. The Ca(2+) signal was reduced by removing extracellular Ca(2+) , but was not affected by nifedipine , verapamil , diltiazem or ICI 182,780 ( an estrogen receptor antagonist ) .\n\nPretreatment with 1 micro M thapsigargin ( an endoplasmic reticulum Ca(2+) pump inhibitor ) to deplete the endoplasmic reticulum Ca(2+) abolished 10 micro M tamoxifen-induced Ca(2+) release .\n\nNeither inhibition of phospholipase C with 2 micro M U73122 nor depletion of ryanodine-sensitive Ca(2+) stores with 50 micro M ryanodine affected tamoxifen-induced Ca(2+) release .\n\nCell proliferation assays using ELISA revealed that overnight incubation with 5-10 micro M tamoxifen inhibited cell proliferation by 20% , and 20 micro M tamoxifen killed all cells .\n\nTogether , the results suggest that , in CHO-K1 cells , tamoxifen induced a [ Ca(2+)](i) increase by causing store-Ca(2+) release from the endoplasmic reticulum in an phospholipase C-independent manner , and by inducing Ca(2+) influx .\n\nThe action of tamoxifen appears to be dissociated from estrogen receptor activation .\n\nLonger incubation with tamoxifen ( >5 micro M ) was cytotoxic .", "output": "Sustaining proliferative signaling" }, { "input": "The identification of molecular markers related to critical biological processes during carcinogenesis may aid in the evaluation of carcinogenic potentials of chemicals and chemical mixtures .\n\nWork from our laboratory demonstrated that a single treatment with N-methyl-N'-nitro-N-nitrosoguanidine ( MNNG ) enhanced spontaneous malignant transformation of the human keratinocyte cell line RHEK-1 .\n\nIn contrast , chronic low-level exposure of cells to arsenic alone or in a mixture containing arsenic , cadmium , chromium , and lead inhibited malignant conversion .\n\nTo identify changes in gene expression that influence these different outcomes , cDNA microarray technology was used .\n\nAnalysis of multiple human arrays in MNNG-transformed RHEK-1 cells , designated OM3 , and those treated with arsenic or the arsenic-containing metal mixture showed unique patterns of gene expression .\n\nGenes that were overexpressed in OM3 included oncogenes , cell cycle regulators , and those involved in signal transduction , whereas genes for DNA repair enzymes and inhibitors of transformation and metastasis were suppressed .\n\nIn arsenic-treated cells , multiple DNA repair proteins were overexpressed .\n\nMixture-treated cells showed increased expression of a variety of genes including metallothioneins and integrin 4 .\n\nThese cells showed decreased expression of oncogenes , DNA repair proteins , and genes involved in the mitogen-activated protein kinase pathway .\n\nFor comparison we are currently analyzing gene expression changes in RHEK-1 cells transformed by other means .\n\nThe goal of these studies is to identify common batteries of genes affected by chemical modulators of the carcinogenic process .\n\nMechanistic studies may allow us to correlate alterations in their expression with sequential stages in the carcinogenic process and may aid in the risk assessment of other xenobiotics .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Malignant gliomas are the most common primary brain tumors in humans .\n\nHowever , poor response to conventional therapeutic approaches , including chemotherapy , leads invariably to disease recurrence and progression .\n\nThe organo-tin derivative triethyltin(IV)lupinylsulfide hydrochloride ( IST-FS 29 ) was identified and developed as potential antiproliferative agent in human cancer cell lines .\n\nHowever , for its peculiar chemical structure and good lipophilicity , this compound also appeared an eligible candidate for the treatment of gliobastoma cells .\n\nThe present experiments were designed to explore the in vitro effects of IST-FS 29 on four human glioblastoma cell lines : A-172 , DBTRG.05MG , U-87MG and CAS-1 .\n\nThe average IC50 values were obtained by MTT assay and ranged between 3 and 10 microM .\n\nTime-course assays with cell recovery after drug withdrawal , demonstrated marked cytotoxicity following exposure to IST-FS 29 for 8 , 24 and 72 h .\n\nCultures treated for 8 h were able to partially re-grow by 144 h ; on the contrary , longer times of exposure did not allow surviving cells to recover from the damage and actively proliferate .\n\nCell morphology of cultures exposed to IST-FS 29 was assessed by inverted light microscopy after 24 and 72 h and was more consistent with cell death by necrosis which included cell size reduction , vacuolation of cytoplasm , round dying cells .\n\nThe present results and our previous data , in vitro and in vivo , indicate the relevant cytotoxic activity of this organo-tin compound and suggest that IST-FS 29 might be a promising novel agent to be developed for the treatment of malignant brain neoplasms .", "output": "Resisting cell death" }, { "input": "Purpose of this work was to synthesize several cis-/trans- isomer pairs of the platinum(II) complexes , and study the extent and the mode of their antiproliferative activity on HeLa cells .\n\nSix platinum(II) isomer pairs have a general formula cis-/trans-[PtA2X2] , where A is ligand : ammonia ( NH3 ) , pyridine ( Py ) ; and X is ligand : chloride ion ( Cl- ) , bromide ion ( Br- ) , iodide ion ( I- ) , thiocyanato ion ( SCN- ) ; four compounds have different structural formulas , and these are cis-/trans-[Pt(NH2OH)2(NH3)2]Cl2 , and cis-/trans-Pt(Gly)2 , where Gly is bidentate glycinato ligand .\n\nResults of the MTT assay , showed that six cis- and one trans-platinum(II) complexes exhibited cytotoxicity ( IC50 ) ranging between 5 and 33 microM .\n\nMost of the cis-platinum(II) isomers caused significant alteration of cell cycle phases progression , and induced apoptosis in degree that varied among different compounds , as evaluated using flowcytometry and morphological study .\n\nSpectrophotometric analysis ( AAS ) indicated that there is no correlation between intracellular platinum(II) accumulation and cytotoxicity of tested complexes .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The effects of deoxycholic acid ( DCA ) and ursodeoxycholic acid ( UDCA ) on 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine ( PhIP)-induced aberrant crypt foci ( ACF ) in the rat colon were examined .\n\nThe effect of these bile acids on DNA adduct formation by PhIP in the colon was then analyzed , since the main action of PhIP is the formation of DNA adducts and subsequent gene mutations .\n\nFor the ACF study , male F344 rats were administered PhIP-HCl ( 75 mg/kg , 10 doses ) by gavage , and a diet containing bile acid ( 0.4% DCA or UDCA ) was provided from 3 days before the first dose of PhIP for 8 weeks .\n\nThe mean number of ACF per colon of DCA , UDCA and controls were 9.9 , 2.4 and 5.5 , respectively .\n\nThe ACF number was significantly increased by DCA and decreased by UDCA ( P<0.001 ) .\n\nTo examine the effect of bile acids on DNA adduct formation , male F344 rats were fed a diet supplemented with bile acids ( 0.1 or 0.4% of DCA and UDCA ) 7 days prior to the PhIP administration .\n\nAll rats were administered a single dose of PhIP-HCl ( 50 mg/kg ) by gavage and sacrificed 48 hours later .\n\nDNA adduct levels of the 0.1% UDCA , 0.1% DCA and controls were 2.93 ( adducts/10(7) nucleotides ) , 2.65 and 1.10 , respectively .\n\nThose of 0.4% UDCA , 0.4% DCA and controls were 1.64 , 1.30 and 1.00 , respectively .\n\nThe PhIP-DNA adduct level was significantly increased by administration of 0.1% UDCA , 0.1% DCA ( P<0.05 ) and 0.4% UDCA ( P<0.01 ) .\n\nThe increasing effect of both DCA and UDCA on PhIP-induced DNA adduct formation was unexpected , and was not directly associated with ACF formation .", "output": "Genomic instability and mutation" }, { "input": "Previously we have found deregulation of collagen metabolism in human pancreatitis and pancreatic cancer tissues .\n\nInsulin-like growth factor-I ( IGF-I ) is known to stimulate collagen biosynthesis through interaction with IGF-I receptor .\n\nIGF-I binding proteins ( BPs ) regulate the activity of IGF-I .\n\nWe investigated whether serum and tissue IGF-I and IGF-BPs as well as tissue IGF-I receptor expression may reflect disturbances of collagen metabolism in patients with pancreatitis and pancreatic cancer .\n\nIn pancreatitis tissue , a significant increase in IGF-I and IGFBP-3 content was accompanied by a distinct increase in IGF-I receptor expression , compared to control pancreas tissue .\n\nIn contrast , serum from patients with pancreatitis did not show significant increases in IGF-I and IGFBP-3 levels , however , significant increases in IGFBP-1 level ( 2.5 fold ) .\n\nMoreover , a distinct decrease in radioactive IGF-binding to the BPs , compared to control serum , was found .\n\nPancreatic cancer tissue and serum of patients with pancreatic cancer showed significant increases in IGF-I , IGFBP-3 and IGFBP-1 content , accompanied by dramatic increases in IGF-I tissue receptor expression , compared to controls .\n\nIn serum of patients with pancreatic cancer distinct increases in radioactive IGF-binding to 46 kDa BP , compared to control serum , were observed .\n\nThe data suggest that disturbances in tissue collagen metabolism during pancreatic diseases may result from deregulation of IGF-I homeostasis and that elevated serum levels of IGF-I , IGFBP-3 and IGFBP-1 may serve as markers of pancreatic cancer .", "output": "Sustaining proliferative signaling" }, { "input": "The epidemiologic association between asbestos exposure and human malignant mesothelioma is well established .\n\nHowever , the molecular mechanisms linking asbestos exposure of humans and the subsequent mesothelioma formation is not well understood .\n\nThe most frequent genetic changes found so far in human malignant mesothelioma ( HMM ) are deletions and point mutations in the tumor suppressor genes p16INK4a and NF2 .\n\nWhereas homozygous deletions appear to be the predominant mechanism leading to p16/CDKN2A inactivation , inactivating point mutations coupled with allelic loss mainly occur at the NF2 locus .\n\nIn the present study , asbestos-treated human mesothelial cells ( HMC ) , SV40-transformed human mesothelial cells ( MeT-5A ) and a human mesothelioma cell line ( COLO ) were investigated for genetic changes of cell cycle genes ( cyclin D1 , p16INK4a , RB1 , CDK2 ) using multicolor fluorescence in situ hybridization ( mFISH ) in interphase cells .\n\nThe results show that cyclin D1 is unaffected in all investigated cells .\n\nThe p16INK4a gene locus was shown to be mutated in COLO cells but not in HMC .\n\nAfter labeling of CDK2 and RB1 , hemizygous loss of one allele of each gene was observed in asbestos-treated HMC whereas gene amplification of these genes was detectable in MeT-5A and COLO cells .\n\nOur data indicate that disarrangement of the RB1 dependent pathway seems to be involved in mesothelioma formation .", "output": "Sustaining proliferative signaling" }, { "input": "The early stages of head and neck cancer are presumed to require a senes of genetic alterations that are not represented by a distinct clinical phenotype .\n\nTherefore , genes with altered expression in the preneoplasia may be useful for the early detection of this highly recurrent cancer .\n\nIn this study , we immortalized normal human oral keratinocytes ( NHOK ) by retroviral-mediated infection of HPV 16 transforming oncogenes , E6 and E7 ( HOK16E6E7 ) .\n\nUsing the Affymetrix gene chip ( U95Av2 ) , we identified 177 known genes and EST that were overexpressed at least 3-fold or above in the immortalized cells , while 133 were down-regulated compared to NHOK .\n\nNorthern blot analysis showed elevated levels of p55CDC in the immortalized cells , while NHOK showed high basal expression of small proline rich protein ( SPRR2 ) .\n\nThe altered expression of these genes maybe associated with cellular proliferation or differentiation and the early stages of oral carcinogenesis .", "output": "Enabling replicative immortality" }, { "input": "BACKGROUND Angiogenesis is of crucial importance for tumor growth and development of metastases .\n\nVascular endothelial growth factor ( VEGF ) has a potent angiogenic activity and mutations of the p53 gene has been thought to upregulate VEGF .\n\nThe purpose of our study was to evaluate the prognostic significance of these tumor biomarkers for angiogenesis relative to the information derived from established clinicopathological parameters in gastric cancer .\n\nMETHODS In this study , we conducted an immunohistochemical investigation of VEGF and p53 expression in 145 tissue samples obtained from gastric cancer patients undergoing curative surgical treatment .\n\nTo evaluate angiogenesis , microvessel density ( MVD ) was counted by staining endothelial cells immunohistochemically using anti-CD34 monoclonal antibody .\n\nRESULTS High MVD was significantly associated with depth of tumor invasion and distant metastasis ( p = 0.004 , 0.021 , respectively ) .\n\nMoreover , overall survival for patients with high MVD were significantly lower than that of low MVD ( p = 0.048 ) .\n\nPositive expression of VEGF correlated significantly with lymph node and distant metastasis ( p = 0.040 , 0.048 , respectively ) .\n\nHowever , no significant correlation was found between p53 expression and various clinicopathological parameters .\n\nVEGF positive tumors showed a higher MVD than VEGF negative tumors ( p = 0.028 ) .\n\nThe expression of p53 did not correlate with VEGF expression .\n\nAlso , the relationship between the status of p53 expression and MVD had not statistically significant differences .\n\nIn the multivariate analysis , status of VEGF , p53 expression and MVD were not an independent prognostic factor .\n\nCONCLUSION VEGF seems to be an important , clinically relevant inducer of angiogenesis and angiogenesis assessed by the MVD may be a useful marker for predicting metastasis in gastric cancer .\n\nHowever , further studies are warranted to clarify the impact of p53 on the angiogenesis and the prognostic significance of angiogenesis in gastric cancer .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "The cellular mechanisms of anti-androgen-induced tumor regression have not been investigated in great detail .\n\nWe have compared the induction of cell death in the androgen-dependent , non-invasive LNCaP prostate cancer cell line by Casodex and TNF-alpha .\n\nBoth agents induce a dose and time-dependent decrease in cell viability in vitro .\n\nHowever , Casodex does not induce classical DNA fragmentation to oligonucleosomes typically induced by TNF-alpha , but rather induces cleavage to form intermediate 60 kb DNA fragments .\n\nRT-PCR based analysis demonstrates that in LNCaP cells Casodex coordinately alters the expression of steady-state level of mRNAs of several matrix metalloproteases and their cognate inhibitors ( most notably MMP2 and TIMP1 ) .\n\nZymography and reverse zymography confirm that the ratio of metalloprotease(s) to inhibitor(s) is altered in favor of activation of the proteases .\n\nIn a small percentage of the treated LNCaP cells , the activation of the extracellular matrix ( ECM)-proteases by Casodex also induces an invasive phenotype .\n\nThe acquisition of an invasive phenotype is not seen when LNCaP cells are treated with TNF-alpha , and is not seen when the LNCaP cells are treated with both compounds simultaneously , suggesting that the phenomenon may be specific to particular classes of compounds .\n\nThese observations have significant implications in the treatment of prostate cancer , since the appearance of a more aggressive phenotype following treatment is clearly undesirable .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "To elucidate role of the three enzymes in hepatocarcinogenesis , hMTH1 , hOGG1 and hMYH , mRNA expression were examined by using RT/semi-quantitative real-time PCR and 8-O-HdG levels was studied by HPLC/ECD in HCC and non-tumorous liver tissue of 21 patients with hepatocellular carcinoma ( HCC ) .\n\nIt was found that the 8-OHdG level in non-tumourous liver tissue was significantly higher than in HCC tissue ( P = 0.006 ) , and this was correlated with the degree of inflammation .\n\nThe hMTH1 expression in HCC tissue was significantly higher than in non-tumorous liver tissue ( P = 0.014 ) .\n\nInversely , The hMYH alpha expression was significantly increased ( P = 0.039 ) in non-tumorous liver tissue .\n\nNo difference was seen in hOGG1 expression in non-tumorous liver and HCC tissue .\n\nA significant linear correlation between hMTH1 and hOGG1 expression was found both in HCC tissue ( r = 0.809 , P < 0.001 ) and in non-tumorous liver tissue ( r = 0.883 , P < 0.001 ) .\n\nOur findings suggested a reactive rather than pathogenic role of the DNA repair enzymes in the hepatocarcinogenesis .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "OBJECTIVE The aims of this study were to analyse the results and long term outcome in a prospective non randomised trial of 74 patients treated by laparoscopic colo-rectal resection for cancer , and to determine wether survival and recurrence are or are not compromised by an initial laparoscopic approach .\n\nPATIENTS AND METHODS Seventy-four patients with colo-rectal carcinoma were included in a prospective trial and treated by laparoscopic resection .\n\nAll patients were reviewed at 1 , 3 , and 6 months interval .\n\nA median of 5 years follow up was available .\n\nForty-eight patients ( 65% ) had more than 3 years of follow up .\n\nRESULTS Six conversions ( 8.1% ) were necessary : 2 for tumor invasion of adjacent organs , 2 for limited margin resection in lower rectal tumors , 1 for small bowel injury and 1 for obesity .\n\nAfter surgery , passing flatus occurred at 34.3 +/- 16.7 h and oral intake could be reinstaured at 42.6 +/- 22 h .\n\nMean postoperative stay was 8.2 +/- 3.4 days .\n\nNo death occurred .\n\nThe overall morbidity was about 13.5% .\n\nThe rate of late complications was 5.4% .\n\nTwo port site metastasis ( 2.6% ) were seen in locally advanced carcinoma .\n\nRecurrence rate at 5 years was 0% for Dukes A , 20% for Dukes B , 39.2% for Dukes C. Survival rate at 5 years was 100% for Dukes A , 80% for Dukes B , and 60.7% for Dukes C. These results are similar to those of conventional open surgery .\n\nCONCLUSION Laparoscopic colorectal resection for cancer can be performed safely , with a low morbidity and rare late complications .\n\nLong term follow up ( 5 years ) assessment shows similar outcome compared with conventional surgery .", "output": "Activating invasion and metastasis" }, { "input": "Solar ultraviolet radiation is considered to be injurious rather than necessary for most organisms living on the earth .\n\nIt is reported that the risk of skin cancer in humans increases by the depletion of the ozone layer .\n\nWe have examined the genotoxicity of solar ultraviolet , especially the longer wavelength light , using Drosophila .\n\nRecently , we have demonstrated that light of wavelength up to 340 nm is mutagenic on Drosophila larvae .\n\nUsing an excision repair-deficient Drosophila strain ( mus201 ) , we have obtained results suggesting that the lesion caused in larvae by the 320 nm-light irradiation may be similar to the damage induced by irradiation at 310 nm , and that light of 330 and 340 nm may induce damage different from that induced by 310 and 320 nm-light .\n\nTo examine the difference in DNA damage induced by light of a particular wavelength , we performed monochromatic irradiation on larvae of two Drosophila strains ; one excision repair-deficient ( mei-9 ) and another postreplication repair-deficient ( mei-41). 310 and 320 nm-light was more mutagenic in the mei-9 strain than in mei-41 , whereas 330 and 340 nm-light was more mutagenic in mei-41 than in mei-9 .\n\nIt is demonstrated that the mei-41 gene is a homologue of the human atm gene which is responsible for a cell cycle checkpoint .\n\nThis result suggests that 310-320 nm-light induces DNA damage that is subject to nucleotide excision repair ( NER ) and that 330-360 nm-light causes damage to be recognized by the cell cycle checkpoint but it is not repairable by NER .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "In the course of a medicinal chemistry program aimed at discovering novel tumour-active rebeccamycin derivatives targeting DNA and/or topoisomerase I , a series of analogues with the sugar residue linked to the two indole nitrogens was recently developed .\n\nTwo promising drug candidates in this staurosporine-rebeccamycin hybrid series were selected for a DNA-binding study reported here .\n\nThe DNA interaction of the cationic indolocarbazole glycosides MP059 bearing a N,N-diethylaminoethyl side chain and MP072 containing a sugar bearing an amino group was compared with that of the uncharged analogue MP024 .\n\nThe results show that the addition of a cationic substituent , either directly on the indolocarbazole chromophore or on the carbohydrate residue , significantly reinforces the interaction of the drugs with nucleic acids .\n\nThe two cationic molecules MP059 and MP072 recognise preferentially sequences containing GpT.ApC and TpG.CpA steps but they do not inhibit topoisomerase I , in contrast to the parent uncharged derivative MP024 which stimulates DNA single strand breaks by topoisomerase I. The cytotoxic activity of the indolocarbazole derivatives bearing positively charged groups is one order of magnitude higher than that of the neutral compound MP024 .\n\nThe high cytotoxic potential can be attributed to the enhanced DNA binding and sequence recognition capacity of the cationic compounds .\n\nThe study provides useful information for further structure-activity relationship studies in the indolocarbazole series .", "output": "Genomic instability and mutation" }, { "input": "Acetyl-11-keto-beta-boswellic acid ( AKBA ) is a naturally occurring pentacyclic triterpene isolated from the gum resin exudate from the stem of the tree Boswellia serrata ( frankincense ) .\n\nAKBA has been recently identified as a novel , orally active , non-redox and non-competitive 5-lipoxygenase inhibitor that also inhibits topisomerase I and II in vitro .\n\nBecause natural pentacyclic triterpenes have an antiproliferative effect against different tumor types , we investigated the effects of AKBA on the proliferation of 11 primary cell cultures established from human surgical specimens of meningiomas , common central nervous system tumors .\n\nTreatment of meningioma cells by AKBA revealed a potent cytotoxic activity with half-maximal inhibitory concentrations in the range of 2-8 microM .\n\nAt similar , physiologically achievable concentrations , AKBA rapidly ( within minutes ) and potently inhibited the phosphorylation of extracellular signal-regulated kinase 1 and 2 ( Erk-1 and Erk-2 ) in meningioma cells stimulated with platelet-derived growth factor BB .\n\nHigh expression level of 5-LO was detected in primary meningioma cells and surgical specimens by immunoblotting analysis , suggesting the possible role of 5-LO in meningioma tumorigenesis .\n\nConsidering the critical importance of the Erk-1/2 signal transduction pathway not only in meningiomas but in other human neoplasms , the interruption of signaling through this evolutionarily conserved pathway might be one of the mechanisms by which AKBA induces suppression of proliferation and apoptosis of different tumor types .", "output": "Sustaining proliferative signaling" }, { "input": "Recent epidemiological and experimental investigations suggest a close relationship between cyclooxygenase ( COX ) and pathogenesis of colorectal cancer .\n\nThere are two isoforms , COX-1 and COX-2 , which differ in physiological functions and distribution .\n\nThis study is to investigate the possible roles of both isoforms in the proliferation of colon carcinoma cells .\n\nA human colon carcinoma cell line , COLO 320DM , was transfected with an eukaryotic expression vector ( pEF-BOS ) carrying cDNA of either COX-1 or COX-2 .\n\nBoth COX-1 and COX-2-expressing cells exhibited a similar enzyme activity , 8-10 nmol/10 min/mg of protein .\n\nGrowth rates of both COX-expressing cells were increased by about 2 fold as compared with mock-transfected cells .\n\nThe stimulated growth of the COX-expressing cells was confirmed by the increased DNA synthesis as assessed by [ 3H]thymidine incorporation .\n\nFurthermore , expression of epidermal growth factor receptor ( EGFR ) was markedly increased in the COX-expressing cells as examined by reverse transcriptase-polymerase chain reaction ( RT-PCR ) .\n\nA COX inhibitor , indomethacin , suppressed the stimulated growth , increased DNA synthesis and induction of epidermal growth factor receptor in the COX-1 and COX-2-transfected cells .\n\nThese results suggest that not only COX-2 but COX-1 is involved in the proliferation of human colon carcinoma cells through the induction of EGFR .", "output": "Sustaining proliferative signaling" }, { "input": "Neutrophils have become recognised as important contributors to the effectiveness of tumour eradication by photodynamic therapy ( PDT ) .\n\nIn this study , we have used the mouse SCCVII squamous cell carcinoma model to investigate the activity of neutrophils in tumours treated by PDT .\n\nTumour levels of neutrophilic myeloperoxidase ( MPO ) demonstrated not only a massive and sustained sequestration of these cells in PDT-treated tumours but also revealed their activated state evidenced by the presence of released MPO .\n\nAmong the adhesion molecules expressed on tumour vascular endothelium , ICAM-1 appears to be of primary importance in the invasion of neutrophils into PDT-treated tumours , because its functional blocking with monoclonal antibodies reduced the tumour cure rate .\n\nA marked upregulation of its ligands CD11b/CD18 and CD11c/CD18 found on neutrophils associated with PDT-treated tumours supports this assumption .\n\nTo evaluate the role of inflammatory cytokines regulating neutrophil activity , neutralising antibodies were given to mice before PDT treatment .\n\nThe results suggest that IL-1beta activity is critical for the therapeutic outcome , since its neutralisation diminished the cure rates of PDT-treated tumours .\n\nNo significant effect was observed with anti-IL-6 and anti-TNF-alpha treatment .\n\nFurther flow cytometry-based examination of neutrophils round in PDT-treated tumours revealed that these cells express MHC class II molecules , which suggests their engagement as antigen-presenting cells and involvement in the development of antitumour immune response .", "output": "Tumor promoting inflammation" }, { "input": "Common use of antimutagens and anticarcinogens in everyday life is an effective measure for preventing human cancer and genetic diseases .\n\nAntioxidant properties of tea have vast potential as protective agents against diverse toxic effects .\n\nThe present study was aimed to evaluate the role of aqueous clonal tea extracts ( green tea , oolong tea and black tea ) in modulating the genotoxic damage induced by cyclophosphamide ( CP ) , a commonly used chemotherapeutic drug and a well-known mutagen and clastogen .\n\nAll the three tea extracts at 1 and 2% concentration did not increase the frequency of micronucleated polychromatic erythrocytes ( MPE ) in bone marrow cells of mice when administered individually .\n\nThe tea extracts decreased the micronuclei ( MN ) induced by CP .\n\nTherefore , regular intake of tea may improve the antioxidant status in in vivo and thereby reduce the risk of cancer and coronary heart disease .", "output": "Genomic instability and mutation" }, { "input": "To investigate whether the Bcl-2 gene family is involved in modulating mechanism of apoptosis and change of cell cycle protein induced by curcumin in acute myeloid leukemia HL-60 cell line and primary acute myelogenous leukemic cells , the Bcl-2 family member Mcl-1 , Bax and Bak and cell cycle proteins including P27kipl , P21wafl , cyclin D3 and pRbp- were selected and their expression detected by SABC immuno-histochemical stain method .\n\nThe attitude of sub-G1 peak in DNA histogram was determined by FCM .\n\nThe TUNEL positive cell percentage was identified by terminal deoxynucleotidyl transferase ( TdT)-mediated Biotin dUNP end labeling technique .\n\nIt was found that when HL-60 cells were treated with 25 mumol/L curcumin for 24 h , the expression level of Mcl-1 was down-regulated , but that of Bax and Bak up-regulated time-dependently .\n\nThere was significant difference in the expression level of Mcl-1 , Bax and Bak between the curcumin-treated groups and control group ( P < 0.05-0.01 ) .\n\nAt the same time , curcumin had no effect on progress of cell cycle in primaty acute myelogenous leukemia at newly diagnosis , but could increase the peak of Sub-G1 ( P < 0.05 ) , and down-regulate the expression of Mcl-1 and up-regulate the expression of Bax and Bak with the difference being statistically significant .\n\nThe expression of P27kipl , P21wafl and pRbp- were elevated and that of cyclin D3 decreased in the presence of curcumin .\n\nThese findings suggested that the Bcl-2 gene family indeed participated in the regulatory process of apoptosis induced by curcumin in HL-60 cells and AML cells .\n\nCurcumin can induce apoptosis of primary acute myelogenous leukemic cells and disturb cell cycle progression of HL-60 cells .\n\nThe mechanism appeared to be mediated by perturbing G0/G1 phases checkpoints which associated with up-regulation of P27kipl , P21wafl and pRbp- expression , and down-regulation of cyclin D3 .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "To evaluate the effects of adenovirus ( Ad)-mediated transfer of p53 and p16 on human bladder cancer cells EJ , EJ were transfected with Ad-p53 and Ad-p16 .\n\nCell growth , morphological change , cell cycle , apoptosis were measured using MTT assay , flow cytometry , cloning formation , immunocytochemical assays .\n\nAd-p16 or Ad-p53 alone could inhibit the proliferating activity of EJ cells in vitro .\n\nAd-p53 could induce apoptosis of partial EJ cells .\n\nG1 arrest was observed 72 h after infection with Ad-p16 , but apoptosis was not obvious .\n\nThe transfer of Ad-p16 and Ad-p53 could significantly inhibit the growth of EJ cells , decrease the cloning formation rate and induce apoptosis of large number of EJ cells .\n\nThe occurrence time of subcutaneous tumor was delayed and the tumor volume in 4 weeks was diminished by using Ad-p53 combined with Ad-p16 and the difference was significant compared with using Ad-p53 or Ad-p16 alone .\n\nIt was suggested that the transfer of wild-type p53 and p16 could significantly inhibit the growth of human bladder cancer in vitro and in vivo .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "A comparative study was undertaken between cancer of the uterine cervix ( n = 50 ) and female breast cancer ( n = 50 ) with reference to the expression of c-erbB-2 oncoprotein ( HER-2/neu ) and that of epidermal growth factor receptor ( EGF-R ) , both being highly homologous structurally .\n\nExpressions of EGF-R and c-erbB-2 oncoprotein were viewed in breast and cervical cancer tissues by immunochemical staining .\n\nCervical cancer cases showed much higher expression of EGF-R which also revealed significant association with the expression of c-erbB-2 oncoprotein and tumour grading .\n\nAmong breast cancer cases , over-expression of EGF-R correlated significantly with metastasis of lymph node ; and expression of c-erbB-2 oncoprotein showed a significant relationship with histological grading of the tumour .\n\nMoreover , an association was noticed between the tumour grade and the concomitant immuno positive expression of EGF-R and c-erbB-2 .\n\nOur study revealed an existence of a conflicting pattern in the expression of EGF-R and c-erbB-2 oncoprotein between carcinomas of the breast and uterine cervix .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Transforming growth factor-beta1 ( TGF-beta1 ) exerts potent immunosuppressive effects .\n\nIn this study , we investigated the potential role of TGF-beta1 produced by hepatocellular carcinoma ( HCC ) cell lines in immunosuppression mechanisms .\n\nUsing the Mv1Lu cell-growth inhibition assay and an enzyme-linked immunosorbent assay ( ELISA ) , we detected optimal levels of TGF-beta1 in the culture supernatants conditioned by the HCC cell lines PLC/PRF/5 , Hep3B , and HepG2 .\n\nTo determine the biological activity of TGF-beta1 in the supernatants , we examined the effects of the culture supernatants on the production of interferon ( IFN)-gamma induced during the culture of peripheral blood mononuclear cells ( PBMCs ) stimulated with interleukin ( IL)-12 .\n\nIFN-gamma production of IL-12-stimulated PBMCs in the 1:1 dilution of the acid-activated conditioned medium of PLC/PRF/5 , Hep3B , and HepG2 reduced to 14.7 +/- 0.8 , 17.3 +/- 9.0 , and 35.9 +/- 14.6% , respectively , compared with the value in the culture with control medium ( complete culture medium ) .\n\nThese results suggest that HCC cells producing TGF-beta1 may reduce the generation or activation of cytotoxic T lymphocytes ( CTL ) and natural killer ( NK ) cells , and thus could enhance their ability to escape immune-mediated surveillance .", "output": "Avoiding immune destruction" }, { "input": "Cell lines of human T-cell acute lymphoblastic leukemias ( T-ALL ) have gained high interest for study of mechanisms of cytostatic drug resistance .\n\nHowever , they should also be suited to examine the validity and reliability of molecular cytogenetic techniques in detecting genomic alterations in neoplastic cells .\n\nTherefore , comparative genomic hybridization ( CGH ) and 24-color-fluorescence-in-situ-hybridization ( M-FISH ) were applied to eight sublines of CCRF-CEM leukemia cells selected in vitro for drug resistance and to their drug-sensitive parental counterparts .\n\nAll cell lines were characterized by altered chromosome numbers and by a variety of chromosomal structural aberrations as shown by M-FISH .\n\nThe great majority of anomalies detected by this technique were confirmed by CGH .\n\nInterestingly , a considerable number of the rearrangements found were imbalanced .\n\nAmplifications of 5q13 in the six methotrexate-resistant cell lines , a del(9)(p21pter) in all lines examined , and a gain of chromosome 20 in 9 of the 10 lines examined were readily detected by both techniques .\n\nThe same held true for losses of chromosomes 17 and 18 in the near tetraploid cell lines which could also be confirmed by CGH .\n\nSome imbalances of genomic material detected by CGH were , however , not observed by means of M-FISH , possibly due to the limited extension of the corresponding chromosomal segment involved or the small subpopulation of cells affected .\n\nOn the other hand , reciprocal translocations , balanced isochromosomes , and small deletions remained mainly undetected by CGH .\n\nA comparison of chromosomal alterations in drug-resistant and parental cell lines showed not only amplifications of chromosomal segments harboring well-known drug resistance genes , e.g. , the dihydrofolate reductase gene , but also chromosomal changes which may involve novel genes associated with drug resistance .\n\nThus , the present study has clearly unveiled the strengths and weaknesses of both techniques which can excellently complement each other .\n\nTheir combination allowed a distinct improvement of the definition of the complex karyotypes of drug-resistant cell lines .", "output": "Genomic instability and mutation" }, { "input": "INTRODUCTION Cell-cell interaction is an essential component of atherosclerotic plaque development .\n\nActivated monocytes appear to play a central role in the development of atherosclerosis , not only through foam cell formation but also via the production of various growth factors that induce proliferation of different cell types that are involved in the plaque development .\n\nUsing serum free co-culture method , we determined the effect of monocytes on endothelial cell proliferation .\n\nMETHODS Endothelial cell proliferation is determined by the amount of [ 3H]thymidine incorporated in to the DNA .\n\nBasic fibroblast growth factor ( b-FGF ) , vascular endothelial growth factor ( VEGF ) and interleukin-8 ( IL-8 ) levels in the conditioned medium were determined by ELISA .\n\nRESULTS Conditioned medium from unactivated monocytes partially inhibited endothelial cell proliferation , whereas conditioned medium from activated monocytes promoted endothelial cell proliferation .\n\nThe mitogenic effect of conditioned medium derived from activated monocytes is due to the presence of b-FGF , VEGF and IL-8 .\n\nNeutralizing antibodies against b-FGF , VEGF and IL-8 partially reversed the mitogenic effect of conditioned medium derived from activated monocytes .\n\nWhen b-FGF , VEGF and IL-8 were immunoprecipitated from conditioned medium derived from activated monocytes , it is less mitogenic to endothelial cells .\n\nCONCLUSION Activated monocytes may play an important role in the development of atherosclerotic plaque by producing endothelial cell growth factors .", "output": "Inducing angiogenesis" }, { "input": "Previous experiments have shown that emodin is highly active in suppressing the proliferation of several tumor cell lines .\n\nHowever , it is not clear that emodin can induce growth inhibition of hepatoma cells .\n\nWe have found that emodin induces apoptotic responses in the human hepatocellular carcinoma cell lines ( HCC ) Mahlavu , PLC/PRF/5 and HepG2 .\n\nThe addition of emodin to these three cell lines led to inhibition of growth in a time- and dose-dependent manner .\n\nEmodin generated reactive oxygen species ( ROS ) in these cells which brought about a reduction of the intracellular mitochondrial transmembrane potential ( DeltaPsim ) , followed by the activation of caspase-9 and caspase-3 , leading to DNA fragmentation and apoptosis .\n\nOur findings demonstrate that ROS and the resulting oxidative stress play a pivotal role in apoptosis .\n\nPreincubation of hepatoma cell lines with the hydrogen peroxide-scavenging enzyme , catalase ( CAT ) and cyclosporin A ( CsA ) , partially inhibited apoptosis .\n\nThese results demonstrate that enhancement of generation of ROS , DeltaPsim disruption and caspase activation may be involved in the apoptotic pathway induced by emodin .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Tea ( Camellia sinensis ) is one of the most popular beverages , consumed worldwide .\n\nThe health promoting properties of tea have been attributed to its antioxidative polyphenolic constituents and their oxidative products .\n\nThe aim of the present study was to evaluate the chemopreventive efficacy of a black tea infusion on azoxymethane induced colonic preneoplastic lesions , the aberrant crypt foci in Sprague-Dawley rats .\n\nRats were injected with azoxymethane ( 15mg/kg.b.w. ) and received oral administration of 1% and 2% ( w/v ) tea infusions from the 1(st)day of carcinogen application .\n\nThe treatment was continued for 12 weeks .\n\nThe colons were then assessed for aberrant crypt foci and compared with the untreated carcinogen control group .\n\nIn situ cell proliferation and in situ apoptosis were also estimated using Brdu incorporation and the TUNEL method , respectively .\n\nAberrant crypt foci were reduced significantly ( by 44% in the 1% tea-treated and by about 40% in 2% tea-treated group ) .\n\nSignificant decrease in proliferation and increase in apoptosis suggest a possible interplay between the two processes resulting in inhibition of colon carcinogenesis by black tea .", "output": "Resisting cell death" }, { "input": "Investigation has been conducted to delineate the action of some phenolic compounds of natural origin in four human tumor cell lines : acute myeloblastic leukemia ( HL-60 ) , chronic myelogenic leukemia ( K-562 ) , breast adenocarcinoma ( MCF-7 ) and cervical epithelial carcinoma ( HeLa ) .\n\nIn cells grown in appropriate media the phenolics curcumin , yakuchinone B , resveratrol and capsaicin exhibited growth inhibition as assessed by trypan blue dye exclusion .\n\nIt was evident from the results of the MTT reduction assay and [ (3)H]thymidine incorporation into nuclear DNA that the phenolics were cytotoxic and inhibited cell proliferation .\n\nDose response studies indicated curcumin to be most cytotoxic towards HL-60 , K-562 and MCF-7 but did not show much activity in HeLa cells .\n\nOn the other hand , yakuchinone B , although less active than curcumin , displayed cytotoxicity towards all four cell lines .\n\nResveratrol was cytotoxic only in leukemic cells , while capsaicin was marginally cytotoxic .\n\nAll these phenolics did not elicit any cytotoxic activity as judged by the above parameters towards lymphocytes purified from normal human blood .\n\nWhen cells treated with phenolics were stained with propidium iodide and examined under a fluorescent microscope , characteristic apoptotic features such as chromatin condensation and nuclear fragmentation were observed .\n\nScoring of cells with apoptotic and non-apoptotic features showed positive correlation of apoptotic index with dose of phenolic , and fragmented DNA extracted free of genomic DNA displayed on gel electrophoresis a typical ladder pattern .\n\nThese phenolics which have human exposure are known cancer chemopreventive agents and their action as inducers of apoptosis in tumor cells suggest their potential use in a strategy for cancer control .", "output": "Resisting cell death" }, { "input": "In the present study we examined the effects of menadione , a redox cycling agent , on structural changes of human osteosarcoma line 143B cells .\n\nIt has been previously reported that menadione can cause necrotic or apoptotic cell death in a concentration- depending manner .\n\nIn our experimental model , cells were treated with 100 microM menadione for 24 hours .\n\nUsing electron microscopy technique cells carrying three kinds of morphological changes were detected : necrotic cells , apoptotic cells and those demonstrating a co-existence of apoptotic and necrotic features in one single cell .", "output": "Resisting cell death" }, { "input": "We examined the influence of the level of dietary protein or vitamin E ( VE ) on oxidative damage to DNA , lipids , and protein in the liver after total body irradiation ( TBI ) with X-rays at 1 or 4 Gy .\n\nLevels of 8-hydroxydeoxyguanosine , thiobarbituric acid-reactive substances , and protein carbonyls in the liver did not differ among the groups that did not receive TBI .\n\nHowever , oxidative damage to lipids and protein was increased by TBI only in the 1% protein group .\n\nDNA damage , lipid peroxidation , or protein oxidation in the liver was increased by TBI in a dose-dependent manner , and the damage was consistently higher in the 1% than in the 20% protein group .\n\nIn the 1% protein group , a greater decrease in relative spleen weight by TBI was also observed .\n\nConcentrations of antioxidants ( vitamins C and E and glutathione ) in the liver were lower and the concentration of nonheme iron in the liver was higher in the 1% than in the 20% protein group .\n\nMice fed a 1% protein diet became susceptible to TBI-induced oxidative damage , and decreases in antioxidant levels and an increase in iron level were involved in the mechanism of this susceptibility .\n\nThese results suggest that dietary VE and protein can prevent oxidative damage to DNA , lipid , and protein in mice subjected to TBI .\n\nConsumption of a VE-free diet significantly increased 8-hydroxydeoxyguanosine levels in DNA from mice fed the 1% protein diet with TBI , but such changes were not detected in DNA from mice fed the 20% protein diet .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Resveratrol , a phytoalexin found in grapes and wine , has been shown to exhibit a wide range of pharmacological properties and is believed to play a role in the chemoprevention of human cancer .\n\nResveratrol has also been shown to induce antiproliferation and apoptosis of several leukemia cell lines .\n\nIn the present study , we investigated the effect of resveratrol in adult T cell leukemia .\n\nOur present observations showed that resveratrol induced growth inhibition in all five human T cell lymphotrophic virus-1-infected cell lines examined , with 50% effective dose of 10.4-85.6 mM .\n\nIn the resveratrol-treated cells , induction of apoptosis was confirmed by annexin V-based analyses and morphological changes .\n\nThe most surprising observation was that resveratrol treatment resulted in a gradual decrease in the expression of survivin , an antiapoptotic protein , during cell apoptosis .\n\nThese findings indicate that resveratrol inhibits the growth of human T cell lymphotrophic virus-1-infected cell lines , at least in part , by inducing apoptosis mediated by downregulation in survivin expression .\n\nIn view of the accumulating evidence that survivin may be an important determinant of a clinical response in adult T cell leukemia , our present findings have led to the suggestion that resveratrol , a common constituent of the human diet , merits further investigation as a potential therapeutic agent for this incurable disease .", "output": "Resisting cell death" }, { "input": "Intra-arterial infusion with cisplatin ( CDDP ) and bleomycin ( BLM ) was carried out in 21 patients with locally recurrent uterine cervical cancer who were previously treated with irradiation alone .\n\nPatients were treated with a bolus infusion into both internal iliac arteries of 50 mg/m2 of CDDP and 30 mg/m2 of BLM .\n\nTwo to four courses of the infusion therapy were given to each patient , and the response rate , the tumor and serum drug concentrations , and the cell kinetics in tumor tissue were evaluated .\n\nThe response rate ( CR+PR ) was 71.4% according to the WHO criteria .\n\nThere was no difference , in the tumor tissue concentrations of CDDP and BLM between responders and nonresponders .\n\nAlthough the DNA ploidy of tumor cells was not significantly different between the two groups before treatment , both the labeling index with BrdU and the proliferation index with flow cytometry significantly increased 24 hours after treatment in responding tumors but not in nonresponding tumors .\n\nThese results show that intra-arterial infusion with CDDP and BLM improves the prognosis of recurrent cervical cancer and that labeling and proliferation indices may be useful for determining the response of cervical cancer to intra-arterial chemotherapy .", "output": "Sustaining proliferative signaling" }, { "input": "We have isolated rabbit kidney proximal tubular epithelial cell lines .\n\nThe selection was based on their ability to form confluent monolayers on porous supports and to maintain receptor-mediated signal transduction and ion transport , characteristic of the proximal tubule .\n\nThe isolation method consisted of several steps : ( 1 ) superficial cortical proximal tubule segments were microdissected and cultured on a matrix-coated porous support until cells formed a confluent monolayer ; ( 2 ) primary cultures showing hormone-regulated ion transport typical for the proximal tubule were selected and co-cultured with irradiated fibroblasts ; and ( 3 ) the epithelial cells surviving after several passages were expanded and passaged on porous substrates .\n\nMost of the cell lines developed in this manner were obtained by co-culture with irradiated fibroblasts producing a recombinant retrovirus encoding SV40 large T antigen and G418 resistance .\n\nHowever , SV40 T antigen expression was not essential for immortalization , since neither T antigen nor G418 resistance was detected in the isolated cell lines and co-culture with non-producing 3T3 cells gave similar results .\n\nOne cell line ( vEPT ) has been characterized in some detail with respect to morphological , biochemical , and ion transport properties .\n\nThis line forms confluent monolayers with apical microvilli , tight junctions , and convolutions of the basolateral plasma membrane .\n\nOnce confluent , monolayers maintain conductances of 25 to 32 mS/cm2 for several weeks in culture and possess phlorizin-sensitive short-circuit current ( Isc ) in glucose containing media , indicative of apical Na(+)-glucose co-transport. vEPT cells also retain receptor and signaling mechanisms for angiotensin II ( Ang II ) .\n\nApical and basal Ang II and 5,6-epoxy-eicosatrienoic acid ( 5,6-EET ) modulate the Isc in a manner similar to primary cultures .\n\nThe cell lines share with primary cultures expression of the cytokeratins K8 , K10/K11 , and K19 ( \"nomenclature \" [ 21] ) .\n\nThey also retain several receptor and signal transduction mechanisms .\n\nFor example , Ang II , arachidonate , bradykinin , 5,6-EET , parathyroid hormone ( residues 1 through 34 ) , and purine nucleotides increase cytosolic Ca2+ , PTH elevates cAMP levels , and Ang II enhances proximal tubule-specific arachidonic acid metabolism .", "output": "Enabling replicative immortality" }, { "input": "In human fibroblasts , the expression of SV40 large T antigen is known to cause a variety of chromosomal aberrations and especially dicentric chromosomes .\n\nIn some cases , the later aberrations have been reported to be reversible telomeric associations .\n\nWe report here aberration and chromosome number studies of twenty-nine T antigen positive lineages , studied from their initiation by transfection of T antigen sequences into human diploid fibroblasts , until crisis or immortalization occurred or , in some cases until the lines became tumorigenic in nude mice .\n\nThe data show that T antigen consistently produced chromosomal instability of both number and structure by an active process that began before transformation indicators were positive and continued throughout neoplastic progression .\n\nThe most frequently observed aberrations were dicentric chromosomes , which were shown to be true dicentrics by examination by in situ hybridization with telomeric sequences .\n\nThese data are consistent with the hypothesis that T antigen causes human fibroblasts to become neoplastically transformed by successive rounds of chromosomal mutation and lineage evolution .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Basal cell hyperplasia classically has been described as having bland cytologic features .\n\nDuring the past 2 years , we have seen 12 cases ( 11 in consultation ) with atypical features that were confused with adenocarcinoma of the prostate .\n\nEleven of these 12 cases contained prominent nucleoli mimicking carcinoma ; in the 12th case , nuclei were enlarged , hyperchromatic , and moderately pleomorphic .\n\nImmunohistochemistry with antibodies against high-molecular-weight cytokeratin ( 34 beta E12 ) was performed in nine of the cases , verifying their basal cell nature .\n\nAdditional findings in these cases were necrotic intraluminal secretions ( two cases ) , immature squamous metaplasia ( two cases ) , peculiar hyaline cytoplasmic globules ( two cases ) , adenosis ( one case ) , markedly atypical nuclei of uncertain nature occurring elsewhere in the specimen ( one case ) , and intraluminal blue mucin ( two cases ) .\n\nWe analyzed nine cases of typical basal cell hyperplasia , all of which showed classic features of basal cell hyperplasia with benign cytology .\n\nBoth atypical and classical basal cell hyperplasia were frequently infiltrated by lymphocytes such that the cytologic changes could not be attributable to inflammation .\n\nAtypical basal cell hyperplasia must be differentiated from ordinary adenocarcinoma of the prostate , prostatic intraepithelial neoplasia , and basaloid carcinoma ( adenoid cystic carcinoma ) of the prostate .", "output": "Tumor promoting inflammation" }, { "input": "The telomerase activity and the senescence profile of cultured breast fibroblasts from normal human interstitial and malignant stromal tissue were studied in comparison with their proliferation and differentiation pattern .\n\nFibroblasts were grown either in the presence or absence of a conditioned medium ( CM ) obtained from cultures of the oestrogen receptor-positive breast cancer MCF-7 cell line .\n\nAt different passages ( from the 2nd up to the 48th ) , fibroblasts were examined for the telomerase activity by the Telomerase Repeats Amplification Protocol ( TRAP ) assay , for proliferation profile by Ki-67 antigen expression , and the myofibroblast or smooth muscle cell-like differentiation pattern by immunofluorescence with monoclonal antibodies specific for smooth muscle markers .\n\nSerial passages of fibroblasts from normal or tumour breast reveal that the relationship between the levels of telomerase activity and phenotypic/proliferation profile changes with cell subcultivation in a different manner in the two cell populations .\n\nThe fibroblasts from normal tissue completed 12 passages in a CM-independent way prior to senescence whereas fibroblasts from tumour stroma senescence were attained after 48 passages .\n\nThese cells showed a marked decrease of telomerase activity , growth rate and smooth muscle alpha-actin expressing myofibroblasts after the 32nd passage .\n\nCM treatment of this fibroblast population induces a decline in the myofibroblast content , which precedes the changes in telomerase activity .\n\nPassaged fibroblasts from normal breast tissue can be converted to myofibroblasts upon CM treatment whereas those from tumour stroma were CM-insensitive .\n\nTaken together our data suggest that a heterogeneous fibroblast population with different life span is activated/recruited in the breast interstitium and poses the problem of a unique activation/recruitment of fibroblasts in neoplastic conditions .", "output": "Enabling replicative immortality" }, { "input": "Contradictory results have been reported regarding the association between vascularity ( used as an index of angiogenesis ) and thrombospondin-1 ( TSP-1 ) in human tumours .\n\nIn previous studies , the reported association was based on the estimated average TSP-1 value per tumour , with a sufficient number of specimens collectively analysed per tumour type .\n\nGiven the extent of intra-tumour heterogeneity , we determined the association between TSP-1 and vascularity within individual specimens , based on the average values of TSP-1 and vascularity in 10-20 pre-selected areas per tumour .\n\nCells expressing TSP-1 mRNA were visualised by in situ hybridisation and quantified by point counting .\n\nVascularity was quantified by point counting and vessel density of von Willebrand Factor-positive vessels .\n\nIn 10 ductal breast carcinomas , a direct correlation between TSP-1 and vascularity was found in 4 tumours , no correlation in 3 and an inverse correlation in 3 .\n\nThe effect of TSP-1 on endothelial cell migration in vitro was assessed in the Boyden chamber assay .\n\nTSP-1 stimulated cell migration at low concentrations ( 0.1-10 microg/ml ) and was inhibitory at high concentrations ( 25-100 microg/ml ) .\n\nThese results suggest that TSP-1 may elicit a concentration-dependent , bi-phasic , effect on angiogenesis .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "This article describes evaluation of plasma membrane fluidity and intracellular SOD with relation to apoptotic death of cervical carcinoma cells after radiation therapy .\n\nCells from biopsies of cancer patients ( stage IIIB ) prior to and 24 h after radiation dose of 2 Gy were examined .\n\nPlasma membrane fluidity , measured by fluorescence polarization of DPH incorporated into lipid bilayer and superoxide dismutase ( SOD ) activity , determined by epinephrine method , showed significant decrease but per cent apoptotic cells , as determined by annexin-V and TUNEL methods , were found increased by two folds after radiotherapy .\n\nIt is suggested that decrease in DPH polarization in membrane , reduction in SOD activity and increased apoptosis in cervical cells of cancer patients treated with radiation may be consequent to oxidative damage induced by reactive oxygen species ( ROS ) , which may have implications in developing predictive protocol in cancer radiotherapy .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Angiogenic factors produced by monocytes-macrophages are involved in the pathogenesis of chronic inflammatory disorders characterized by persistent angiogenesis .\n\nThe possibility was tested that interleukin-8 ( IL-8 ) , which is a cytokine that is chemotactic for lymphocytes and neutrophils , is also angiogenic .\n\nHuman recombinant IL-8 was potently angiogenic when implanted in the rat cornea and induced proliferation and chemotaxis of human umbilical vein endothelial cells .\n\nAngiogenic activity present in the conditioned media of inflamed human rheumatoid synovial tissue macrophages or lipopolysaccharide-stimulated blood monocytes was equally blocked by antibodies to either IL-8 or tumor necrosis factor-alpha .\n\nAn IL-8 antisense oligonucleotide specifically blocked the production of monocyte-induced angiogenic activity .\n\nThese data suggest a function for macrophage-derived IL-8 in angiogenesis-dependent disorders such as rheumatoid arthritis , tumor growth , and wound repair .", "output": "Inducing angiogenesis" }, { "input": "Vascular endothelial growth factor ( VEGF ) , also known as vascular permeability factor or vasculotropin , is a recently characterized endothelial-specific mitogen which is angiogenic in vivo .\n\nHere we demonstrate that VEGF is angiogenic in vitro : when added to microvascular endothelial cells grown on the surface of three-dimensional collagen gels , VEGF induces the cells to invade the underlying matrix and to form capillary-like tubules , with an optimal effect at approximately 2.2nM ( 100ng/ml ) .\n\nWhen compared to basic fibroblast growth factor ( bFGF ) at equimolar ( 0.5nM ) concentrations , VEGF was about half as potent .\n\nThe most striking effect was seen in combination with bFGF : when added simultaneously , VEGF and bFGF induced an in vitro angiogenic response which was far greater than additive , and which occurred with greater rapidity than the response to either cytokine alone .\n\nThese results demonstrate that like bFGF , VEGF induces an angiogenic response via a direct effect on endothelial cells , and that by acting in concert , these two cytokines have a potent synergistic effect on the induction of angiogenesis in vitro .\n\nWe suggest that the synergism between VEGF and bFGF plays an important role in the control of angiogenesis in vivo .", "output": "Inducing angiogenesis" }, { "input": "We have observed previously that treatment of plateau-phase L5178Y murine lymphoblasts in vitro with 2'-deoxycoformycin plus deoxyadenosine ( dCF/dAdo ) can inhibit the repair of X-irradiation-induced DNA single-strand breaks ( SSB ) in these cells and that this effect is associated with synergistic cell kill .\n\nIn this study we examined the effect of a combination treatment of plateau-phase L5178Y cells with bleomycin ( BLM ) plus dCF/dAdo .\n\nIncubation of BLM-treated cells with dCF/dAdo resulted in significant inhibition of the repair of BLM-induced DNA SSB .\n\nHowever , an additive , but not a synergistic , increase in cell kill was observed when cells were treated with a combination of BLM plus dCF/dAdo .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "There is increasing evidence for the implication of tumor-derived angiogenic and anti-angiogenic factors in controlling tumor growth in vivo .\n\nIn this study , we documented the production of inhibitors of angiogenesis by pancreatic cancer cells and examined how changes in the balance between pro- and anti-angiogenic factors regulate tumor growth in vivo .\n\nThe human pancreatic cancer cell line Hs-776T ( HS-W ) produces slow-growing tumors in SCID mice .\n\nCells of a variant form ( HS-R ) of Hs-776T produced faster-growing tumors compared to HS-W .\n\nCharacterization of HS-W and HS-R cells in vitro showed similar proliferation rates and production of the angiogenic factors vascular endothelial growth factor ( VEGF ) and basic fibroblast growth factor ( bFGF ) .\n\nAnalyzes of anti-angiogenic factors showed comparable levels of angiostatin and thrombospondin 1 and 2 , but endostatin was only detected in conditioned media of HS-W cells and was absent in HS-R .\n\nCell proliferation was similar in both tumor types in vivo , whereas HS-W tumors demonstrated increased apoptosis with a high percentage of apoptotic endothelial cells ( EC ) .\n\nSubsequently , VEGF was over-expressed in Hs-776T cells ( HS-VF ) , resulting in rapidly growing tumors and lowering tumor and EC apoptosis .\n\nCollectively , our study confirms that tumor growth is dependent on its ability to increase the angiogenic stimulus or to reduce the amounts of endogenous anti-angiogenic factors .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "We have examined the effect of FK506 on the Adriamycin sensitivity of the multidrug resistant human chronic myelocytic leukemia cell line ( K562/ADM ) .\n\nIn K562/ADM cells , 1.0 microgram/ml FK506 reversed the resistance of Adriamycin , and increased the IC50 value for Adriamycin up to 17 fold .\n\nHowever , IC50 value for the parent cells ( K562 ) increased only 1.5 fold .\n\nBy cell cycle analysis , the accumulation in late S-G2M phase was confirmed on K562/ADM cells , treated with 1.0 microgram/ml FK506 and low-dose of Adriamycin .\n\nCyclosporin A ( CsA ) could also restored the Adriamycin sensitivity in the K562/ADM cells , as previously reported. 1.0 microgram/ml FK506 as well as CsA significantly increased radioactive Adriamycin accumulation in K562/ADM cells and blocked [ 3H]azidopien photoaffinity labeling of P-glycoprotein .\n\nThese results suggest that 1.0 microgram/ml FK506 could reverse the Adriamycin resistance in a MDR human leukemia cells through the interaction with P-glycoprotein .", "output": "Sustaining proliferative signaling" }, { "input": "There is little understanding of the factors controlling the mobilization of blast cells from bone marrow to peripheral blood and tissues .\n\nThe aim of this study was to evaluate the soluble hepatocyte growth factor ( sHGF ) and vascular endothelial growth factor ( sVEGF ) levels in newly diagnosed patients with acute myeloid leukemia ( AML ) and to correlate these levels with the clinico-pathological features .\n\nSixty-three patients with AML and 15 normal controls were included in this study .\n\nThe levels of sHGF and sVEGF were determined by enzyme linked immunosorbent assay at diagnosis and after remission induction chemotherapy .\n\nOur results revealed significantly increased plasma levels of sHGF and sVEGF at diagnosis when compared to both control and remission levels ( P=0.000 for both ) .\n\nThe sHGF and sVEGF levels differed between AML FAB subtypes ( P=0.000 ) .\n\nThe highest concentrations were found in M5 followed by M4 .\n\nSHGF and sVEGF were directly correlated with peripheral white cell counts ( WBC ) ( r=0.836 , P=0.000 , r=0.718 ; P=0.000 , respectively ) , but inversely correlated with blast cell distribution ratio ( BCDR ) ( r=-0.785 , P=0.000 , r=-0.664 , P=0.000 , respectively ) .\n\nMoreover , both sHGF and sVEGF levels were significantly elevated in AML patients with extra-medullary infiltration as compared to those without ( P=0.000 , 0.006 , respectively ) .\n\nThe sHGF but not sVEGF levels were significantly elevated in patients who died compared to those who relapsed and to patients in complete remission ( P=0.02 , 0.08 , respectively ) .\n\nLogistic regression analysis revealed that the sHGF level at diagnoses is a powerful predictor of the patient outcome , compared to sVEGF .\n\nIn conclusion : our data support the hypothesis that angiogenic factors play a functional role in blast cell movement from the bone marrow to peripheral tissues .\n\nAssessment of sHGF at AML diagnosis is likely to be helpful in predicting patient outcome and selecting optimal therapeutic regimen .", "output": "Inducing angiogenesis" }, { "input": "Expression of angiogenic factors is upregulated in hyperplastic mucosa adjacent to colon cancer , and this upregulation is closely associated with cancer growth and metastasis .\n\nWe investigated the role of histone acetylation in vascular endothelial growth factor ( VEGF ) expression in hyperplastic mucosa adjacent to orthotopic colon cancer in mice .\n\nIn the hyperplastic mucosa adjacent to KM12SM tumor in the cecum of athymic mice , VEGF upregulation was associated with hypoxia-inducible factor ( HIF)-1alpha induction .\n\nThe hyperplastic mucosa also showed hypoacetylation of histone H4 and reduction of both p53 and von Hippel-Lindau ( VHL ) proteins .\n\nTo examine the effects of growth factors and cytokines on histone acetylation and levels of p53 , VHL and HIF-1alpha , the rat intestinal epithelial cell line IEC6 was treated with epidermal growth factor ( EGF ) and interleukin ( IL)-15 for 35 days .\n\nAcetylated histone H4 , p53 protein and ubiquitinated protein levels were reduced , whereas HIF-1alpha production was upregulated in EGF- and IL-15-treated IEC6 cells .\n\nThese findings suggest that EGF- or IL-15-induced histone H4 hypoacetylation is associated with repression of p53 and VHL genes in intestinal epithelial cells .\n\nThe subsequent suppression of protein ubiquitination leads to upregulation of VEGF production by HIF-1alpha retention .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "OBJECTIVES Epiphyseal cartilage is a barrier to osteosarcoma invasion , however the mechanisms behind this resistance remain unclear .\n\nThe aim of this study was to examine the chronological and spatial patterns of osteosarcoma growth and invasion of local tissue structures including epiphyseal cartilage .\n\nMETHODS We used an in vivomouse model of osteosarcoma to histologically examine tumors at different stages of disease progression .\n\nWe compared the pattern of osteosarcoma penetration of epiphyseal cartilage with the expression pattern of two potent mediators of angiogenesis ; proangiogenic vascular endothelial growth factor ( VEGF ) and antiangiogenic pigment epithelium-derived factor ( PEDF ) .\n\nRESULTS Epiphyseal cartilage remained intact across its entire length in all sections examined , despite increasing tumor size as well as intra- and extraosseous destruction .\n\nIn the most advanced cases , only the proangiogenic lowermost layers of the hypertrophic zone of the growth plate were eroded .\n\nThis corresponded with the growth plate layers which highly expressed the angiogenic factor VEGF .\n\nIn contrast , the resting , proliferative and upper hypertrophic layers were resistant to osteosarcoma invasion in all cases .\n\nThis corresponded to the layers with the highest expression of the potent antiangiogenic factor PEDF .\n\nCONCLUSION Epiphyseal cartilage is resistant to local invasion by osteosarcoma .\n\nThe balance of angiogenesis , influenced by pro- and antiangiogenic factors , is likely to play an important role in this resistance .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Mitochondrial dysfunction and oxidative stress have been implicated in cellular senescence , apoptosis , aging and aging-associated pathologies .\n\nTelomere shortening and genomic instability have also been associated with replicative senescence , aging and cancer .\n\nHere we show that mitochondrial dysfunction leads to telomere attrition , telomere loss , and chromosome fusion and breakage , accompanied by apoptosis .\n\nAn antioxidant prevented telomere loss and genomic instability in cells with dysfunctional mitochondria , suggesting that reactive oxygen species are mediators linking mitochondrial dysfunction and genomic instability .\n\nFurther , nuclear transfer protected genomes from telomere dysfunction and promoted cell survival by reconstitution with functional mitochondria .\n\nThis work links mitochondrial dysfunction and genomic instability and may provide new therapeutic strategies to combat certain mitochondrial and aging-associated pathologies .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Resisting cell death" }, { "input": "We have investigated the cellular requirements for IL-2 production by autocrine proliferating tumor cells from four patients with adult T cell leukemia ( ATL ) .\n\nCultures of these ATL cells both produced endogenous IL-2 protein in the absence of added mitogen and proliferated at higher levels when exogenous recombinant IL-2 was added .\n\nDepletion of macrophages in the tumor cell cultures resulted in a sharp decline in tumor cell IL-2 production , while re-addition of macrophages reconstituted this response .\n\nMacrophage-derived factors including IL-6 and IL-1 also reconstituted IL-2 production in these macrophage depleted cultures .\n\nThese results raise the possibility that macrophages may play a central role in HTLV-I mediated immortalization of T cells .", "output": "Enabling replicative immortality" }, { "input": "We present a new cell line , EJ established from an invasive endometrioid adenocarcinoma of the uterine corpus in a 56-year-old patient .\n\nThe cells show rapid growth in culture with a doubling time of 16 h and high migration activity .\n\nMonolayer-cultured cells were polygonal in shape showing a tendency to pile up without contact inhibition .\n\nSubcutaneous transplantation of the EJ cells into nude mice formed solid tumors that were histologically diagnosed as adenocarcinoma , whereas no metastasis was observed .\n\nCultured EJ cells produced tissue polypeptide antigen ( IPA ) .\n\nGenetic and molecular analyses revealed high telomerase activity but not estrogen receptor alpha expression .\n\nUsing the DNA sequencing technique , we have screened EJ cells for p53 mutation in exon 5 to 8 but no mutation of p53 was observed .\n\nThis cell line appears to represent the development of a more malignant clone with divergent receptor function and growth behavior , and provides us with an interesting new tool for the study of tumorigenesis in the human endometrium .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "We have studied the mutagenic specificity of abasic sites using the yeast oligonucleotides transformation assay .\n\nWe introduced oligonucleotide containing a natural abasic site and a tetrahydrofuran abasic site into Rev1 mutants , rev1AA , which contains mutations of Asp467 and Glu468 residues of Rev1p to Ala in order to inactivate dCMP transferase activity , and rev1 delta , which lacks its whole coding sequence .\n\nThe transformation efficiencies of rev1AA with abasic-containing oligonucleotides were lower than those of B7528 , a strain proficient in REV1 gene , but much higher than rev1 delta mutant .\n\nSequence analysis opposite the lesions showed that the mutation spectra were different among these three strains .", "output": "Genomic instability and mutation" }, { "input": "Vascular endothelial growth factor A ( VEGF-A ) and its receptor tyrosine kinases located on endothelial cells seem to play an important role in the multistep pathway of angiogenesis .\n\nSU5416 is a small molecule which inhibits angiogenesis by acting as an inhibitor of VEGF receptor-2 tyrosine kinase .\n\nWe have developed a reproducible murine model for neuroblastoma , a childhood cancer , based on s.c. xenotransplantation of SH-SY5Y neuroblastoma cells .\n\nWe found that SH-SY5Y cells expressed VEGF-A on both the mRNA and protein levels , that plasma concentrations of VEGF-A were significantly elevated in animals with neuroblastoma with a volume > 1.4 ml , and that there was a correlation between VEGF-A levels in plasma and tumor size in untreated tumor-bearing animals .\n\nTreatment with SU5416 reduced the growth of neuroblastoma tumors by 65% without apparent toxicity .\n\nSU5416 treatment also suppressed tumor angiogenesis , despite an increase in plasma VEGF-A levels per ml tumor volume during therapy .\n\nOur experimental data suggest that the angiogenesis inhibitor SU5416 may be beneficial in the treatment of solid tumors of childhood such as neuroblastoma .", "output": "Inducing angiogenesis" }, { "input": "Epidermal growth factor ( EGF ) and transforming growth factor alpha ( TGF alpha ) content was measured in normal ovaries and benign ovarian tumours .\n\nEpidermal growth factor was present in 12.7% of normal ovaries , with a range 0.030-0.533 ng/mg DNA , and in 31.8% of benign ovarian tumours , with a range 0.1335-2.080 ng/ml DNA .\n\nTGF alpha was present in 84.5% of normal ovaries , with a range of values from 0.037-18.2 ng/mg DNA , and in 84.1% of benign ovarian tumours , with a range of 0.083-195 ng/mg DNA .\n\nThe TGF alpha content in post menopausal benign ovarian tumours was significantly higher ( P < 0.0001 ) than TGF alpha in the pre-menopausal normal ovarian group .\n\nThe frequency of detection and levels of TGF alpha measured were significantly higher than those of EGF in the normal ovary group ( P < 0.001 ) and also in the benign ovarian group ( P < 0.005 ) .\n\nWe conclude that TGF alpha is the predominant growth factor present in normal ovaries and benign ovarian tumours .", "output": "Sustaining proliferative signaling" }, { "input": "FCE 23762 ( 3 ' desamino-3'[2(s)methoxyl-4-morpholinyl]doxorubicin ) is a new doxorubicin ( Dx ) derivative that has been selected for clinical testing for its favourable antitumor characteristics , which include efficacy on Dx-resistant tumors .\n\nImmunosuppression is an undesirable side-effect of anti-cancer chemotherapy and the therapeutic efficacy of Dx is probably also related to its low immunotoxicity .\n\nIt was , thus , of interest to compare the effects of FCE 23762 and its parental drug on the immune responses .\n\nBoth compounds were injected i.v. into healthy mice at equitoxic doses and according to different treatment schedules .\n\nSingle doses of FCE 23762 and Dx , given concomitant or after the antigen , suppressed at the same degree and dose-dependently the primary anti-SRBC antibody response .\n\nFollowing a multiple treatment schedule after the antigen , FCE 23762 was less suppressive than Dx on both primary and secondary antibody production .\n\nDifferently from Dx , that was completely inactive , FCE 23762 moderately inhibited DTH reaction to SRBC , only at the highest single dose tested or for repeated administrations given simultaneously or after priming .\n\nBoth drugs were totally ineffective in delaying skin allograft rejection .\n\nSince spleen cellularity and ex vivo lymphocyte proliferation to Con A and LPS were similarly impaired by the two drugs , the differentiated immunodepressive activity of FCE 23762 and Dx cannot be merely associated to their cytotoxic and antiproliferative action .\n\nThe hypothesis of a selective effect on different regulatory cell subsets and/or immune mechanisms is discussed .", "output": "Avoiding immune destruction" }, { "input": "The effects of an anti-CD3 mAb on induction of non-MHC restricted cytolysis was investigated .\n\nPeripheral blood mononuclear cells ( PBMC ) from normal donors ( 29 ) and cancer patients ( 18 ) were cultured in 100 U/ml of interleukin-2 ( rIL-2 ) with and without anti-CD3 mAb ( OKT3 , 10 ng/ml ) for the first 48 hours of incubation .\n\nThereafter , both PBMC cultures were maintained on rIL-2 up to 20 days .\n\nPBMC proliferation was enhanced 17-fold in number by day 20 when anti-CD3 mAb and rIL-2 was present during the first 48 hours but only 3-fold by day 20 when rIL-2 alone was present .\n\nConcomitantly anti-CD3 mAb but not Lym-1 , an isotype matched control , inhibited the induction of lytic activity against both NK sensitive ( K562 ) and NK resistant ( Raji ) target cell lines .\n\nThus the inhibitory effect is dependent on anti-CD3 mAb stimulating the CD3/TCR T-cell receptor complex .\n\nWhile lytic activity was dependent on the concentration of rIL-2 , inhibition of the induction phase of non-MHC restricted lytic activity was independent of the concentration of rIL-2 .\n\nFlow cytometry analysis indicated that treatment with the anti-CD3 mAb increased the percentage of CD3 positive cells , CD4 positive cells and especially CD25 positive cells , but decreased th percentage of CD56 positive cells .\n\nSupernatants from anti-CD3 mAb stimulated cultures also inhibited the induction of non-MHC restricted lytic activity .\n\nLymphokine analysis showed that supernatants of anti-CD3 mAb stimulated cultures had higher levels of TNF-alpha and IFN-gamma .\n\nHowever , TNF-alpha and IFN-gamma alone or in combination could not mediate the inhibitory effect .\n\nThe inhibitory factor(s) was partially purified by sequential chromatography on matrices of controlled pore glass and Sepharose CL-6B .\n\nThe molecular weight of the inhibitory factor(s) was less than 67K .\n\nThese studies have identified a novel regulatory pathway controlling non-MHC restricted cytolysis .\n\nPerturbation of the T-cell CD3/TCR complex with the anti-CD3 mAb results in the secretion of a soluble mediator that down-regulates the induction of rIL-2 dependent non-MHC restricted cytolysis .", "output": "Sustaining proliferative signaling" }, { "input": "The effects of high and low dietary fat ( 20% vs. 0.5% corn oil ) , and of the prostaglandin synthetase inhibitor indomethacin ( 0.005% w/w ) , on tumour incidence , tumour growth , hormone-receptor status and growth-factor expression were examined in dimethylbenzanthracene ( DMBA)-induced rat breast cancer .\n\nThe high dietary-fat group showed a significantly higher tumour incidence , larger tumour size and larger number of bromodeoxyuridine(BrdU)-positive cells of tumours as compared with those in the low dietary-fat group .\n\nIndomethacin reduced tumour incidence significantly , but conversely increased the tumour size and the number of BrdU-positive cells in both the high and the low dietary-fat groups .\n\nNo significant difference was noted in the hormone-receptor status of the tumours .\n\nGrowth factors ( TGF-alpha and IGF-II ) were somewhat highly expressed in the high dietary-fat group as compared with the low dietary-fat group , but indomethacin rather reduced the growth-factor expression .\n\nIt is concluded that high dietary fat stimulates tumour incidence and tumour proliferation , while indomethacin has dual effects : a stimulating effect on tumour proliferation , but an inhibiting effect on tumour incidence .\n\nIt is also suggested that hormone-receptor status and growth-factor expression do not play an important role in their stimulating effects on tumour proliferation .", "output": "Sustaining proliferative signaling" }, { "input": "Induction of the expression of the Mr 67,000 high-affinity laminin receptor gene has been postulated as playing a role in the progression of human tumors to invasive cancers .\n\nWe tested this hypothesis by examining histopathological sections of a large number of epithelial lesions of the genital tract associated with human papillomaviruses .\n\nIn situ hybridization was performed with a riboprobe generated from a laminin receptor complementary DNA .\n\nLaminin receptor mRNA was expressed primarily in the less differentiated cells in normal squamous tissues and in a spectrum of squamous neoplasms .\n\nThere was no net induction of mRNA per cell in intraepithelial or invasive squamous neoplasms relative to normal tissue .\n\nIn contrast , laminin receptor mRNA was not expressed at a detectable level in normal glands of the uterine cervix but was dramatically induced in morphologically abnormal , human papillomavirus-positive glands , irrespective of the genotype of human papillomaviruses present .\n\nThe induction occurred before any evidence of invasion , and there was no further increase during the transition from adenocarcinoma in situ to invasive carcinoma .\n\nWe conclude that induction of high-affinity laminin receptor gene expression is associated with the development of malignancies of cervical glandular epithelia , but the increased expression appears to correlate with the proliferative rather than the invasive properties of these cells .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Activated lymphocytes and malignant lymphoma cells derived from them ( Ki-1 positive lymphoma cells ) share similar mechanisms of proliferation .\n\nTo further examine the inhibitory role of endogenous transforming growth factor beta ( TGF beta ) in Ki-1 positive lymphoma cells , the authors studied anti-TGF beta antibodies and measured their effect on proliferation .\n\nA monoclonal antibody ( T1A5 ) prepared against a unique antigenic epitope of high molecular weight Hodgkin's TGF beta and a polyclonal rabbit antibody prepared against highly purified 25,000 D porcine platelet TGF beta 1 were used .\n\nBoth antibodies are shown here to inhibit the biological activity of Hodgkin's TGF beta and to crossreact with their respective antigens in immunoblotting .\n\nDNA synthesis by Ki-1 lymphoma cells was increased 138-fold by anti-TGF beta 1 antibody and 262-fold by anti-Hodgkin's TGF beta .\n\nExogenous TGF beta 1 suppression was completely reversed by anti-TGF beta 1 antibody and IL-2-induced proliferation was markedly potentiated ( 41 fold ) .\n\nL-428 Reed-Sternberg cells secrete physiologically active TGF beta but have fewer than 500 TGF beta receptor sites per cell ; no significant proliferative response was measured for either anti-TGF beta 1 or anti-Hodgkin's TGF beta .\n\nThese results show the suppressive effect of exogenous TGF beta 1 on indolent Ki-1 lymphoma cells and suggest that the endogenous secretion of high molecular weight physiologically active TGF beta is important in maintaining the indolent nature of this low-grade Ki-1 positive lymphoma .", "output": "Sustaining proliferative signaling" }, { "input": "Previous studies have shown that the E7 gene of human papillomavirus ( HPV ) type 16 or 18 alone was sufficient for immortalization of human foreskin epithelial cells ( HFE ) and that the efficiency was increased in cooperation with the respective E6 gene , whereas the HPV6 E6 or E7 gene was not active in HFE .\n\nTo detect weak immortalizing activities of the HPV6 genes , cells were infected with recombinant retroviruses containing HPV genes , alone and in homologous and heterologous combinations .\n\nThe HPV6 genes , alone or together ( HPV6 E6 plus HPV6 E7 ) , were not able to immortalize cells .\n\nHowever the HPV6 E6 gene , in concert with HPV16 E7 , increased the frequency of immortalization threefold over that obtained with HPV16 E7 alone .\n\nInterestingly , 6 of 20 clones containing the HPV16 E6 gene and the HPV6 E7 gene were immortalized , whereas neither gene alone was sufficient .\n\nThus , the HPV6 E6 and E7 genes have weak immortalizing activities which can be detected in cooperation with the more active transforming genes of HPV16 .\n\nAcute expression of the HPV6 and HPV16 E6 and E7 genes revealed that only HPV16 E7 was able to stimulate the proliferation of cells in organotypic culture , resulting in increased expression of the proliferative cell nuclear antigen and the formation of a disorganized epithelial layer .\n\nAdditionally , combinations of genes that immortalized HFE cells ( HPV16 E6 plus HPV16 E7 , HPV16 E6 plus HPV6 E7 , and HPV6 E6 plus HPV16 E7 ) also stimulated proliferation .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "The oncogenic potential of a human papillomavirus type 16 ( HPV16 ) variant cloned from normal human cervical keratinocytes has been tested in vitro using primary rodent epithelial cells and human cervical keratinocytes .\n\nThe HPV16 variant was able to extend the lifespan of , but failed to immortalize , human keratinocytes .\n\nIt could however cooperate with an activated ras oncogene to transform primary rodent cells .\n\nRadioimmunoprecipitation assays of the rodent cells showed that they expressed the E7 protein .\n\nDNA sequence analysis of the URR/E6/E7 and E5 regions of the HPV16 showed them to be fully functional , but a deletion in the viral E2 open reading frame was detected .\n\nThis truncated E2 only weakly stimulated transcription of the viral regulatory region .\n\nComplementation assays using the HPV16 variant and a full-length E2 enabled the cloned variant to immortalize human cervical keratinocytes with wild-type efficiency .\n\nThese results suggest that other viral gene products in addition to E6/E7 may play an important role in the in vitro immortalization of cervical keratinocytes in HPV16 and the development of cervical cancer .", "output": "Enabling replicative immortality" }, { "input": "The \" high-risk \" human papillomavirus types 16 ( HPV-16 ) and 18 ( HPV-18 ) have been etiologically implicated in the majority of human cervical carcinomas .\n\nIn these cancers , the viral DNAs are often integrated into the host genome so that expression of the E1 and the E2 genes is lost , suggesting that disruption of these regulatory genes plays an important role in carcinogenic progression .\n\nPrevious studies defining the viral genes affecting HPV-16 transformation functions have used the \" prototype \" viral genome , which was cloned from a human cervical carcinoma and later discovered to harbor a mutation in the E1 gene .\n\nIn this study , we have corrected this mutation and have evaluated the effect of mutations of either the E1 or the E2 gene on the efficiency of HPV-16 immortalization of human keratinocytes .\n\nMutation of either the E1 gene or the E2 gene in the background of a \" wild-type \" HPV-16 genome markedly increased immortalization capacity .\n\nMutations were also generated in the E2-binding sites located upstream of the P97 promoter , which directs synthesis of the viral E6 and E7 transforming genes .\n\nE2 negatively regulates the P97 promoter through binding at adjacent sites .\n\nSurprisingly , the mutation of these sites only partially relieved the negative effect of E2 on viral immortalization , implicating additional mechanisms in the E2 repression of viral immortalization functions .\n\nOur results provide genetic evidence that the E1 and E2 gene products each can repress HPV-16 immortalization and support the hypothesis that a selective growth advantage is provided by integration of the viral genome in a manner that causes the loss of expression of either E1 or E2 .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "5-HT1c receptors have been shown to act as protooncogenes in NIH 3T3 cells , inducing ligand-dependent focus formation .\n\nIn order to assess their mitogenic and oncogenic potential in a different cell system , we transfected these receptors into CCL39 hamster fibroblasts , a well-characterized growth factor-dependent cell line .\n\nCell clones expressing functional receptors were isolated and tested for ( a ) growth factor dependence of proliferation measuring thymidine incorporation in response to varying doses of serum , ( b ) the response to serotonin alone or in combination with other growth factors , and ( c ) the capacity for anchorage-independent proliferation .\n\nIn the absence or presence of serotonin , the large majority of the clones isolated showed normal morphology and normal growth factor dependence and was unable to grow in soft agar .\n\nNone of the clones showed a significant response to serotonin alone in DNA synthesis reinitiation experiments , but synergy was observed between serotonin and the tyrosine kinase activating growth factors EGF and FGF .\n\nHowever , the major part of this effect could be abolished by an antagonist of 5-HT1b receptors , which are endogenous in CCL39 cells .\n\nThe same receptor was found to mediate a significant mitogenic response to the neurotransmitter in Ha-ras-transfected cells .\n\nThe fact that 5-HT1c receptors do not readily induce a transformed phenotype in CCL39 cells clearly distinguishes them from strong dominantly acting oncogene products like RAS , SRC , or FMS .", "output": "Sustaining proliferative signaling" }, { "input": "Human papillomavirus type 16 ( HPV16 ) E6 and E7 are selectively retained and expressed in HPV16-associated human genital tumors .\n\nE6 is active in several cell culture assays , including transformation of NIH 3T3 cells , trans activation of the adenovirus E2 promoter , and cooperation with E7 to immortalize normal human keratinocytes .\n\nBiochemically , the HPV16 E6 protein has been shown to bind to tumor suppressor protein p53 in vitro and induce its degradation in a rabbit reticulocyte lysate .\n\nTo examine the relationship between the various biological activities of E6 and inactivation of p53 , we tested the abilities of dominant negative mutants of p53 to substitute functionally for E6 in the three cell culture assays .\n\nWhile wild-type p53 inhibited keratinocyte proliferation , both mouse and human mutant p53s , in conjunction with E7 , increased proliferation of the keratinocytes , resulting in generation of immortalized lines .\n\nHowever , in contrast to E6 , mutant p53 was unable to induce transformation or trans activate the adenovirus E2 promoter in NIH 3T3 cells .\n\nThese results suggest that inactivation of wild-type p53 is necessary for HPV-induced immortalization of human keratinocytes and that different or additional activities are required for E6-dependent transformation and trans activation of NIH 3T3 cells .", "output": "Enabling replicative immortality" }, { "input": "In order to determine the in vivo immune response in glioblastoma , monoclonal and polyclonal antibodies specific for inflammatory leukocytes and immunoregulatory products were utilized to stain tissue from four surgical specimens .\n\nThe more activated the inflammatory cells , the more activated the tumors appeared to be .\n\nIn the tumor with the largest infiltration ( Case 3 ) , inflammatory cells were stained for interferon-gamma , interleukin-2 , interleukin-1 beta , lymphotoxin , tumor necrosis factor-alpha , and transforming growth factor-beta .\n\nThe tumor cells also expressed interleukin-1 beta , interleukin-6 , transforming growth factor-beta , tumor necrosis factor-alpha , and prostaglandin E. In contrast , in the tumor with the least inflammatory response ( Case 1 ) , the tumor cells did not express any cytokines .\n\nExpression of cytokines by glioma cells was modest in the two cases with modest inflammatory responses .\n\nCellular inflammation , primarily consisting of T cells and macrophages with few or no B cells or natural killer cells , was two- to 15-fold greater outside the tumor than within .\n\nIn contrast to leukocytes outside the tumor , which were activated and expressing class II major histocompatibility antigens , leukocytes within the tumor parenchyma or at the tumor's edge were negative for these antigens .\n\nIn the four specimens studied here , the tumor cells themselves were also negative for class II major histocompatibility antigens .\n\nThese findings , although preliminary , suggest that inflammatory cells within gliomas are inactivated and that glioma cells may increase the expression of immunosuppressive cytokines in response to an increased lymphocyte infiltrate .\n\nThis observation , if corroborated by more extensive studies , may help to explain the failure of immune treatments in glioblastoma multiforme .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "NIH-3T3 cells are non-tumorigenic when injected into athymic mice .\n\nIf these cells are mixed with an extract of basement-membrane proteins ( matrigel ) and injected s.c. , they form locally invasive and highly vascularized tumors .\n\nCells cultured from the NIH-3T3-matrigel-induced tumors showed a transformed phenotype and lacked contact inhibition .\n\nWhen cultured in a gel of matrigel , they proliferated and formed branched and invasive colonies .\n\nIn contrast , the parental NIH-3T3 cells cultured on matrigel remained as cell aggregates and were not invasive .\n\nI.V. injections of the tumor-derived NIH-3T3 cells produced many colonies on the surface of the lungs , whereas the parental NIH-3T3 cells were not metastatic .\n\nZymographic analysis of the conditioned media obtained from both the tumor-derived and parental NIH-3T3 cells demonstrated higher amounts of the 72-kDa gelatinase ( type-IV collagenase ) enzyme in the tumor-derived cells .\n\nAlso , tumor-derived NIH-3T3 cells , but not parental NIH-3T3 cells , secreted the 92-kDa type-IV collagenase .\n\nThese studies suggest that the interaction of pre-malignant NIH-3T3 cells with extracellular matrix components may contribute to the process of tumor progression .", "output": "Inducing angiogenesis, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Mas oncogene has been shown to have focus-inducing ability in NIH 3T3 cells which are tumorigenic in vivo in nude mice .\n\nIts stable expression in a variety of cell lines conferred some angiotensin responsiveness .\n\nTo understand why mas-transfected cells exhibit a transformed phenotype and if angiotensin responsiveness plays any role in this process , we studied the growth characteristics of mas-transfected 3T3 cells and demonstrated that they lose contact inhibition , exhibit foci formation , and increased DNA synthesis even in absence of serum .\n\nOur results suggest that the transformed phenotype is due to the production of a mas receptor ligand distinct from angiotensin .", "output": "Evading growth suppressors" }, { "input": "Elevated serum levels of vasoactive intestinal peptide ( VIP ) are associated with some cases of neuroblastoma and correlate with a favorable prognosis .\n\nVIP has previously been shown in our laboratory to cause the in vitro growth inhibition and morphological differentiation of the human neuroblastoma cell line , LA-N-5 .\n\nIt is now shown that LA-N-5 cells express immunoreactive VIP and bear specific VIP receptors .\n\nAntagonism of endogenous VIP , either by competitive inhibition or receptor blockade , increased cell proliferation , suggesting that VIP is operative in normal growth regulation .\n\nIntracellular and extracellular levels of VIP were also shown to increase significantly during the retinoic acid-induced differentiation of these cells .\n\nFurthermore , a concomitant marked increase in VIP receptor expression was demonstrated with cellular differentiation .\n\nThese receptors remain functional as evidenced by a matching increase in the level of detectable cAMP generated in response to exogenous VIP .\n\nIt is concluded that VIP is a normal autoregulator of neuroblastoma cell growth and differentiation , and that retinoic acid-mediated differentiation may be , in part , due to endogenous VIP .", "output": "Sustaining proliferative signaling" }, { "input": "The mouse polyomavirus encodes a tumor-suppressor gene inactivator in its large T protein and a proto-oncogene activator in its middle T protein .\n\nWe have used site-directed mutagenesis to selectively inactivate the former function without affecting the latter .\n\nTwo mutant viruses were constructed to encode altered large T proteins that fail to bind the retinoblastoma tumor-suppressor gene product pRB , along with normal small and middle T proteins .\n\nThe pRB-binding mutants proved to be defective in immortalization of primary rat embryo fibroblasts by a variety of tests .\n\nYet they proved capable of transforming both primary and established fibroblasts in culture .\n\nMost importantly , the inability of these mutants to bind pRB had little effect on their ability to induce tumors in mice .\n\nWe conclude that induction of multiple tumor types in this system does not depend on large T-pRB interactions but rather on middle T-dependent pathways .\n\nIn addition , the ability of this virus to immortalize cells in culture is not essential to its ability to induce tumors in the animal .", "output": "Enabling replicative immortality" }, { "input": "Retrovirally induced immunosuppression may elevate the incidence of chemically induced cancers .\n\nA proposed hypothesis to explain this relationship is the increased free radical activity observed during retroviral infection and carcinogen activation .\n\nWe previously found that vitamin E retarded growth of esophageal tumors accompanied by reductions of free radical products .\n\nThis study investigated the contribution that retroviral immunosuppression has on esophageal cancer induced by the carcinogen N-nitrosomethylbenzylamine ( NMBzA ) , and the response that increased levels of dietary vitamin E has on this induced carcinogenesis .\n\nFemale C57BL/6 mice received NMBzA or vehicle ( corn oil ) i.p. weekly for 3 weeks .\n\nThen some of the mice were infected with LP-BM5 murine retrovirus and fed diets containing 30 IU vitamin E or 172 IU vitamin E/kg of diet .\n\nAs an assessment of free radical activity , exhaled ethane was measured prior to killing the animals at 26 weeks .\n\nEsophagi from the various mice groups were assessed for size and frequency of tumors .\n\nLivers homogenates were analyzed for vitamins A and E , lipid fluorescence , conjugated dienes and malondialdehyde .\n\nHepatic levels of vitamin A and E were decreased ( P < 0.05 ) and indices of lipid peroxidation were greater ( P < 0.05 ) in NMBzA-treated mice relative to controls .\n\nLipid peroxidation and serum transaminases ( ALT and AST ) were greatest in mice given NMBzA and infected with the retroviruses .\n\nIncidence of esophageal tumors were also greatest in the NMBzA-treated , immunocompromised animals .\n\nMice fed vitamin E-supplemented diets showed increased ( P < 0.05 ) hepatic concentrations of vitamin E and vitamin A , decreased activities of serum transaminases , decreased indices of lipid peroxidation , and decreased size and frequency of esophageal tumors in both the immunocompromised and non-immunocompromised mice .\n\nThese results suggest that vitamin E plays an antioxidant function that retards the incidence of esophageal cancers in immunocompromised and non-immunocompromised animals .", "output": "Avoiding immune destruction" }, { "input": "The immune status of spleen and the effect of surgical treatment in advanced gastric cancer ( AGC ) patients were evaluated by means of testing NK cell activity , T-lymphocyte subsets and circulating immune complex ( CIC ) .\n\nThe results showed ( 1 ) the significant impairment of NK cell activity and T-lymphocyte subsets , were decreased in CD3+ , CD4+ cells and increased in CD6+ cells resulting in CD4+/CD8+ cell ratio decrease in peripheral blood lymphocytes ( PBL ) , splenic venous blood lymphocytes ( SVL ) and spleen cells ( SC ) of AGC , as compared with PBL of normal population ; ( 2 ) NK cell activity or CD4+/CD8+ cell ratio of SVL and SC were significantly lower than those of PBL in AGC patients , mainly caused by marked decrease of CD4+ cells in SC ; ( 3 ) NK cell activity , CD4+ cells and CD4+/CD8+ cell ratio were significantly elevated in most of AGC cases receiving either radical gastrectomy ( R2+ ) or extensive radical gastrectomy ( R3 ) .\n\nA striking declination in CD8+ cells were found only after R3 operation ; ( 4 ) after a short period ( 10-14 days ) or a long period ( 2-4 years ) of radical gastrectomy , NK cell activity and T-lymphocyte subsets showed no significant differences between R2+ and R3 operation .\n\nAs a rule , AGC would weaken immune function of patients , and along with the development of the tumor , the immunosuppression in the spleen would be generated gradually .\n\nFor these reasons , a complete tumor resection would be necessary to improve the immunocompetence , and the combined splenectomy might be advisable if indicated .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND Although chemotherapy offers promise of increased survival for children with medulloblastoma and glioblastoma multiforme , drug resistance occurs frequently , resulting in tumor progression and death .\n\nResistance to nitrosoureas and methylating agents , which damage DNA , can be mediated by a DNA repair protein , O6-alkylguanine-DNA alkyltransferase ( AGAT ) .\n\nDepletion of this protein with alkylguanines or methylating agents , however , restores tumor cell sensitivity to the cytotoxicity of chloroethylnitrosoureas ( e.g. , carmustine [ BCNU] ) .\n\nPURPOSE This study was designed to determine whether resistance to the activity of nitrosourea ( the drug BCNU ) in BCNU-resistant human medulloblastoma ( D341 Med ) and human glioblastoma multiforme ( D-456 MG ) can be reversed by the methylating agent streptozocin and the O6-substituted guanines O6-methylguanine and O6-benzylguanine .\n\nMETHODS Xenografts were grown subcutaneously in athymic BALB/c mice .\n\nBCNU was administered as a single intraperitoneal injection at doses of 100 mg/m2 , 75 mg/m2 , or 38 mg/m2--i.e. , 1.0 , 0.75 , or 0.38 , respectively , of the dose lethal to 10% of treated animals ( LD10 ) .\n\nMice were treated intraperitoneally with a single dose of O6-benzylguanine or O6-methylguanine ( 240 mg/m2 ) or with streptozocin ( 600 mg/m2 ) daily for 4 days .\n\nResponse was assessed by tumor growth delay and tumor regression .\n\nAGAT activity in the xenografts was measured at 1 and 6 hours after pretreatment , at the time tumors were excised .\n\nRESULTS Pretreatment with O6-benzylguanine , O6-methylguanine , or streptozocin reduced AGAT activity to 4% , 25% , and 95% of control values , respectively , in D341 Med and 0% , 0% , and 25% of control values , respectively , in D-456 MG 1 hour after injection .\n\nAfter 6 hours , levels changed to 7% , 61% , and 116% of control values in D341 Med and 0% , 79% , and 21% of control values in D-456 MG , respectively .\n\nBoth D341 Med and D-456 MG xenografts were completely resistant to BCNU at its LD10 .\n\nPretreatment with O6-benzylguanine increased BCNU sensitivity in both types of xenograft .\n\nIn contrast , treatment with BCNU plus O6-methylguanine or streptozocin did not produce growth delays substantially different from those produced by BCNU alone , reflecting the more efficient depletion of AGAT by O6-benzylguanine .\n\nFollowing therapy with BCNU plus O6-benzylguanine at 0.38 LD10 , tumor regressions were seen in eight of 10 D341 Med and in all 10 D-456 MG xenografts .\n\nCONCLUSION We recommend comprehensive clinical toxicologic evaluation of combination therapy with O6-benzylguanine plus BCNU , which would allow subsequent design of phase I clinical trials .", "output": "Genomic instability and mutation" }, { "input": "Semi-conservative replication of double-stranded DNA in eukaryotic cells is an asymmetric process involving leading and lagging strand synthesis and different DNA polymerases .\n\nWe report a study to analyze the effect of these asymmetries when the replication machinery encounters alkylation-induced DNA adducts .\n\nThe model system is an EBV-derived shuttle vector which replicates in synchrony with the host human cells and carries as marker gene the bacterial gpt gene .\n\nA preferential distribution of N-methyl-N-nitrosourea ( MNU)-induced mutations in the non transcribed DNA strand of the shuttle vector pF1-EBV was previously reported .\n\nThe hypermutated strand was the leading strand .\n\nTo test whether the different fidelity of DNA polymerases synthesizing the leading and the lagging strands might contribute to MNU-induced mutation distribution the mutagenesis study was repeated on the shuttle vector pTF-EBV which contains the gpt gene in the inverted orientation .\n\nWe show that the base substitution error rates on an alkylated substrate are similar for the replication of the leading and lagging strands .\n\nMoreover , we present evidence that the fidelity of replication opposite O6-methylguanine adducts of both the leading and lagging strands is not affected by the 3 ' flanking base .\n\nThe preferential targeting of mutations after replication of alkylated DNA is mainly driven by the base at the 5 ' side of the G residues .", "output": "Genomic instability and mutation" }, { "input": "Melanocyte stimulating hormone ( alpha-MSH , alpha-melanotropin),Ac-Ser-Tyr-Ser-Met-Glu-His-Phe-Arg-Trp-Gly-Ly-Pro-Va l-NH2 , regulates melanogenesis within epidermal melanocytes of many animals .\n\nAn MSH analogue ( [ Nle4,D-Phe7]alpha-MSH ) that exhibits superpotency and prolonged biological activity has been synthesized , biologically characterized , and is presently in clinical trials to determine its possible clinical use in tanning of the skin .\n\nIt also has potential for the diagnosis , localization , and chemotherapy of melanoma .\n\nThe effects of this analogue on the growth , metastatic behavior , and invasive potential of a melanotic variant of Cloudman S-91 murine melanoma are reported here .\n\nIn an intracutaneous murine model of melanoma cell tumor growth , the analogue did not increase primary tumor growth ( size ) after the period of administration of the peptide hormone analogue and did not affect spontaneous lung metastases .\n\nSurvival times for the control and melanotropin-treated groups were similar , suggesting that overall tumor burden was not affected by treatment with the hormone analogue .\n\nLast , melanoma cell invasion through a human amniotic basement membrane in vitro was not enhanced compared to untreated cells .", "output": "Activating invasion and metastasis" }, { "input": "The involvement of signal transduction systems in the initial attachment of two murine B16 melanoma clones of differing metastatic potential to extracellular matrix components was examined to learn more of the early events in cell-matrix interaction .\n\nClones of high and low metastatic capacity attached similarly in the absence of any stimulators , exhibiting a two phase time course of attachment with 100% attachment by 60 min .\n\nA slight difference in attachment characteristics between the clones was seen in response to phorbol ester stimulation , which significantly inhibited attachment of the low metastatic clone but which had no effect on the highly metastatic clone .\n\nTotal protein kinase C activity and distribution was similar for both clones .\n\nAttachment of both clones was severely reduced , however , if intracellular calcium was elevated or intracellular calmodulin inhibited .\n\nThis study suggests that signal transduction mechanisms are involved in melanoma cell attachment to matrix proteins and offers an approach to pharmacological manipulation of these cell-matrix interactions which may be relevant to reducing metastatic spread .", "output": "Activating invasion and metastasis" }, { "input": "Normal human lung fibroblast diploid cells , WI-38 , become senescent after a definite number of divisions .\n\nVA-13 is a line of immortalized cells established by transformation of WI-38 cells by SV40 virus .\n\nTo determine whether SV40 large T ( SV40-T ) antigen is essential for this immortalization of WI-38 cells we introduced an antisense gene for T antigen into VA-13 .\n\nTwo morphologically different types of antisense transformant ( VA-AS5-8 and VA-AS37-8 ) were obtained .\n\nIn both antisense transformants the expression of T antigen was reduced by more than 70% as compared to that in the parent cells .\n\nThe morphology of the antisense transformants indicated a partial conversion to the senescent phenotype of WI-38 .\n\nThe relative number of cells in the S phase of the antisense transformants was decreased as compared to that in cultures of VA-13 and about 50% of cells were at G1/0 .\n\nThe doubling time of the transformants was prolonged to close to the doubling time of WI-38 .\n\nThe level of expression of retinoblastoma protein ( pRB ) complexed with SV40-T antigen of the antisense transformants was significantly decreased although the level of total pRB was much higher than that in VA-13 .\n\nThe pRB was present exclusively in the underphosphorylated form .\n\nThus , the decreased level of formation of the complex between SV40-T and pRB or the underphosphorylation of pRB may explain the suppression of growth of antisense transformants .\n\nTogether , these results show that an antisense gene for SV40-T antigen can efficiently block the cell proliferation and the cell immortalization of VA-13 cells .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "c-myc , c-erbB-2 , and Ki-67 expression was examined by immunohistochemistry in 11 normal breast tissues and 42 invasive and 14 noninvasive breast carcinomas .\n\nThe c-myc product was detected in all breast carcinoma specimens and in 7 of 11 normal breast tissues .\n\nInvasive tumors stained more frequently with the anti-myc monoclonal antibody than did noninvasive tumors , while the level of expression in normal breast tissue was much less than that in breast cancer .\n\nMembrane staining of the c-erbB-2 protein was demonstrated in 29% ( 4 of 14 ) of noninvasive ductal carcinomas and in 45% ( 19 of 42 ) of invasive breast carcinomas .\n\nNone of the 11 normal breast tissue samples was positive .\n\nThe mean value of Ki-67-positive cells was 0.91 +/- 0.31% for normal breast tissue , 4.57 +/- 1.36% for noninvasive ductal carcinoma , and 12.76 +/- 2.18% for invasive breast cancer .\n\nIn 42 invasive breast carcinomas , the expression of c-myc , c-erbB-2 , and Ki-67 proliferation marker were compared with lymph node status , estrogen receptor status , progesterone receptor status , and age of patients at diagnosis. c-erbB-2 overexpression and Ki-67 overexpression were identified as the only factors associated with lymph node status .\n\nWe concluded that they might be additional prognostic factors for breast carcinoma .", "output": "Sustaining proliferative signaling" }, { "input": "The modifying effects of an immunosuppressive agent , 6-mercaptopurine ( 6-MP ) , on development of focal lesions in liver cirrhosis models induced by carbon tetrachloride ( CCl4 ) or furfural were studied in male F344 rats .\n\nFeeding of 6-MP at 50 p.p.m. for 20 weeks to animals with pre-existing liver cirrhosis caused immunosuppression , and significantly enhanced the induction of gamma-glutamyltranspeptidase ( GGT)-positive foci and nodules in the CCl4 but not furfural case .\n\nGlutathione S-transferase P ( GST-P)-positive preneoplastic lesions were not affected .\n\nMoreover , phenobarbital ( PB ) also enhanced the induction of GGT-positive hepatocellular lesions only in the CCl4-induced liver cirrhosis model , no promotion influence being exerted after treatment with the non-carcinogenic furfural .\n\nThis study , therefore , suggests that 6-MP can enhance the induction of one type of preneoplastic foci and nodules and that essential differences exist between focal lesions arising in cirrhotic livers caused by CCl4 as opposed to furfural .", "output": "Avoiding immune destruction" }, { "input": "Amplification of the HER-2 ( c-erbB-2 ) gene and overexpression of the p185HER-2 gene product is found in approximately one-third of primary human breast and ovarian cancers and is associated with a poor clinical outcome of early relapse and death .\n\nThe HER-2 gene encodes a cell-surface growth factor receptor with intrinsic tyrosine kinase activity .\n\nWild-type human HER-2 has been shown to act as a potent oncogene when over-expressed in mouse fibroblasts .\n\nRecent data suggest that the mechanism by which HER-2 mediates transformation requires the interaction of the epidermal growth factor ( EGF ) receptor .\n\nTo test whether overexpression of normal human HER-2 can transform cells independently of the EGF receptor , we have introduced multiple copies of HER-2 into the EGF receptor-negative cell line , NR6 , and have performed assays for both transformation and tumorigenicity .\n\nEngineered NR6 cells that overexpress the HER-2 gene product display a highly transformed and tumorigenic phenotype as compared with control cells .\n\nAdditionally , a monoclonal antibody to the extracellular domain of the HER-2 receptor is able to inhibit the proliferation of the overexpressing cells in vitro as well as tumor growth in vivo .\n\nThis study provides clear evidence that HER-2-mediated transformation can be achieved independently of the EGF receptor .", "output": "Sustaining proliferative signaling" }, { "input": "Three normal human diploid cell strains were transfected with an activated Ha-ras oncogene ( EJ ras ) or SV40 T-antigen .\n\nMultiple clones were examined for morphological alterations , growth requirements , ability to grow under anchorage independent conditions , immortality and tumorigenicity in nude mice .\n\nClones expressing SV40 T-antigen alone or in combination with ras protein p21 were significantly radioresistant as compared with their parent cells or clones transfected with the neo gene only .\n\nThis radioresistant phenotype persisted in post-crisis , immortalized cell lines .\n\nCells transfected with EJ ras alone showed no morphological alterations nor significant changes in radiosensitivity .\n\nCell clones expressing ras and/or SV40 T-antigen showed a reduced requirement for serum supplements , an increase in aneuploidy and chromosomal aberrations , and enhanced growth in soft agar as an early cellular response to SV40 T-antigen expression .\n\nThe sequential order of transfection with SV40 T-antigen and ras influenced radio-sensitivity but not the induction of morphological changes .\n\nThese data suggest that expression of the SV40 T-antigen but not activated Ha-ras plays an important role in the radiosensitivity of human diploid cells .\n\nThe radioresistant phenotype in SV40 T transfected cells was not related to the enhanced level of genetic instability seen in pre-crisis and newly immortalized cells , nor to the process of immortalization itself .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "One of the immunosuppressive effects of both ultraviolet ( UV ) light and chemical carcinogens is to deplete Langerhans cells ( LC ) from the epidermis , suggesting that these cells play an important role in inducing immune responses to developing tumors during the early phases of carcinogenesis .\n\nRetinoids such as all-trans-retinoic acid ( RA ) are natural or synthetic derivatives of vitamin A ; RA binds to nuclear receptors in the skin , effecting transcription of a wide range of genes .\n\nTopical application of RA prevents the tumor promotor 12-O-tetradecanoylphorbol-13-acetate ( TPA ) from depleting the density of LC in murine epidermis .\n\nIn contrast , topical RA did not itself alter the normal LC density .\n\nRA also inhibited the development of TPA-induced immunosuppression to a locally applied contact sensitizer .\n\nTopical RA also prevented UV light from reducing the density of both LC and Thy-1+ dendritic epidermal cells ( Thy-1+ dEC ) .\n\nHowever , the RA treatment did not prevent local immunosuppression to the contact sensitizer from developing in response to UV irradiation .\n\nThe reasons for this are unclear , however , it is possible that RA does not inhibit some other immunosuppressive effect of UV light .\n\nTemarotene , a recently developed synthetic retinoid also inhibited UV light from reducing the LC and Thy-1+ dEC density from murine epidermis .\n\nThus part of the anti-carcinogenic activity of retinoids may be due to their ability to protect LC during the early stages of carcinogenesis .", "output": "Avoiding immune destruction" }, { "input": "Results generated by the immunohistochemical staining with PC10 , a new monoclonal antibody recognizing PCNA ( a nuclear protein associated with cell proliferation ) in formalin-fixed and paraffin-embedded tissue were compared with those of Ki-67 labeling and DNA flow cytometry in 47 consecutive non-small cell lung cancer ( NSCLC ) .\n\nPCNA reactivity was observed in all samples and confined to the nuclei of cancer cells .\n\nIts frequency ranged from 0 to 80% ( 37.7 +/- 23.6 ) and larger sized , early-staged and DNA aneuploid tumors expressed a significant higher number of PCNA-reactive cells .\n\nThe PCNA and Ki-67 labeling rates were closely correlated ( r = 0.383 , P = 0.009 ) .\n\nBy flow cytometry , we observed a good correlation among PCNA labeling and S-phase fraction ( r = 0.422 , P = .0093 ) and G1 phase ( r = 0.303 , P = .051 ) of the cell cycle .\n\nResults indicate that PCNA labeling with PC10 is a simple method for assessing the proliferative activity in formalin-fixed , paraffin-embedded tissue of NSCLC and correlates well with Ki-67 labeling and S-phase fraction of the cell cycle .", "output": "Sustaining proliferative signaling" }, { "input": "HER-2/neu gene expression , DNA ploidy and proliferation index were studied in 250 cases of breast cancer .\n\nExpression of HER-2/neu was determined by using an antibody to the HER-2/neu receptor .\n\nKi-67 antibody was used to determine the proliferation index of the breast cancers , and the Feulgen method was used to assess DNA amounts in the tumor cells .\n\nHistochemical staining was quantitated by image analysis .\n\nOf the cancers studied , 72 were positive for overexpression of HER-2/neu protein ; of these , 62 ( 86% ) possessed near-tetraploid DNA content , and 47 ( 65% ) had more than one G0G1 stem line ( polyploid ) of DNA distribution .\n\nCells from the cases negative for HER/2-neu overexpression contained DNA amounts that ranged from diploid to varying degrees of aneuploid .\n\nA significant difference in the amounts of cellular proliferation in HER-2/neu overexpressing cancers was found between those that expressed the HER-2/neu receptor on their membranes and those that exhibited mainly cytoplasmic receptors .", "output": "Sustaining proliferative signaling" }, { "input": "Reactive oxygen species ( ROS ) produced by phagocytic cells induce oxidative stress during chronic inflammation .\n\nROS play a role in the pathogenesis of a broad range of diseases including autoimmune , cardiac and neoplastic abnormalities .\n\nWe found that sera of patients with a variety of inflammatory dermatoses contain elevated levels of antibodies ( Ab ) binding to an oxidized DNA base derivative , 5-hydroxymethyl-2'deoxyuridine ( HMdU ) coupled to bovine serum albumin , as determined by the enzyme-linked immunosorbent assay .\n\nPatients with immune complex diseases and a history of neoplasm elaborated the highest titers of anti-HMdU Ab .\n\nTiters from sera of psoriatic subjects were lower than from the aforementioned groups but were still significantly elevated ( p < 0.001 ) above those of healthy controls .\n\nTreatment of inflammatory dermatoses with systemic antiinflammatory and cytotoxic drugs significantly lowered the titers [ p < 0.005 ( immune complex ) or p < 0.001 ( psoriasis and neoplastic ) diseases ] , suggesting that this assay may be of value in monitoring the response to therapy in these diseases .", "output": "Tumor promoting inflammation" }, { "input": "A panel of primary human cells and virus-transformed derivatives were tested for events that coincide with immortalization .\n\nIn all primary and precrisis cells , two proteins of 92 and 150 kDa that shared an epitope with p53 were found ; in most of their immortalized derivatives , however , they were absent .\n\nExpression of these proteins may be involved in senescence .", "output": "Enabling replicative immortality" }, { "input": "Lipoprotein lipase ( LPL ) plays a central role in normal lipid metabolism as the key enzyme involved in the hydrolysis of triglycerides present in chylomicrons and very low density lipoproteins .\n\nLPL is a member of a family of hydrolytic enzymes that include hepatic lipase and pancreatic lipase .\n\nBased on primary sequence homology of LPL to pancreatic lipase , Ser-132 , Asp-156 , and His-241 have been proposed to be part of a domain required for normal enzymic activity .\n\nWe have analyzed the role of these potential catalytic residues by site-directed mutagenesis and expression of the mutant LPL in human embryonic kidney-293 cells .\n\nSubstitution of Ser-132 , Asp-156 , and His-241 by several different residues resulted in the expression of an enzyme that lacked both triolein and tributyrin esterase activities .\n\nMutation of other conserved residues , including Ser-97 , Ser-307 , Asp-78 , Asp-371 , Asp-440 , His-93 , and His-439 resulted in the expression of active enzymes .\n\nDespite their effect on LPL activity , substitutions of Ser-132 , Asp-156 , and His-241 did not change either the heparin affinity or lipid binding properties of the mutant LPL .\n\nIn summary , mutation of Ser-132 , Asp-156 , and His-241 specifically abolishes total hydrolytic activity without disrupting other important functional domains of LPL .\n\nThese combined results strongly support the conclusion that Ser-132 , Asp-156 , and His-241 form the catalytic triad of LPL and are essential for LPL hydrolytic activity .", "output": "Genomic instability and mutation" }, { "input": "Immunosuppression of immunoglobulin synthesis seen in patients with multiple myeloma is in part due to immunosuppressive CD5 positive B cells .\n\nIn a 13 year longitudinal study of an IgA-deficient blood donor who developed multiple myeloma , the presence of immunosuppressive CD5 positive B cells and T cells preceded the diagnosis of overt multiple myeloma and the appearance of immunosuppressive monocytes .\n\nThese data argue that certain immune defects may be involved in the development of myeloma and are not simply a consequence of overt malignancy .", "output": "Avoiding immune destruction" }, { "input": "Human diploid fibroblasts have a limited life span in vitro , and spontaneous immortalization is an extremely rare event .\n\nWe have used transformation of human diploid fibroblasts by an origin-defective simian virus 40 genome to develop series of genetically matched immortal cell lines to analyze immortalization .\n\nComparison of a preimmortal transformant ( SVtsA/HF-A ) with its uncloned and cloned immortalized derivatives ( AR5 and HAL ) has failed to reveal any major alteration involving the simian virus 40 genome .\n\nKaryotypic analysis , however , demonstrated that all of the immortal cell lines in this series have alterations of chromosome 6 involving loss of the portion distal to 6q21 .\n\nThe karyotypic analysis was corroborated by DNA analyses .\n\nSouthern analysis demonstrated that only one copy of three proto-oncogene loci ( ros1 , c-myb , and mas1 ) on 6q was retained in immortal cells .\n\nPolymerase chain reaction analysis of the microsatellite polymorphism at 6q22 ( D6S87 ) showed loss of heterozygosity .\n\nIn addition , elevated expression of c-myb ( 6q22-23 ) was observed .\n\nWe hypothesize that the region at and/or distal to 6q21 plays a role in immortalization , consistent with the presence of a growth suppressor gene .", "output": "Enabling replicative immortality" }, { "input": "A new cell line , CB109 , has been established from a human glioblastoma multiforme .\n\nThe cytoskeleton was positive for glial fibrillary acidic protein , vimentin and fibronectin .\n\nHyaluronan ( HA ) and the HA-binding protein hyaluronectin ( HN ) were expressed in the cell cytoplasm and in the extracellular matrix of spheroids and plated cells .\n\nHyaluronidase did not prevent spheroid formation suggesting that HA was not involved in the cell-cell adhesion .\n\nHA precoating prevented cell adherence to the plates and favoured spheroid formation .\n\nHA was secreted in relatively large amounts into the culture medium .\n\nHigh performance liquid chromatography demonstrated that HA was in the high molecular weight form .\n\nThe rate of HN secretion by cells was very low .\n\nBasic fibroblast growth factor significantly increased the proliferation in vitro and tumour growth after grafting into nude mice .\n\nThe epidermal growth factor receptor was not expressed on cultivated CB109 cells .\n\nCytogenetic analysis showed polysomy 7 , structural rearrangement of chromosome 10 short arm and a translocation 13q13-q14 without detectable alteration of the RB gene .", "output": "Sustaining proliferative signaling" }, { "input": "Among salivary gland neoplasms are a group of rare tumors that are histologically identical to benign mixed tumors that inexplicably metastasize ; they have been called metastasizing mixed tumor ( MZMT ) of salivary glands .\n\nWe report the clinicopathologic features and flow cytometric findings for 11 cases of MZMT .\n\nAt the time of discovery of metastatic disease , the patients , six women and five men , ranged in age from 20 to 83 years .\n\nPrimary sites of involvement included the parotid gland ( eight cases ) , submandibular gland ( two cases ) , and the nasal septum ( one case ) .\n\nWith one exception , all the patients had at least a single recurrences of their primary mixed tumor , but two or more recurrences were the norm before development of metastatic foci .\n\nThe metastases were discovered from six to 52 years following the occurrence of the primary tumor .\n\nMetastatic deposits were identified in bone , lung , regional lymph nodes , skin , kidney , retroperitoneum , oral cavity , pharynx , calvarium , and central nervous system .\n\nThe metastases either occurred simultaneously with an episode of recurrent mixed tumor ( n = 5 ) or from 5 to 29 years after a recurrence ( n = 6 ) .\n\nThe treatment of the primary , recurrent , and metastatic neoplasms was surgical excision .\n\nFollow-up , ranging from 8 months to 16 years following the diagnosis of MZMT , revealed seven patients to be alive without disease ( 64% ) and two dead of causes unrelated to metastatic disease ( 18% ) .\n\nTwo patients ( 18% ) died as a direct result of metastatic tumor at 3 and 2 years after metastasis of their mixed tumors .\n\nFlow cytometric analysis revealed a diploid DNA cell population in the primary and/or metastatic tumors in nine cases .\n\nAneuploid DNA cell content was identified in two of the cases .\n\nDNA ploidy levels and cell proliferation rates were compared with those of conventional benign mixed tumors and also with malignant mixed tumors .\n\nRetrospective analysis of histologic parameters ( mitotic rate , cellular pleomorphism , infiltrative growth , vascular or lymphatic invasion ) and flow cytometric analysis failed to identify criteria to predict the development of metastasis in these neoplasms .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "The influence of cell shape on the expression of proto-oncogenes was examined in normal and malignant human cells that varied in their sensitivities to contact-inhibition of proliferation .\n\nCells were constrained into varying degrees of roundness by plating onto culture surfaces coated with different concentrations of poly(2-hydroxyethyl methacrylate ) ( poly[HEMA] ) and assayed for proliferation capacity and levels of c-myc , c-ras , c-fos , and c-fes mRNAs .\n\nProliferation of contact-inhibited normal CUA-1 fibroblasts and the variant HT-IFNr cells was highly coupled to cell shape .\n\nAs these cells became more rounded , a critical degree of roundness was reached at which proliferation ceased .\n\nIn contrast , proliferation of non-contact-inhibited malignant HT-1080 cells was independent of cell shape .\n\nNorthern analysis revealed that expression of c-myc and c-ras was highly sensitive to cell shape in the normal CUA-1 cells but not in the malignant HT-1080 or variant HT-IFNr cells .\n\nLevels of c-myc and c-ras mRNAs declined to nearly undetectable levels in CUA-1 cells at degrees of roundness that correlated with loss of proliferative ability .\n\nExpression of c-fos and c-fes oncogenes were independent of cell shape in all cells tested .\n\nQuantification of transcription rates by the nuclear run-off assay showed that shape modulation of c-myc and c-ras oncogene expression occurred at the transcriptional level .\n\nThese data suggest that changes in cell shape can modulate expression of certain oncogenes and that these changes correlate with the cell's ability to proliferate .\n\nMoreover , inability to regulate c-myc and c-ras oncogene expression is associated with loss of shape-dependent growth controls and contact inhibition but that loss of this regulation alone is not sufficient to release cells from contact-inhibited controls .", "output": "Evading growth suppressors" }, { "input": "Despite an increasing incidence of human breast cancer , its etiology remains unknown .\n\nSince some environmental chemicals are stored in human breast fat and are rodent mammary carcinogens , determining the genotoxic potential of environmental agents in this key target tissue is important .\n\nAn assay was developed for detecting genotoxic activity , as unscheduled DNA synthesis ( UDS ) , induced by chemicals and UV radiation in early passage cultures of normal human mammary epithelial cells ( HMEC ) derived from 5 different women .\n\nIn order to measure UDS in culture , reduction in the percentage of cells in S-phase was accomplished either by depriving the cells of epidermal growth factor and bovine pituitary extract or by contact inhibition of growth .\n\nCultures were incubated with test chemicals for 24 h in the presence of [ 3H]-thymidine .\n\nUDS was quantitated autoradiographically as net grains per nucleus ( nuclear grains minus cytoplasmic background , population average ) with > or = 6 net nuclear grains considered in repair for any individual cell .\n\nA positive response was observed with UV radiation , benzo(a)-pyrene , aflatoxin B1 , ethylmethanesulfonate , 1,6-dinitropyrene , 2-acetylaminofluorene , and tobacco smoke condensate but not 7,12-dimethylbenz(a)anthracene or 2,3,7,8-tetrachlorodibenzo-p-dioxin .\n\nThese results demonstrate that HMEC from all 5 women examined have the ability to metabolize a variety of environmental chemicals to DNA-reactive forms .\n\nFurthermore , some chemicals known either to cause mammary cancer in rodents or to be contaminants in human breast tissue are genotoxic in HMEC .\n\nA positive response in passage 9 cultures was observed only with direct acting agents , suggesting that HMEC may lose their metabolic capabilities in longer-term cultures .\n\nThe HMEC UDS assay may be used to address the role of environmental agents in human breast cancer by determining whether chemicals are DNA reactive or metabolized to DNA reactive species in this critical target tissue .", "output": "Genomic instability and mutation" }, { "input": "Since tumor cells are more dependent on glycolysis for energy supply than other cells , we tested whether its inhibition by 2-deoxy-D-glucose ( 2-DG ) affects tumor growth .\n\nMale Wistar rats were inoculated in the liver with tumor cells from a chemically induced colonic adenocarcinoma .\n\nFrom day 5 after inoculation 2-DG ( 400 mg/kg/24 h ) was continuously infused into the hepatic artery for 5 days ; controls received saline in the same fashion .\n\nSeven days after the end of infusion , the animals were sacrificed .\n\nA second experimental group of rats was treated with isolated liver perfusion for 30 min with oxygenated blood through the portal vein and hepatic artery simultaneously .\n\nIn the perfusate , 400 mg/kg 2-DG were added , and the rats were sacrificed at 10 days after perfusion .\n\nA first control group underwent perfusion without 2-DG , and a second control group received i.v. infusion of 2-DG ( 400 mg/kg/30 min ) for 30 min over 5 days .\n\nA nontreated control group was also added .\n\nAll animals survived the procedures .\n\nThe concentration of blood glucose increased in the rats receiving 2-DG i.v. and intraarterially but was unchanged in the other groups .\n\nThe tumor growth was significantly reduced by 2-DG in all experimental groups , with no difference between the groups .\n\nIt is therefore concluded that 2-DG is of potential interest in the treatment of malignancies .\n\nSince local application of 2-DG avoids the risk for systemic side effects , this approach should be explored further .", "output": "Cellular energetics" }, { "input": "DNA single-strand breaks and associated growth inhibition induced by the thymidylate synthase inhibitor N-(5-[N-(3,4-dihydro-2-methyl-4-oxoquinazoline-6-ylmethyl)-N -methylamino]-2 - thenoyl)-L-glutamic acid ( ICI D1694 ) were quantitated using the human ileocecal adenocarcinoma cell line , HCT-8 .\n\nThe effects of different concentrations and schedules of [ 6R,S]-5-formyltetrahydrofolate ( [ 6RS]LV ) and 2'-deoxy-thymidine ( dThd ) on drug growth inhibition and DNA damage were also evaluated .\n\nThe drug concentrations for 50% inhibition of cell growth in culture following 2-h and 72-h exposures were 0.073 and 0.003 microM , respectively .\n\nAfter a 2-h drug exposure , the occurrence of DNA single-strand breaks ( SSBs ) was time dependent .\n\nIt was detectable at 8 h and reached a maximum at about 24 h , 34 +/- 3 ( SD ) and 305 +/- 34 rad equivalents with 0.1 microM ( 50% inhibition concentration ) and 1.0 microM ( 90% inhibition concentration ) ICI D1694 , respectively .\n\nA significant level of DNA SSBs ( 101 +/- 13 rad equivalents ) was still detectable at 72 h after the 2-h treatment with 1 microM ICI D1694 .\n\nNo significant level of DNA SSBs was detected when cells were exposed simultaneously to ICI D1694 and 20 microM [ 6RS]LV .\n\nComplete rescue of drug-induced DNA SSBs could be achieved when cells were exposed to 10 microM dThd starting no later than 4 h after drug treatment .\n\nThe growth inhibition of ICI D1694 was abrogated by [ 6RS]LV in a concentration-dependent manner .\n\nComplete protection was achieved when cells were exposed simultaneously to 1 microM ICI D1694 and 5 microMs [ 6RS]LV or to 3 microMs dThd immediately after drug treatment .\n\nThe results demonstrate that : ( a ) the growth inhibition of ICI D1694 is a function of time and schedule ; ( b ) the growth inhibition is accompanied by extensive DNA single-strand breaks and slow repair ; ( c ) at 1 microM ICI D1694 , 3 microMs dThd and 5 microMs [ 6RS]LV can completely rescue cells from drug effects when dThd is added up to 4 h following drug treatment or when [ 6RS]LV is given in combination with the drug ; ( d ) interference of [ 6RS]LV with ICI D1694 action may be occurring at the level of drug uptake and at intracellular targets , while dThd interferes with the drug action at intracellular targets .", "output": "Genomic instability and mutation" }, { "input": "Adhesion molecules play an important role in the functioning of the immune system , particularly with regard to cell-cell interactions and antigen presentation .\n\nSeveral adhesion molecules are expressed on Hodgkin's disease-derived cell lines and these are important in their molecular interactions as antigen presenting cells ( APC ) .\n\nThere are no data regarding the expression of many of these adhesion molecules on Reed-Sternberg cells and its mononuclear variant ( Hodgkin's cells ( HC) ) present in pathological material .\n\nTo obtain this information we undertook an immunohistological study on material from 18 cases of Hodgkin's disease using a panel of MoAbs to examine the expression of adhesion molecules on HC .\n\nThe HC were shown to express the integrin beta 1 subfamily molecules , LFA-1 ( CD11a ) and p150,95 ( CD11c ) in high density but lacked CR3 ( CD11b ) .\n\nAll of the immunoglobulin gene superfamily adhesion molecules studied were present to some degree on HC , with ICAM-2 , in particular , showing moderate to strong expression in most cases .\n\nThe Hermes antigen CD44 was present in high density but leukosialin ( CD43 ) , another molecule present on diverse leucocyte types , was , in general , not detected on HC .\n\nThese new data showing that ICAM-1 , ICAM-2 and LFA-3 are , like LFA-1 , expressed on HC emphasize the ability of HC to act as APC .\n\nThe known adhesion molecule phenotype of the recently defined haematopoietic lineage of human dendritic cells ( DC ) is broadly similar to that of HC , perhaps supporting the hypothesis that some HC represent a malignancy of an APC ( DC ) lineage .", "output": "Avoiding immune destruction" }, { "input": "Numerous studies have reported a correlation between production of 72-kDa ( MMP-2 ) and 92-kDa ( MMP-9 ) type-IV collagenases/gelatinases and the metastatic potential of cancer cells .\n\nAn abrogating effect of tissue inhibitors of metalloproteinases ( TIMP-1 and TIMP-2 ) on metastases has also been noted .\n\nIn this report we have used sensitive enzyme-linked immunoassays to measure MMP-2 , MMP-9 , TIMP-1 and TIMP-2 levels in eight human lung-cancer cell lines which were characterized for biological behavior in nude mice .\n\nWe demonstrated that the Calu-6 and A549 cell lines with the highest metastatic , invasive and tumorigenic potential secreted the highest levels of MMP-2 .\n\nMMP-9 and TIMP-1 secretions were comparatively low in all cell lines .\n\nTIMP-2 secretion , which exceeded MMP-2 secretion for all cell lines , did not correlate with metastatic potential .\n\nTo further explore these correlations , the metastatic Calu-6 cell line was transfected with a K-rev-1 cDNA expression construct .\n\nThe K-rev revertant cell lines demonstrated a more differentiated phenotype and were less tumorigenic , invasive and metastatic in nude mice .\n\nNonetheless , the Calu-6 revertant cell lines secreted higher levels of MMP-2 than the parent cell line .\n\nIn conclusion , invasion and metastasis by lung-cancer cells requires not only enhanced MMP production , but also other less well-understood tumorigenic characteristics .\n\nThe multiplicity of factors required by cancer cells for dissemination helps to explain the minute fraction of cancer cells from a primary tumor that ever develop into a metastasis .", "output": "Activating invasion and metastasis" }, { "input": "This study aims to identify significant predictors of survival in pediatric and adolescent colorectal carcinoma .\n\nWe retrospectively analyzed our experience with 29 histologically verified cases , of which 20 were resected for cure .\n\nVariables analyzed as predictors of survival included : ( 1 ) resectability , ( 2 ) regional nodal involvement , ( 3 ) depth of invasion , ( 4 ) grade , and ( 5 ) interval from symptom onset to diagnosis .\n\nSignet ring or anaplastic lesions were considered high grade .\n\nSurvival curves were generated on both the overall group and those resected for cure .\n\nMultivariate analysis was performed on the overall group .\n\nThe median age at diagnosis was 19 years ( range , 10 to 21 ) .\n\nMedian follow-up in survivors was 4.7 years .\n\nSignet ring tumors occurred in 45% and another 24% were poorly differentiated .\n\nSeventy-six percent presented with regional lymph node metastases .\n\nThe median survival for the overall group was 16 months , whereas that for those undergoing complete resection was 33 months .\n\nIn patients undergoing resection for cure , grade ( P = .005 ) , regional nodal involvement ( P = .007 ) , and depth of invasion ( P = .03 ) were significant predictors of outcome in univariate analysis .\n\nIn the overall group these variables as well as resectability and distant metastases were significant in univariate analysis .\n\nIn multivariate analysis high-grade lesions and lymph node involvement were highly correlated , as were resectability and metastases .\n\nThus , either variable ( but not both ) of each pair added information to the multivariate model .\n\nIn patients resected for cure , positive nodes or high histological grade became the only significant predictors of survival.(ABSTRACT TRUNCATED AT 250 WORDS )", "output": "Activating invasion and metastasis" }, { "input": "The ras oncogenes alone fully transform established ( immortalized ) rodent fibroblasts in a few days , but generally transform early-passage fibroblasts only partially , unless their action is complemented by that of a nuclear , immortalizing , oncogene .\n\nHere we show that transfection of second-passage Syrian hamster embryo fibroblasts ( HEFs ) by the EJ-H-ras oncogene coupled to the neo gene , followed by selection with G418 , gives rise to apparently normal , or only slightly transformed , clonal colonies , only a few of which become established .\n\nThe study of two established clonal lines showed that they acquired only after some weeks , and stepwise , the main characteristics of full neoplastic transformation , i.e. anchorage independence , reduced requirement for serum growth factors and tumorigenicity .\n\nLater both clonal lines became increasingly tumorigenic and completely independent of exogenous growth and attachment factors , without increase in the expression of the H-ras oncogene .\n\nTransfection of one of the clones , early after its isolation , with a truncated derivative of the nuclear v-myb oncogene devoid of its transcriptional negative regulatory domain and able to partially transform chicken embryo fibroblasts [ (myb(KXANM) ] gave rise to more transformed cells , expressing both EJ-H-ras and myb(KXANM) , which became tumorigenic earlier than the controls and remained more tumorigenic later on .\n\nWith more efficient transfection techniques , numerous foci of fully transformed cells were subsequently obtained , in a few days , in cultures transfected sequentially with EJ-H-ras(neo) and myb(KXANM) and in cultures co-transfected with the two oncogenes .\n\nHighly tumorigenic , serum-independent and immortalized clones expressing both oncogenes were obtained from these cultures .\n\nHence , the truncated myb(KXANM) oncogene accelerate the stepwise transformation of unestablished HEFs by the EJ-HH-ras oncogene and , together with this oncogene , fully transforms these same cells in a single step .\n\nThe two oncogenes acting in cooperation also induce cell immortalization , but myb(KXANM) , by itself , is not an immortalizing oncogene .\n\nNo cooperation was observed between EJ-H-ras(neo) and the unaltered v-myb oncogene .", "output": "Enabling replicative immortality" }, { "input": "A number of growth factors have been implicated in the control of the proliferation of breast cancer cells and some have been reported to mediate the proliferative effects of oestradiol .\n\nMCF-7 cells were treated with growth factors in the presence and absence of oestradiol .\n\nOestradiol increased the response of cells to the proliferative effects of epidermal growth factor ( EGF ) , transforming growth factor alpha ( TGF-alpha ) and basic fibroblast growth factor ( bFGF ) .\n\nPlatelet derived growth factor ( PDGF ) and cathepsin D had no effect in the presence or absence of oestradiol while TGF-beta slightly reduced the stimulation by oestradiol .\n\nIn the absence of oestradiol , there was little effect of combinations of growth factors although the effects of bFGF and IGF-I were additive .\n\nIn the presence of oestradiol , the effects of bFGF and TGF-alpha were additive whereas bFGF acted as an IGF-I antagonist .\n\nOverall , bFGF had the greatest effect on cell proliferation although this was less marked than the previously described effect of the IGFs and insulin .\n\nThe effects of oestradiol on the sensitivity of cells to the proliferative effects of bFGF did not appear to result from regulation of bFGF receptor expression .", "output": "Sustaining proliferative signaling" }, { "input": "Previous reports showed that treatment with non-steroidal anti-inflammatory agents ( NSAIA ) can alter the growth profile of a variety of tumours .\n\nIn this study , the effect of NSAIA treatment on the growth of the primary tumour and the appearance of spontaneous pulmonary metastases , was investigated .\n\nA mammary adenocarcinoma of non-detected immunogenicity , C7HI , was grafted subcutaneously in the lateral flank of Balb/c mice .\n\nOral treatment with approximately 1 mg kg-1 day-1 piroxicam delayed both tumour growth and the growth of pulmonary metastases .\n\nSurvival of mice bearing the primary tumour was significantly lengthened by anti-inflammatory treatment .\n\nSimilarly , in separate experiments , after surgical removal of the primary tumour by day 34 after grafting , the group of mice treated orally with piroxicam also exhibited a higher survival rate than the control group .\n\nUpon surgical removal of the primary tumour 34 days after grafting , piroxicam treatment significantly decreased both the number and size of pulmonary metastases .\n\nThe results of this study lends support to the hypothesis that inhibition or modulation of inflammation may delay tumour organisation and growth .\n\nIt is suggested that piroxicam treatment may be an appropriate adjunct therapy to delay the appearance of pulmonary metastases and to increase life-expectancy in a host whose primary tumour has to be surgically removed .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "The chemotherapeutic drug cis-diamminedichloroplatinum ( II ) covalently binds to DNA resulting in a variety of adducts and cross-links which are thought to be responsible for the toxicity of the drug .\n\nWe have used the gel mobility shift assay to detect proteins which bind to DNA treated in vitro with cis-diamminedichloroplatinum ( II ) and have identified two complexes which bind with increased affinity to cis-diamminedichloroplatinum ( II)-damaged DNA .\n\nUsing monoclonal antibodies we have shown that one complex , B1 , contains human single-stranded DNA binding protein , a protein known to be involved in the in vitro repair synthesis assay of mammalian excision repair .", "output": "Genomic instability and mutation" }, { "input": "To elucidate the mechanism of the synergistic cytotoxicity of 5-fluorouracil ( 5-FU ) and cis-diamminedichloroplatinum(II) ( CDDP ) , we studied the interaction of these agents using a human squamous carcinoma cell line ( HST-1 ) .\n\nExposure to 5-FU for 24 h and to CDDP for 1 h produced a 50% inhibitory concentration of 1.0 micrograms/ml ( 7.7 microM ) and 2.5 micrograms/ml ( 8.3 microM ) , respectively .\n\nThe cytotoxic action of CDDP was augmented , and a greater than additive effect was observed when the cells were exposed to 5-FU ( 1.0 micrograms/ml ; 7.7 microM ) for 24 h before the CDDP treatment .\n\nThis synergistic activity was maximal when the interval between 5-FU and CDDP exceeded 24 h .\n\nIn contrast , the cytotoxicity of CDDP was attenuated when it preceded the exposure to 5-FU .\n\nThymidine did not alter the 5-FU-CDDP interaction .\n\nEvaluation of the kinetics of the removal of DNA interstrand cross-links , measured by alkaline elution , showed a significant reduction of this removal in the cells exposed to 5-FU followed by CDDP with a drug-free interval of 48 h , as compared with cells exposed to CDDP alone , or to 5-FU immediately followed by CDDP , although no differences were found in the formation of DNA interstrand cross-links by CDDP among these cells .\n\nNo significant differences in the accumulation of intracellular platinum were detected by atomic absorption spectrophotometry .\n\nThese findings suggest that 5-FU modulates the repair of platinum-DNA adducts , thereby potentiating the antitumor activity of CDDP .", "output": "Genomic instability and mutation" }, { "input": "The effect of quercetin , a flavonoid found in many plants , on the proliferation of human leukemic T-cells was analyzed .\n\nQuercetin reversibly blocked the cell cycle at a point 3-6 h before the start of DNA synthesis .\n\nExpression of the growth-related genes histone H4 , cyclin A and B , and p34cdc2 was suppressed in cells blocked with quercetin .\n\nComparison of the quercetin arrest points with those of the cell cycle inhibitors aphidicolin and mimosine revealed a temporal order of arrest points in G1 of quercetin , mimosine , and aphidicolin .\n\nMimosine and aphidicolin did not inhibit the expression of cyclin A or p34cdc2 , whereas all three reagents inhibited expression of cyclin B. Low concentrations of the protein inhibitor cycloheximide inhibited release of the quercetin but not the mimosine or aphidicolin block .\n\nA [ 35S]methionine-labeled M(r) 60,000 protein disappeared in quercetin-treated cells and was rapidly synthesized after removal of quercetin , suggesting the possibility that the M(r) 60,000 protein induces DNA synthesis after the cell is released from a quercetin block .\n\nThese results suggest the usefulness of quercetin in studies of the regulation of late G1 phase .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Previous studies have demonstrated that the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone ( NNK ) induced liver tumors in F344 rats but not in Syrian golden hamsters .\n\nThe aim of this study was to determine whether there was a correlation between the persistence of O6-methylguanine ( O6-mGua ) adducts and the rate of recovery of O6-methylguanine-DNA methyltransferase ( O6-mGuaT ) after depletion in the liver and susceptibility to NNK in F344 rat and Syrian golden hamster injected s.c. with NNK ( 80 mg/kg ) .\n\nThe levels of both 7-methylguanine and O6-mGua reached a maximum 24 h after NNK treatment .\n\nO6-mGua in NNK-treated rat liver was undetectable after 48 h .\n\nIn the rat , the depletion of O6-mGuaT activity occurred within 4 h following NNK treatment .\n\nA subsequent rapid recovery of enzyme activity was observed 36 h after NNK exposure .\n\nIn contrast , high levels of O6-mGua persisted in hamster liver DNA and no O6-mGuaT activity was detected up to 336 h after NNK injection .\n\nThus , the persistence of O6-mGua in hamster liver is most likely related to a lack of recovery of the O6-mGuaT .\n\nThese results suggested that factors other than O6-mGua may be determining NNK-induced hepatocarcinogenesis in rats .\n\nAn aldehyde generated by alpha-hydroxylation of NNK , 4-oxo-4-(3-pyridyl)butanal , inhibited O6-mGuaT activity in rat hepatocytes , suggesting that this aldehyde contributes to the carcinogenicity of NNK by inhibiting this repair enzyme .", "output": "Genomic instability and mutation" }, { "input": "The aim of this study was to address whether there is a fundamental difference in regulation of histone gene expression in cells that have become quiescent but retain the ability to proliferate , compared with those cells that have differentiated .\n\nWe compared multiple levels of regulation of histone gene expression during 3T3-L1 pre-adipocyte differentiation .\n\nConfluent cells induced to differentiate by treatment with insulin , dexamethasone , and isobutylmethylxanthine initially exhibited an increased proliferative response compared with cells given serum alone .\n\nThis initial differentiation response was associated with a twofold increase in both histone gene transcription and cellular histone mRNA levels , as well as with enhanced sequence-specific binding of nuclear factors to the proximal cell-cycle-regulatory element of the H4 histone promoter .\n\nTransforming growth factor beta 1 , an inhibitor of 3T3-L1 differentiation , increased both the percentage of proliferating cells and the cellular levels of histone mRNA when given in addition to serum stimulation , but no enhancement of these parameters was observed upon addition of TGF beta 1 to the differentiation treatment .\n\nInterestingly , although TGF beta 1 enhanced binding of nuclear factors to the proximal cell cycle regulatory element of the histone promoter , these protein/DNA interactions were not associated with an increase in histone transcription .\n\nOur results are consistent with the down-regulation of histone gene expression at confluency being controlled primarily at the post-transcriptional level , in contrast to an increased involvement of transcriptional down-regulation at the onset of differentiation .", "output": "Sustaining proliferative signaling" }, { "input": "Apoptosis , programmed cell death , was previously shown to be induced by the mAb anti-APO-1 ( IgG3 , kappa ) by binding to the APO-1 cell surface Ag , a new member of the nerve growth factor/TNF receptor superfamily .\n\nTo investigate the role of the Ig H chain Fc regions we compared induction of apoptosis by the original mAb IgG3 anti-APO-1 with anti-APO-1 F(ab')2 fragments and different anti-APO-1 isotypes ( IgG1 , IgG2b , IgG2a , and IgA ) isolated by sequential sublining .\n\nWe found that IgG3 was the most active isotype ; IgG1 , IgG2a , and IgA showed intermediate activity , and IgG2b and F(ab')2 were inactive .\n\nCytotoxic activity of the inactive or less active antibody preparations was fully reconstituted by protein A , anti-mouse Ig , or anti-mouse Ig F(ab')2 , respectively .\n\nThus , APO-1-mediated induction of apoptosis was dependent on efficient cross-linking of APO-1 cell surface Ag , indirectly augmented by anti-APO-1 Fc-Fc self-aggregation .\n\nBecause of their different in vitro activity we selected IgG3- , IgG2b- , and IgA anti-APO-1 to test their antitumor activity against solid human B lymphoblastoid tumors in SCID mice .\n\nThe isotypes showed a different serum half-life ( IgG3 : 9.2-10.4 days , IgG2b : 1.9-2.6 days , and IgA : 14.1-29.2 h ) and a different initial tumor localization 4 h after i.p. injection ( IgG3 around the blood vessels , IgG2b homogeneously , and IgA heterogeneously distributed in the tumor ) .\n\nAll antibody preparations induced tumor regression by induction of apoptosis , even IgG2b anti-APO-1 inactive in vitro without cross-linking .\n\nThe activity of IgA anti-APO-1 , which did not mediate complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity indicates that apoptosis may be used as the main if not the only mechanism of induction of tumor regression in vivo .\n\nAs with in vitro , IgG3 anti-APO-1 was the most effective isotype also in vivo .\n\nThis result suggests that cross-linking of APO-1 on the tumor cell surface may also be required for tumor regression by apoptosis in vivo .\n\nTaken together , our data show that selective targeting of apoptosis to tumors may be an efficient antitumor mechanism .", "output": "Resisting cell death" }, { "input": "Immunosuppression of humoral and cellular responses following chronic oral exposure to 1 , 5 , 10 , and 20 ppm N-nitrosodimethylamine ( NDMA ) was examined in CD-1 mice .\n\nMonitoring of cumulative mortality and the incidence of peritoneal ascites in animals showed an NDMA dose-related mortality and hepatotoxicity .\n\nNo visible changes in immunological parameters were noted at the 1 ppm NDMA dose .\n\nImmunosuppression of immunoglobulin M ( IgM ) antibody response by NDMA to sheep red blood cells ( SRBC ) was time-related , dose-related , and could be reversed within 30 d by removal of the chemical from the drinking water .\n\nCellular immune response , monitored by allogeneic stimulation of cells in mixed lymphocyte reaction ( MLR ) , was markedly suppressed by 10 and 20 ppm NDMA .\n\nThus , chronic exposure to NDMA , except for the low-hepatotoxic doses of nitrosamine , resulted in a marked and persistent immunosuppression of cellular and humoral responses in CD-1 mice .\n\nIn conclusion , chronic exposure to the hepatotoxic ( ascite-inducing ) doses of NDMA suppressed humoral and cellular immunity .\n\nThe persistent immunosuppression could be reversed after the removal of NDMA from the drinking water .\n\nAlthough no direct NDMA-related cancer was reported in humans , our data point to a potential epigenetic carcinogenicity of nitrosamines due to chronic immunosuppression .", "output": "Avoiding immune destruction" }, { "input": "At present , isoniazid ( INH ) is being used prophylactically to reduce the side effects of intravesical BCG therapy for superficial bladder cancer , although it is not clear whether or not this reduces the antitumor efficacy of BCG .\n\nIn this study the impact of INH treatment on the immune response after repeated intravesical BCG administration was investigated in guinea pigs .\n\nINH was given on the 3 days around each BCG instillation .\n\nWe found that the administration of INH severely impaired the immunological effects of BCG .\n\nThe induction of mononuclear cell infiltration in the bladder wall was reduced .\n\nEnlargement of the regional lymph nodes ( weight and number of cells ) , and increase of MHC Class II expression on the lymph node cells , normally observed after intravesical BCG administration , were inhibited by INH .\n\nSystemic immunity , measured by the DTH reaction in the skin to PPD , was also diminished due to the combined treatment of BCG with INH .\n\nWhen INH was administered during the last 4 of 6 BCG instillations , the immune response to BCG was still impaired .\n\nA five-fold increase of the dose of BCG did not overcome the effect of INH .\n\nINH probably did not exert a direct suppression of the immune system of the guinea pig as the DNCB skin reactivity was not influenced .\n\nAlthough INH concentrations in the urine were high at the onset of the instillation , in vitro experiments indicated that the effect of INH may not be caused by killing of the BCG organisms shortly after application in the bladder .\n\nIn conclusion , our data in guinea pigs suggest that the use of INH may impair the immune response to intravesical BCG .\n\nAs this response may be important for the antitumor effect of BCG , urologists should be cautious with the prophylactic use of INH .\n\nThe influence on the antitumor efficacy is now investigated in man .", "output": "Avoiding immune destruction" }, { "input": "Between 1965 and 1985 , 489 patients with advanced gastric cancer who were treated with gastric resection and in whom tumor cells remained after the operation were defined as cases of a \" noncurative resection. \"\n\nThe clinicopathological features and prognosis of these patients were examined and two groups were prepared : locally advanced cancer and cancer with a distant metastasis .\n\nIn locally advanced cancer cases , tumor cells remained in the neighboring organs , lymph nodes , and/or resected margins ; in those with distant metastasis , peritoneal dissemination and/or liver metastasis were present regardless of whether or not the metastasis was removed , with or without locally noncurative factors .\n\nSerosal invasion was prominent and high rates of lymph node metastasis and lymphatic involvement were evident in both groups .\n\nThe survival rate for patients with locally advanced gastric cancer was better than that of patients with distant metastasis ( P < 0.01 ) .\n\nSurvival time in patients with locally advanced cancer can be lengthened by resecting all of the primary tumor and as much of the metastatic lesions as possible , even if the surgical management is \" noncurative. \"\n\nAggressive postoperative chemotherapy for patients with distant metastasis from a gastric cancer is to be recommended .", "output": "Activating invasion and metastasis" }, { "input": "Histopathological features of the lymph node involvement were studied in 104 patients with thoracic esophageal cancer who underwent subtotal esophagectomy combined with extended radical lymph adenectomy in cervicothoracoabdominal region .\n\nMetastatic involvement was found in a total number of 503 lymph nodes from 73 patients by histologic examination .\n\nThe mean of long and short diameter was found to be less than 5mm in 125 ( 24.9% ) of these 503 nodes .\n\nThe involved area on the section was less than one third in 149 nodes ( 29.6% ) , and was significantly smaller in mediastinal lymph nodes than those in cervical or abdominal ones .\n\nSixty-seven ( 13.3% ) of 503 nodes were partially invaded by micrometastasis of 1mm or less in diameter .\n\nMicrometastasis also more frequently occurred in mediastinal nodes with a statistically significant difference .\n\nExtranodal proliferation ( ENP ) of cancer cells was found in 106 nodes ( 21.1% ) , and extranodal lymphatic and/or blood vessel invasion ( ENly , v ) was also recognized in 60 nodes ( 11.9% ) .\n\nMicrometastasis and ENP with or without ENly , v were found in 24 ( 32.9% ) and 29 ( 39.7% ) of 73 patients with positive lymph node metastasis , respectively .\n\nPostoperative survival rate in patients with micrometastasis and/or ENP with or without ENly , v was inferior to that in patients with neither of them .", "output": "Activating invasion and metastasis" }, { "input": "The cytotoxic activity of cyclosporin A ( CsA ) and the three non-immuno-suppressive CsA analogues B3-243 , WO-039 and B3-665 were studied in tumor cell lines representing both classical and atypical forms of multidrug resistance ( MDR ) : T-ALL GM3639 L100 cells selected for vincristine ( vcr ) resistance and displaying characteristics of classical MDR , including P-glycoprotein ( pgp ) expression and increased drug efflux which can be inhibited by pgp blockers ( e.g. verapamil ) , and U-1285/ADR , a small cell lung cancer ( SCLC ) cell line selected for doxorubicin resistance which lacks pgp , is insensitive to pgp-blockers and shows cross resistance to cis-platinum .\n\nAt 1 micrograms/ml CsA was the most active agent in reversing Vcr resistance in L100 cells followed by B3-243 and WO-039 , with no effect of B3-665 .\n\nParental LO cells were only marginally sensitized to Vcr by these agents .\n\nNo reversing effect of any cyclosporin was observed in the U-1285/ADR or its parental cell line .\n\nCompared to LO cells , L100 cells showed a marked hypersensitivity to CsA > B3-243 > WO-039 with B3-665 being inactive .\n\nNo collateral sensitivity was observed for cyclosporins in U-1285/ADR cells .\n\nAlthough of different magnitude , the pattern of cytotoxic activity for the different cyclosporins alone closely parallelled that of L100 cells for U-1285 , U1285/ADR and LO cells .\n\nThe results indicate that not only the collateral sensitivity in classical MDR but also the cytotoxic actions of cyclosporins per se on tumor cells alone are independent of immunosuppressive activity .\n\nThe results also suggest a structure-activity relationship for cyclosporin-induced cytotoxicity similar to , but independent of , MDR reversing activity .", "output": "Avoiding immune destruction" }, { "input": "The effect of transforming growth factor-beta 1 ( TGF-beta 1 ) and interleukin-1 beta ( IL-1 beta ) on LDL receptor in Hep G2 cells was investigated .\n\nA greater than two-fold stimulation of the binding and internalisation of [ 125I]-labelled LDL at 37 degrees C was observed after an 18-h incubation of the cells with TGF-beta 1 at 50 ng/ml and IL-1 beta at 11,700 units/ml compared with control cells .\n\nScatchard analysis of the binding of [ 125I]-labelled LDL at 4 degrees C after an 18-h incubation of the cells with 1170 units/ml IL-1 beta and 5 ng/ml TGF-beta 1 showed that they were both acting primarily by increasing LDL receptor number .\n\nThe increase in LDL receptor activity could not be attributed to an increase in cell proliferation as TGF-beta 1 at concentrations from 0.05 ng/ml to 50 ng/ml had no significant effect on either cell number or [ 3H]thymidine incorporation into DNA whilst IL-1 beta inhibited DNA synthesis by more than 80% at a concentration of 11,700 units/ml but had significant effect on cell number .\n\nCholesterol biosynthesis from [ 14C]acetate , in contrast to the stimulation of LDL receptor activity , was inhibited by approximately two-fold by incubation with TGF-beta 1 at 50 ng/ml and IL-1 beta at 11,700 units/ml .", "output": "Sustaining proliferative signaling" }, { "input": "Using monolayer cultures of clonally isolated C3 and T5 rat prostate cancer cells , we determined that acidic ( aFGF ) and basic ( bFGF ) fibroblast growth factors profoundly enhanced T5 cell thymidine incorporation with half-maximum stimulation at 0.53 and 0.35 ng/ml , respectively .\n\nIn contrast , aFGF or bFGF enhancement of C3 cell thymidine incorporation was about 5% of that of T5 cells , and effects were principally mitogen concentration independent .\n\nSaturation analyses and cross-linking studies established that both C3 and T5 cells contained high-affinity FGF receptors of 120 and 145 kilodaltons and that receptor content and Kd of C3 and T5 cells were comparable. aFGF or bFGF stimulation of T5 cell thymidine incorporation profoundly decreased as cell plating density was reduced from 1.5 x 10(5) to 1.0 x 10(4) cells/well .\n\nThe modest response of C3 cells to either aFGF or bFGF also decreased as cell plating density was reduced .\n\nBecause heparin preserves FGF biological activity and enhances bFGF binding to high-affinity FGF receptors , we examined the effect of heparin on FGF stimulation of C3 cell thymidine incorporation .\n\nWe found that changes in cell plating density and/or medium heparin concentration had variable , inconsistent effects .\n\nThese were C3 cell plating density associated and included inhibition or modest enhancement of FGF effects .\n\nBinding analyses established that high-affinity bFGF binding of C3 and T5 cells immediately prior to assessing FGF-stimulated thymidine incorporation was comparable and independent of cell plating density , implying that C3 cell FGF insensitivity was not attributable to differences in C3 and T5 cell FGF receptor content at the time of mitogen stimulation.(ABSTRACT TRUNCATED AT 250 WORDS )", "output": "Sustaining proliferative signaling" }, { "input": "MCF-7 human breast cancer cells propagated in vitro were treated with adenosine derivatives added to the culture medium .\n\nThe effects on cell proliferation , glycolysis , and glutaminolysis were investigated .\n\nOf all adenosine derivatives tested , AMP was the most efficient inhibitor of cell proliferation .\n\nIn AMP-treated cells , DNA synthesis decreased , whereas RNA and protein syntheses rose normally with time .\n\nIn terms of carbohydrate metabolism , lactate production from glucose was drastically reduced ; therefore , most of lactate produced must have been derived from glutamine .\n\nIncreases in the enzyme activities involved in glutamate degradation and in the malate-aspartate shuttle were observed .\n\nIn contrast , actual glycolytic flux rates declined , whereas key glycolytic enzyme activities increased .\n\nMetabolites such as fructose 1,6-bisphosphate and pyruvate accumulated in AMP-arrested cells .\n\nBased on the lowered NAD level in the AMP-treated cells , lactate dehydrogenase , but not malate dehydrogenase , was impaired ; thereby the whole of glycolysis was inhibited .\n\nIn compensation , glutamine catabolism was increased .\n\nNAD concentrations fell drastically because of the known inhibition of P-ribose-PP synthesis through heightened intracellular AMP levels .\n\nA hypothetical metabolic scheme to explain these results and to show how extracellular AMP may influence carbohydrate metabolism and cell proliferation is presented .", "output": "Cellular energetics" }, { "input": "Gamma-radiation , tetrandrine , bistratene A , and cisplatin were all found to induce pronounced morphological changes characteristic of apoptosis and extensive DNA fragmentation in the human BM13674 cell line 8 h after treatment .\n\nApoptosis induced in BM13674 cells by these diverse agents was markedly inhibited by 1 microM okadaic acid , a tumour promoter that inhibits protein phosphatases 1 and 2A .\n\nThis compound also inhibited the appearance of apoptosis in fresh human leukaemia cells that had been exposed to gamma-radiation .\n\nThe inhibition of apoptosis was confirmed using fluorescence microscopy and DNA gel electrophoresis .\n\nDephosphorylation of a limited number of proteins was shown to be associated with apoptosis and okadaic acid prevented these dephosphorylations .\n\nPrevious studies on the BM13674 cell line showed that an inhibitor of protein synthesis failed to prevent apoptosis in these cells .\n\nThe present data provides further support that posttranslational modification of proteins , in particular , phosphorylation/dephosphorylation status , plays an important role in inhibition/activation of programmed cell death in different human cells after exposure to several cytotoxic agents .", "output": "Resisting cell death" }, { "input": "Intracellular pH ( pHi ) plays a critical role in the entry of cells into the DNA-synthesis phase of the cell cycle .\n\nAlterations in pHi may contribute to abnormal proliferative responses such as those seen in tumorigenic cells .\n\nWe observed that alkaline stress leads to genomic transformation of Madin-Darby canine kidney ( MDCK ) cells .\n\nTransformed cells ( F cells ) form \" foci \" in culture , lack contact inhibition , and are able to migrate , typical characteristics of dedifferentiated tumorigenic cells .\n\nF cells exhibit spontaneous biorhythmicity .\n\nRhythmic transmembrane Ca2+ flux activates plasma membrane K+ channels and Na+/H+ exchange .\n\nThis leads to periodic changes of membrane voltage and pHi at about one cycle per minute .\n\nWe conclude that endogenous oscillatory activity could be a trigger mechanism for DNA synthesis , proliferation , and abnormal growth of renal epithelial cells in culture .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND Matrix metalloproteinases ( MMP ) are a gene family of zinc enzymes capable of degrading almost all of the extracellular matrix macromolecules in vivo .\n\nTheir enzymic activities are believed to be responsible for tumor invasion and metastasis .\n\nMETHODS In this study , using peroxidase-antiperoxidase method , monospecific antisera against MMP-1 ( tissue collagenase ) , MMP-2 ( type IV collagenase/72-kilodalton [ KD ] gelatinase ) , and MMP-3 ( stromelysin ) were applied to 29 squamous cell carcinomas and normal epithelium of the esophagus to identify cells synthesizing and secreting these enzymes .\n\nRESULTS Immunoreactivity of MMP-1 , -2 , and -3 was observed in small cancer nests of the deeply invasive or marginal portion of the tumor .\n\nAmong the 29 patients studied , the presence of at least one MMP was observed in 17 ( 58.6% ) .\n\nAll three enzymes were observed in six ( 20.6% ) patients , MMP-2 and -3 in five ( 17.2% ) patients , only MMP-2 in three ( 10.3% ) patients , and MMP-3 alone in three ( 10.3% ) patients .\n\nThere was a good correlation among histologic stage and tumor invasion , lymph node metastasis , and MMP expression .\n\nIn particular , expression of MMP-2 and -3 was closely related to lymph node metastasis and vascular invasion .\n\nCONCLUSIONS These results suggest that MMP , especially MMP-2 and -3 , play an important role in tumor invasion and metastasis and that analysis of MMP-2 and -3 production is useful for evaluation of malignant potential in esophageal carcinoma .", "output": "Activating invasion and metastasis" }, { "input": "Urokinase-type plasminogen activator ( u-PA ) plays an important role in tumor growth and metastasis .\n\nThe aim of this work was to study the u-PA production , in vitro and in vivo , in a transplantable murine mammary adenocarcinoma ( M3 ) , moderately metastatic to lung , and in a related tumor variant ( MM3 ) , highly metastatic to the same organ , during tumor development .\n\nAt different times post-transplantation , tumors were employed to prepare either primary cell cultures or homogenates .\n\nPA activity from conditioned media ( CM ) , cell lysates ( CLs ) and tumor homogenates ( THs ) was quantitated by means of a fibrinolytic assay .\n\nImmunoneutralization and zymographic assays were performed to identify the PA present in both tumors .\n\nPA activity in CM , CLs and THs , that was undetectable at early stages , increased significantly along the growth of M3 adenocarcinoma .\n\nSecreted PA activity in MM3 CM was measurable at early stages and consistently increased up to 37 days post-transplantation , but a marked fall of activity was found at 48 days .\n\nPA activity in MM3 THs exhibited the same enhancement and late fall found in vitro .\n\nA positive correlation was observed between tumor size and THs PA values in both tumors .\n\nThe PA present in cell cultures and THs was identified as of the u-PA type .\n\nThese results support the hypothesis that high u-PA levels are important for tumor invasion and that the stage of tumor development is a critical factor in their PA activity .", "output": "Activating invasion and metastasis" }, { "input": "Photodynamic therapy ( PDT ) of hepatic tumours has been restricted owing to the preferential retention of photosensitizers in liver tissue .\n\nWe therefore investigated interstitial tumour illumination as a means of selective PDT .\n\nA piece of colon carcinoma CC531 was implanted in the liver of Wag/Rij rats .\n\nPhotofrin was administered ( 5 mg kg-1 i.v. ) 2 days before laser illumination .\n\nTumours with a mean ( +/- s.e. ) diameter of 5.7 +/- 0.1 mm ( n = 106 , 20 days after implantation ) were illuminated with 625 nm light , at 200 mW cm-1 from a 0.5 cm cylindrical diffuser and either 100 , 200 , 400 , 800 or 1600 J cm-1 .\n\nControl groups received either laser illumination only , Photofrin only or diffuser insertion only .\n\nShort-term effects were studied on the second day after illumination by light microscopy and computer-assisted integration of the circumference of damaged areas .\n\nLong-term effects were studied on day 36 .\n\nTo determine the biochemistry of liver damage and function , serum ASAT and ALAT levels were measured on day 1 and 2 , and antipyrine clearance on day 1 .\n\nTumour and surrounding liver necrosis increased with light dose delivered ( P < 0.001 ) .\n\nBest long-term results were obtained at 800 J cm-1 with complete tumour remission in 4 out of 6 animals .\n\nNo deterioration in liver function was found .\n\nThe results of this study show the ability of interstitial PDT to cause major destruction of tumour tissue in the liver combined with minimal liver damage .", "output": "Resisting cell death" }, { "input": "The purpose of this study was to generate a selective radiosensitising effect by the intra-hepatic-arterial infusion of misonidazole ( MISO ) .\n\nMISO ( 10 mg ) was infused after transcatheter hepatic-arterial embolisation into the livers of rabbits bearing VX2 liver cancer .\n\nThis procedure was followed by 15 Gy electron irradiation .\n\nEvaluation of tumour volume and histological examination was carried out on the 7th day after treatment .\n\nThe greatest tumour response was obtained in the group which received MISO followed by radiation and was characterised by extensive fibrosis around the tumour and nearly complete tumour necrosis .\n\nLiver cell regeneration was also noted in adjacent liver tissue .\n\nThe advantages of regional infusion of MISO following hepatic-arterial embolisation are : ( 1 ) Selectivity increased radiosensitivity of liver cancer alongside very low drug concentration in the plasma .\n\n( 2 ) Reduced or absent deleterious side effects of MISO with higher tumour/normal tissue ratios of drug concentration .\n\n( 3 ) Reduced cost due to the lower dosage of MISO required for regional infusion .", "output": "Resisting cell death" }, { "input": "Nine primary pulmonary carcinomas , one metastatic carcinoma , and two malignant pleural mesotheliomas have been analysed for the expression at the mRNA level of metalloproteinases ( MPs ) and tissue inhibitors of MPs ( TIMPs ) .\n\nIn situ hybridisation showed TIMP-1 and TIMP-2 transcripts predominantly over tumour stroma and gelatinases evenly distributed over both stromal and tumour cells .\n\nWhile both TIMP-1 and TIMP-2 were expressed in non-neoplastic lungs ( NNL ) as well as in carcinomas , stromelysin 3 ( ST3 ) , 92 kDa gelatinase and interstitial collagenase were expressed only by carcinomas .\n\nExpression of these MPs by carcinomas was independent of histologic type and such tumour features as fibrosis or necrosis .\n\nThe consistent expression of ST3 by all of the carcinomas examined and absence of its expression in NNL indicates that ST3 production is likely associated with the malignant phenotype .\n\nHowever , since 92 kDa gelatinase and interstitial collagenase transcripts were found in some but not all tumour samples , their expression is not a uniform feature of pulmonary carcinomas .\n\nThe possible prognostic significance of the expression of the latter two enzymes by carcinomas remains to be established .", "output": "Resisting cell death" }, { "input": "Fifty-six previously untreated stage-I ( according to Rai ) chronic lymphocytic leukemia ( CLL ) patients were examined for their clinical data , immunological characteristics , and hormonal values .\n\nDysfunction of T and B lymphocytes was demonstrated by changed lymphocyte blastogenic response to stimulation with phytohemagglutinin ( PHA ) , concanavalin A ( ConA ) , pisum sativatum agglutinin ( PSA ) , wheat germ agglutinin ( WGA ) , recombinant interleukin 2 ( IL 2 ) , and dextran sulfate ( DxS ) ; also by decreased immunoglobulin levels ( IgG , IgA , IgE ) and increased beta 2-microglobulin ( beta 2-M ) values .\n\nSimultaneously , dysregulation of the hypothalamic-pituitary-adrenal axis , immune system integration , imbalance of sex hormones , and changes in thyroid hormones were observed in the same group of patients .\n\nDisturbed immunohormonal interactions in early-stage CLL may be responsible for the pathogenetic mechanisms in this lymphoproliferative malignancy .", "output": "Avoiding immune destruction" }, { "input": "We wanted to investigate the effects of gamma radiation on DNA single-strand breaks ( SSB ) and glutathione ( GSH ) levels in mononuclear blood cells ( MNC ) of radiotherapy technicians .\n\nDNA SSB in MNC of radiotherapy technicians who use ( 60)Co-gamma source in their works were detected by alkaline filter elution and compared to control subjects .\n\nIn addition , GSH levels were measured using the enzymatic method in MNC .\n\nBlood samples were collected from radiotherapy technicians on Monday and Friday .\n\nDNA SSB levels were found to be significantly higher in smoking controls compared to non-smoking controls .\n\nSignificant increases of 36% and 49% in DNA SSB were detected from Monday to Friday for non-smoking and smoking radiotherapy technicians , respectively .\n\nGSH levels were found to be decreased significantly from Monday to Friday .\n\nGamma-radiation resulted in increased DNA SSB levels of MNC in radiotherapy technicians throughout the working week and these breaks have been observed to be repaired at the weekend .\n\nSmoking habit caused an additional increase in the SSBs observed in radiotherapy technicians .", "output": "Genomic instability and mutation" }, { "input": "Studies were performed to test the hypothesis that urethane-induced murine lung tumors exhibit xenobiotic resistance and alterations in pulmonary cytochrome P-450 enzymes. 1,1-Dichloroethylene , naphthalene , and paraquat were administered to tumor-bearing and control mice to elicit acute lung cytotoxicity , and responses were evaluated in tumors ( papillary and solid ) , uninvolved surrounding tissue , and untreated control lung. 1,1-Dichloroethylene ( 125 mg/kg , i.p. ) and naphthalene ( 225 mg/kg , i.p. ) caused preferential necrosis of Clara cells in control lungs and uninvolved tissue of tumor-bearing lungs .\n\nIn contrast , papillary and solid tumors were both resistant to 1,1-dichloroethylene-induced cytotoxicity .\n\nParaquat ( 10 , 20 mg/kg , i.v. ) elicited Clara cell damage in control lungs and uninvolved lung tissue of tumor-bearing mice , with minor disruption of the alveolar epithelium .\n\nNeither papillary nor solid tumors sustained any apparent cell damage from paraquat .\n\nImmunoblots of P-450 enzymes confirmed constitutive expression of CYP2B1 in control lung and uninvolved lung tissue of tumor-bearing mice , but this P-450 enzyme was not detected in either adenomas or carcinomas .\n\nLung CYP1A1 was inducible by beta-naphthoflavone in non-tumor-bearing mice and uninvolved tissue of tumor-bearing mice ; however , inducibility was decreased in adenomas and abolished in carcinomas .\n\nThese results demonstrate resistance of lung tumor cells to chemically induced cytotoxicity and diminished expression of cytochrome P-450 enzymes in tumors .", "output": "Resisting cell death" }, { "input": "The growth-inhibitory effects of ketoconazole , an antifungal agent which inhibits arachidonic acid lipoxygenases and cytochrome P-450 enzymes , were tested in human colon and breast cancer cell lines .\n\nIn the serum independent HT29-S-B6 colon cell clone , ketoconazole reduced cell proliferation and [ 3H]thymidine incorporation in a dose-dependent fashion , with a 50% inhibitory concentration of approximately 2.5 microM .\n\nFlow cytometry showed an accumulation of cells in the G0-G1 phase of the cell cycle and a concomitant decrease of the percentage of cells in S phase .\n\nKetoconazole also inhibited [ 3H]thymidine incorporation in the hormone-independent breast cancer cells MDA-MB-231 and Evsa-T , with respective 50% inhibitory concentration of approximately 13 and 2 microM .\n\nThe mechanism of action of ketoconazole is unknown .\n\nHowever , another lipoxygenase inhibitor , BW755C , inhibited only weakly [ 3H]-thymidine incorporation and accumulated the cells in S and G2 .\n\nConversely , clotrimazole and SKF525A , inhibitors of cytochrome P-450 enzymes , had effects similar to those of ketoconazole on HT29-S-B6 cells whereas metronidazole and secnidazole , other azole derivatives which do not inhibit cytochrome P-450 enzymes , had no effect .\n\nThe results suggest that cytochrome P-450 enzyme(s) activity(ies) could be implicated in the antiproliferative effects of ketoconazole .", "output": "Sustaining proliferative signaling" }, { "input": "We found that the human colon cancer cell line SW480 consists of two distinct subpopulations which we have designated E-type ( epithelial ) and R-type ( round ) .\n\nPure cultures of each type were obtained by subcloning , and both have maintained their characteristic phenotypes for at least 1 year ( 40 passages ) .\n\nE-type cells are the major ( > 98% ) type in the parental SW480 cell line .\n\nThey form flat epithelial-like colonies .\n\nIn contrast , R-type cells , which constitute a minor fraction ( < 2% ) of the parental cell line , have a rounded shape and grow in clusters of piled-up cells .\n\nCompared to E-type cells or the parental SW480 cells , isolated R-type cells display decreased doubling time , loss of contact inhibition , less adhesiveness to culture plates , higher anchorage-independent growth in soft agar , and a much more aneuploid karyotype .\n\nWhen injected s.c. into nude mice , R-type cells produce much larger tumors within the same period of time than E-type cells , and the tumors are less differentiated than those produced by the E-type cells .\n\nCell fusion experiments between R-type and E-type cells revealed that the R-type phenotype is dominant , and the results suggest that this is due to one or a few genetic changes .\n\nTaken together , these findings suggest that the R-type cells represent a more malignant variant of the E-type cells .\n\nThey may be useful , therefore , for studying mechanisms involved in tumor progression .", "output": "Evading growth suppressors" }, { "input": "Many reports have emphasized the role of gastrin as a growth factor for normal gastrointestinal mucosa and gastrointestinal cancers .\n\nRecent studies have pointed out that this peptide acts also as a growth factor for the pancreatic cancer cell line AR42J .\n\nThis effect is mediated by gastrin [ cholecystokinin ( CCK)-B ] receptors .\n\nIn the present study , we investigated gastrin ( CCK-B ) receptor expression in the azaserine-induced rat pancreatic carcinoma DSL-6 , comparing it to normal rat pancreas , and we also characterized CCK receptor subtypes in this tumor .\n\nThe results showed that there is extensive gastrin binding to the DSL-6 pancreatic carcinoma .\n\nNo evidence of specific gastrin binding to normal pancreas was found .\n\nAnalysis of the ability of gastrin-17-I to inhibit 125I-gastrin-I binding demonstrated that gastrin bound to a single class of receptors with a Kd of 0.21 +/- 0.04 nM and a binding capacity of 184 +/- 29 fmol/mg protein. 125I-Gastrin-I binding was inhibited by the specific CCK-B receptor antagonist L365,260 approximately 40 times more effectively than by the specific CCK-A receptor antagonist L364,718 .\n\nAnalysis of the ability of cholecystokinin octapeptide ( CCK-8 ) to inhibit 125I-Bolton-Hunter-CCK-8 binding revealed two CCK binding sites , i.e. , a high affinity site and a low affinity site .\n\nThe observed binding affinities of CCK-8 were then introduced into the computer analysis of the dose-inhibition curve of the ability of gastrin-17-I to inhibit binding of 125I-Bolton-Hunter-CCK-8 , which was significantly better fit by a three-site model than by a two-site model .\n\nThe three sites meet the criteria for CCK-B , high affinity CCK-A , and low affinity CCK-A receptors .\n\nThe binding capacity of CCK-B receptors constitutes 34% of the total high affinity CCK binding sites .\n\nThis study demonstrated that DSL-6 pancreatic carcinoma expresses three subtypes of CCK receptors .\n\nGastrin ( CCK-B ) receptors , which were not detected in normal rat pancreas , constitute about one third of the total high affinity CCK receptors .\n\nWe suggest that novel expression of gastrin ( CCK-B ) receptors may be generated by gene mutation or amplification during carcinogenesis and may play an important role in promoting tumor growth .", "output": "Sustaining proliferative signaling" }, { "input": "We studied the effects of double-strand breaks on intramolecular extrachromosomal homologous recombination in mammalian cells .\n\nPairs of defective herpes thymidine kinase ( tk ) sequences were introduced into mouse Ltk- cells on a DNA molecule that also contained a neo gene under control of the SV40 early promoter/enhancer .\n\nWith the majority of the constructs used , gene conversions or double crossovers , but not single crossovers , were recoverable .\n\nDNA was linearized with various restriction enzymes prior to transfection .\n\nRecombination events producing a functional tk gene were monitored by selecting for tk-positive colonies .\n\nFor double-strand breaks placed outside of the region of homology , maximal recombination frequencies were measured when a break placed the two tk sequences downstream from the SV40 early promoter/enhancer .\n\nWe observed no relationship between recombination frequency and either the distance between a break and the tk sequences or the distance between the tk sequences .\n\nThe quantitative effects of the breaks appeared to depend on the degree of homology between the tk sequences .\n\nWe also observed that inverted repeats recombined as efficiently as direct repeats .\n\nThe data indicated that the breaks influenced recombination indirectly , perhaps by affecting the binding of a factor(s) to the SV40 promoter region which in turn stimulated or inhibited recombination of the tk sequences .\n\nTaken together , we believe that our results provide strong evidence for the existence of a pathway for extrachromosomal homologous recombination in mammalian cells that is distinct from single-strand annealing .\n\nWe discuss the possibility that intrachromosomal and extrachromosomal recombination have mechanisms in common .", "output": "Genomic instability and mutation" }, { "input": "We analyzed several factors which could influence the immunogenicity of colon tumor cells , using a series of clones derived from a single chemically induced rat adenocarcinoma cell line .\n\nThese clones display variable tumorigenic potential in syngeneic immunocompetent animals , and it has been established that in this model the tumorigenicity of the cells depends on their ability to escape immune surveillance .\n\nThe results show an absence of relationship between tumorigenicity and expression of MHC-class-I antigens , cell adhesion to rat fibroblasts or fibroblast extracellular matrix .\n\nThe secretion of latent and active TGF beta I appeared to be quite variable from one clone to the other , but was unrelated to tumorigenicity .\n\nUnexpectedly , some regressive clones produced elevated levels of this cytokine , suggesting that in this model , spontaneous secretion of TGF beta I is not sufficient to impair the immune system of the host .\n\nIn contrast , the more tumorigenic clones were more resistant than less tumorigenic ones to cytotoxicity mediated by NK or LAK cells .\n\nThey also showed arrest of cell proliferation after reaching confluence , something not observed in the less tumorigenic clones .\n\nFinally , the strongest relationship with tumorigenicity was found for expression of blood-group carbohydrate antigens .\n\nIncreased expression of blood-group-H antigen and , conversely , decreased expression of beta-galactoside precursors of this antigen correlated with increased tumorigenicity .", "output": "Avoiding immune destruction" }, { "input": "Hepatocyte growth factor ( HGF ) , a mesenchymal-derived factor which regulates growth , motility , and morphogenesis of epithelial and endothelial cells , functions as a hepatotrophic and renotrophic factor for regeneration of the liver and kidney .\n\nWe have now obtained evidence that transforming growth factor-beta 1 ( TGF-beta 1 ) and glucocorticoids are negative regulators for HGF gene expression .\n\nWhen TGF-beta 1 or dexamethasone was added to cultures of MRC-5 human embryonic lung fibroblasts and HL-60 human promyelocytic leukemic cells , the amount of HGF secreted into the culture medium was inhibited to 30-40% of that of control cultures by 10 ng/ml TGF-beta 1 and to 40-50% by 10(-6) M dexamethasone .\n\nThe inhibitory effect of TGF-beta 1 and dexamethasone on HGF synthesis in MRC-5 cells was additive , thereby suggesting that TGF-beta 1 and dexamethasone exert effects through distinct mechanisms .\n\nHydrocortisone also inhibited HGF synthesis with the same potency as dexamethasone ; however , testosterone , estriol , and beta-estradiol had no effect .\n\nThe rate of HGF synthesis in MRC-5 cells , as measured by pulse labeling with [ 35S]methionine and subsequent immunoprecipitation , was suppressed to 30-40% of the control with 10 ng/ml TGF-beta 1 , and to 30-45% by 10(-6) M dexamethasone .\n\nHGF mRNA levels in MRC-5 cells and HL-60 cells were dose-dependently suppressed by TGF-beta 1 and dexamethasone ; 10 ng/ml TGF-beta 1 suppressed HGF mRNA levels to 32% and 35% of control culture , respectively , in MRC-5 cells and HL-60 cells , and 10(-6) M dexamethasone suppressed to 43% and 38% , respectively .\n\nThus , TGF-beta 1 and glucocorticoids seem to inhibit HGF synthesis by suppressing the expression of the HGF gene .\n\nWe propose that a negative regulation of HGF gene expression by TGF-beta 1 or glucocorticoids may be involved in physiological or pathological processes during tissue regeneration .", "output": "Sustaining proliferative signaling" }, { "input": "Peptide 11 , CDPGYIGSR-NH2 , is a segment of laminin which blocks tumor cell invasion .\n\nA high affinity laminin receptor in tumor cells is thought to be blocked by the carboxyl-terminal YIGSR , and conformational energy calculations suggest that the glycine in YIGSR allows an important conformational bend .\n\nWe replaced the YIGSR glycine residue in peptide 11 with either D-alanine or L-alanine to allow or disfavor the proposed glycine bend .\n\nWe found the Gly7-->D-Ala7 analog to be equal to peptide 11 in inhibiting tumor cell invasion of basement membrane matrix .\n\nThe Gly7-->L-Ala7 analog was much less capable of invasion inhibition .\n\nTwo-dimensional 1H-1H NMR was used to study the solution conformations of the peptide 11 analogs .\n\nNOESY experiments revealed close NH-NH contacts in peptide 11 and the D-Ala7 analog , but not in the L-Ala7 analog .\n\nMolecular dynamics generated low energy structures with excellent NOE agreement for peptide 11 and its analogs .\n\nBoth peptide 11 and the D-Ala7 analog , but not the less active L-Ala7 analog , were predicted to have similar bends around Gly7 or D-Ala7 .\n\nThese results suggest that a bend in the YIGSR region of peptide 11 may be important for the binding of laminin to its metastasis-associated receptor .", "output": "Activating invasion and metastasis" }, { "input": "Many breast cancers are estrogen independent , and even in patients who initially respond to estrogen suppression therapy , the regression is often temporary .\n\nWe have recently shown that antagonists of bombesin and gastrin-releasing peptide , including RC-3095 , inhibit the growth of pancreatic , colonic , and prostatic cancers in experimental animals .\n\nThis effect was associated with a substantial decrease in epidermal growth factor ( EGF ) receptor levels in pancreatic and colon cancers .", "output": "Sustaining proliferative signaling" }, { "input": "OBJECTIVE To study the effects of angiostatin(AS) gene mediated by liposome on human pancreatic cancer cell line SW1990 .\n\nMETHODS Angiostatin gene was cloned into the eukaryotic expression vector pRC/CMV .\n\nThe recombinant of pRC/CMV-AS was introduced into the pancreatic cancer cell line , SW1990 .\n\nThe mechanism of anti-tumor was studied and tested .\n\nRESULTS The eukaryotic expression vector pRC/CMV-AS was identified by the restriction digest. pRC/CMV-AS was stably integrated into the target cells and expressed by Western blot and drug-sensitivity tests , and inhibited the vascular endothelial cells proliferation in vitro .\n\nIn addition , the effects of the angiostatin vector on reducing the volume of tumors implanted in nude mouse models were also noted .\n\nCONCLUSION This study demonstrated that the recombinant pRC/CMV-AS mediated by liposome may play a potential role in the treatment of pancreatic cancer in the future .", "output": "Inducing angiogenesis" }, { "input": "Thirty-five cases of clear cell sarcoma of soft tissues were studied to determine the clinical or morphologic features that are important in predicting prognosis .\n\nTumors occurred most commonly in the extremities , and the majority of the patients were young women .\n\nSurgery was the elected treatment in every case .\n\nFive patients experienced local recurrences , and metastases developed in 22 .\n\nFifty-four percent of the patients died of tumor , 11% are alive with disease , and the remaining 34% are alive and well ; the average survival for each group was 67 months , 113 months , and 103.5 months , respectively .\n\nThis sarcoma is characterized by small clusters of polygonal to spindle cells featuring clear to slightly basophilic cytoplasm and vesicular nuclei with prominent nucleoli .\n\nThe clusters are separated by delicate fibrous septa .\n\nIn a deletion , clear cell sarcoma has low mitotic activity , little or no necrosis , and mild nuclear pleomorphism .\n\nTumor size and the presence of necrosis are statistically significant predictors of prognosis .\n\nAll 12 patients with tumors measuring > 5 cm died of disease or are alive with disease .\n\nEleven of the 20 patients with tumors measuring < 5 cm are alive with no evidence of disease .\n\nTumor necrosis was present in 10 cases ; eight of these patients died of disease and one is alive with disseminated metastases .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "Cytokines are known to play an important role in host defense by regulating the function , growth , and differentiation of the cells of the immune system .\n\nWe hypothesize that , in the tumor microenvironment , tumor cells and resident tissue cells ( e.g. , fibroblasts ) also produce cytokines that may regulate the local immune response to tumors .\n\nInitially , homogenates of eight head and neck squamous cell carcinomas ( HNSCC ) were assayed for the presence of interleukin-1 ( IL-1 ) , interleukin-4 ( IL-4 ) , interleukin-6 ( IL-6 ) , and granulocyte-macrophage colony-stimulating factor ( GM-CSF ) to establish the presence of these cytokines in the tumors in vivo .\n\nWe detected IL-1 in all tumor homogenates and IL-4 , IL-6 , and GM-CSF in some homogenates .\n\nTo assess the ability of HNSCC to produce these cytokines , supernatants of short-term primary cultures of HNSCC were assayed for the same cytokines .\n\nNo IL-1 was detected , although baseline levels of IL-4 , IL-6 , and GM-CSF were present .\n\nHowever , the stimulation of primary tumor cultures with exogenous IL-1 induced or significantly enhanced production of IL-4 ( p < 0.01 ) , IL-6 ( p < 0.001 ) , and GM-CSF ( p < 0.02 ) .\n\nThese results support our hypothesis that HNSCC secrete cytokines that may influence the response of local immune cells .\n\nOur data also suggest that IL-1 may have a central role in regulating the local immune response through the enhancement or induction of cytokine production by tumor and/or resident tissue cells .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "The high affinity fibronectin receptor ( FNR ) is expressed by hematopoietic cells , fibroblasts , and proliferating epidermal cells .\n\nExpression of this integrin is altered by chemical and viral transformation , suggesting that FNR dysfunction may play a role in growth control .\n\nThis study demonstrates that exposing FA-K562 cells to glycine-arginine-glycine-aspartate-serine ( GRGDS ) , a peptide ligand of the FNR , specifically stimulates p34/cdc2- and cyclin A-associated kinase activities .\n\nThis occurs within 2 h of peptide addition .\n\nThe 110-kDa form of the retinoblastoma protein appears within 3 h of GRGDS addition , consistent with activation of a G1/S kinase .\n\nDNA staining profiles demonstrate that GRGDS induces cell cycle progression within 24 h .\n\nIncreased anchorage-independent growth is subsequently observed in GRGDS-treated FA-K562 cells .\n\nThe control peptide , GRGES , which cannot bind the FNR , has none of these effects .\n\nThis demonstrates that an extracellular integrin ligand can regulate cell proliferation .\n\nFurthermore , these results suggest that integrins link the extracellular environment and intracellular growth regulators .", "output": "Sustaining proliferative signaling" }, { "input": "Capillary endothelial cells can be induced to form capillary-like structures in vitro by plating on fibronectin-coated dishes ( Ingber , D. E. , and Folkman , J .\n\n( 1989 ) J. Cell Biol. 109 , 317-330 ) , thereby mimicking angiogenesis .\n\nTo assess the role of glycoproteins bearing asparagine-linked oligosaccharides in this process , we tested the effect of oligosaccharide processing inhibitors on the formation of capillary tubes .\n\nDeoxymannojirimycin , a compound that prevents synthesis of hybrid and complex-type oligosaccharides , inhibited the formation of capillary tubes .\n\nIn contrast , swainsonine , an inhibitor that blocks synthesis of complex- but not hybrid-type oligosaccharides , did not inhibit tube formation .\n\nLectin affinity chromatography of 2-[3H] mannose-labeled glycopeptides from endothelial cells induced to form tubes did not reveal a striking difference in the spectrum of oligosaccharides compared to uninduced cells .\n\nSince endothelial cells formed tubes normally in the presence of swainsonine , we analyzed glycopeptides from swainsonine-treated induced and uninduced cells .\n\nCells induced to form tubes were enriched in monosialylated hybrid-type oligosaccharides sensitive to alpha-fucosidase , beta-galactosidase , and beta-N-acetylhexosaminidase , suggestive of sialyl Lewis-X determinants .\n\nWe used an enzyme-linked immunoassay to measure sialyl Lewis-X epitopes on capillary endothelial cells and found that both induced and uninduced cells expressed sialyl Lewis-X epitopes .\n\nDeoxymannojirimycin and , to a lesser extent , swainsonine reduced the level of sialyl Lewis-X epitopes in cells induced to form capillary tubes , but neither compound affected the level of epitopes in cell monolayers .\n\nWe conclude that synthesis of at least hybrid-type oligosaccharides is required for capillary tube formation in vitro and that an increase in monosialylated , fucosylated glycans on asparagine-linked oligosaccharides occurs during this process .", "output": "Inducing angiogenesis" }, { "input": "The UT-7 cell line was established from a patient with a megakaryoblastic leukemia ( Komatsu et al , Cancer Res 51 : 341 , 1991 ) .\n\nIts proliferation is strictly dependent on the presence of hematopoietic growth factors including erythropoietin ( Epo ) , granulocyte-macrophage colony-stimulating factor ( GM-CSF ) , and interleukin-3 ( IL-3 ) .\n\nWe investigated the differentiation capacities of this cell line under the action of several growth factors , using immunomarkers , flow cytometry , and ultrastructural techniques .\n\nIn the presence of GM-CSF and IL-3 , eosinophil and basophil promyelocytes were detected , as well as a few cells with erythroid and megakaryocytic ( MK ) differentiation features .\n\nIn contrast , Epo induced a marked erythroid differentiation with an increase of glycophorin A expression , accompanied by a few hemoglobinized cells .\n\nDifferentiation induced by the growth factors took 24 to 48 hours to begin , and increased with cell passages to a plateau at 2 weeks of culture .\n\nHowever , this was not only due to a cell selection because the differential effects of Epo and GM-CSF were observed from a single cell clone and the phenotype could be reversed by opposite growth factors , even after a long period of culture .\n\nWe subsequently investigated the phenotype of UT-7 in the presence of combinations of Epo , IL-3 , and GM-CSF , and showed that GM-CSF and IL-3 act predominantly over Epo .\n\nThis effect was mediated by a rapid downmodulation of Epo receptors by GM-CSF at messenger RNA and binding sites levels , without a change in receptor affinities .\n\nOn the other hand , Epo had no effect on number and affinity of GM-CSF receptors .\n\nThis study shows that UT-7 is a growth factor-dependent pluripotent cell line in which commitment may be directed by a hierarchical action of growth factors through an early and rapid transmodulation of growth factor receptors .", "output": "Sustaining proliferative signaling" }, { "input": "Transforming growth factor-Beta ( TGF-beta ) is a potent growth inhibitor for several cell types including epithelial cells and hematopoietic progenitor cells .\n\nUsing a human promonocytic leukemia cell line , THP-1 , we have shown that TGF-beta inhibits their proliferation and promotes differentiation into cells exhibiting macrophage-like properties .\n\nTherefore , a key question is whether TGF-beta influences the expression of genes associated with proliferation and/or growth inhibition .\n\nTGF-beta treatment of THP-1 cells results in downregulation of expression of c-myc .\n\nWe also observe that TGF-beta 1-treated cells express reduced levels of the cell cycle regulated histone , H2B , but express elevated levels of an RNA splicing variant of this histone that has been observed to be upregulated in growth inhibited and terminally differentiated cells .\n\nIn addition , a nuclear protein associated with senescence and withdrawal of cells from the cell cycle , statin , is also expressed by THP-1 cells in response to TGF-beta 1 treatment .\n\nThese results suggest that TGF-beta 1 is capable of inducing expression of specific nuclear proteins associated with differentiation and/or cessation of proliferation that may result in changes in nuclear organization and altered gene expression .\n\nSuch changes in nuclear organization may be incompatible with continued proliferation of the cells .", "output": "Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "OBJECTIVE To explore the adduct characteristics of styrene and DNA .\n\nMETHODS The adduct reactions between styrene , urinary mandalic acid(MA) , phenylglyoxalic acid(PGA) , mercapturic acid of styrene ( UMA ) and DNA were studied by ultraviolet spectral analysis .\n\nThe SO-DNA adducts by 32P-post labeled method , the chemical structures of SO-DNA adducts by GC-MS and NMR were also studied .\n\nRESULTS SO combined with DNA at O6 , N2 positions of dGMP to form six adducts , but styrene , urinary mandalic acid , phenylglyoxalic acid and mercapturic acid of styrene did not react with DNA to form adduct .\n\nCONCLUSIONS Styrene formed adduct with DNA through its active center metabolite--SO after entering the body .\n\nSO combined with DNA at O6 , N2 positions of dGMP to form adducts .\n\nIf these DNA adducts are not repaired or are mis-repaired before cell duplication , the gene mutation and chemical damage would happen .\n\nNo adduct reactions are seen among other metabolites of styrene .", "output": "Genomic instability and mutation" }, { "input": "This is a case report of a 69-year-old woman with sarcomatoid hepatocellular carcinoma ( HCC ) , which was diagnosed clinically as hemangioma .\n\nShe was first admitted to our university hospital , complaining of general fatigue in December , 1988 , and cholelithiasis and liver cirrhosis with hepatic tumor in Segment 8 were diagnosed .\n\nThe serum AFP level was within normal range , and the tumor was diagnosed as hemangioma radiologically .\n\nShe underwent only cholecystectomy and was well without any therapy for the liver tumor up until March in 1991 when she was readmitted to our university hospital due to rapidly progressive liver dysfunction .\n\nThe size of the liver tumor was unchanged .\n\nDespite intensive care , she died of hepatic failure due to cirrhosis in a decompensation state .\n\nAt autopsy , a well defined yellowish white tumor of 3 cm in maximum diameter was seen in the cirrhotic liver .\n\nAlthough the largest part of the tumor revealed necrosis and hyalinization , a sarcomatoid part composed of spindle-shaped cells was noted in the peripheral portion .\n\nIn addition , some necrotic ghost cells , probably hepatocellular carcinoma , were also noted .\n\nLow molecular cytokeratin , which is always found in HCCs , was seen in spindle-shaped sarcomatoid cells .\n\nThe liver tumor was diagnosed as sarcomatoid HCC from these pathological findings .\n\nWe report this histologically unusual HCC with an immunohistochemical study .", "output": "Resisting cell death" }, { "input": "Chromatographic , peptide mapping and mass spectrometric analysis were used to examine hemoglobin ( Hb ) from heavy drinkers and abstainers for alcohol consumption-related modifications .\n\nHeavy drinker and abstainer hemoglobin samples contained similar amounts of glycosylated Hb and significantly different ( p < 0.05 ) amounts of \" fast \" hemoglobin .\n\nThe presence of higher amounts of \" fast \" Hb in heavy drinker relative to abstainer samples suggested the presence of alcohol-consumption related modifications .\n\nTo further examine Hb for modifications , tryptic peptides of the \" fast \" hemoglobin HbA1c were isolated and analyzed by plasma desorption mass spectrometry ( PDMS). [ 14C]acetaldehyde ( AcH)-Hb was synthesized in vivo for use as a standard .\n\nSpecific peptides were chosen based on co-migration with radiolabeled peptides from a tryptic digest of the [ 14C]acetaldehyde-Hb .\n\nThe masses obtained by PDMS for two heavy drinker peptides were identical to two radiolabeled peptides ; the two pairs of peptides co-migrated on HPLC .\n\nA comparison of the observed mass for the peptides with the theoretical masses for acetaldehyde-modified Hb peptides suggested that the peptides were AcH-modified alpha and beta chain N-termini of Hb .\n\nThe modified peptides were found in five of six heavy drinker samples .\n\nThis is the first description of site-specific AcH-Hb adducts occurring in vivo .\n\nThe routine detection of such adducts has potential for characterizing usual alcohol intake .", "output": "Genomic instability and mutation" }, { "input": "A tumor growth-dependent elevation in the hepatic levels of Zn and metallothionein ( MT ) , without a change in the level of Cu , was found in mice and rats bearing solid tumors in the inguinal region .\n\nThe levels of Zn and MT thus elevated gave a significant correlation ( r = 0.95 ) between them .\n\nNevertheless , when tumor-bearing mice and rats were fed a Zn-deficient diet , the hepatic levels of Zn and MT did not increase .\n\nIn mice in which inflammation was induced at the same region , on the other hand , hepatic levels of Zn and MT increased transiently after the injection of turpentine or carrageenan even when they were fed the Zn-deficient diet .\n\nThese results suggest that the elevation of MT and Zn levels can be a helpful marker for detecting malignancy .", "output": "Tumor promoting inflammation" }, { "input": "The induction of apoptosis by glucocorticoids in isolated thymocytes has been studied extensively .\n\nHowever , it is not known whether or not the same changes occur after in vivo glucocorticoid treatment .\n\nIn order to investigate this , we have studied the changes occurring in thymocytes isolated from rats , from 2-24 hr after a dose of dexamethasone ( 1 mg/kg ) , which caused 50% thymic atrophy .\n\nThymocytes were separated into four fractions by isopycnic Percoll gradients .\n\nA loss of cells occurred within 2-8 hr , primarily in only one of the two major fractions of normal thymocytes .\n\nThis loss of normal thymocytes coincided with the appearance of small dense cells with characteristic features of apoptosis including condensed chromatin , increased DNA fragmentation , internucleosomal DNA cleavage and a \" hypodiploid \" peak on flow cytometric analysis .\n\nStriking differences occurred in the cellular composition of the different Percoll fractions with time .\n\nInitially ( up to 4 hr ) , the pattern of changes occurring in vivo resembled those found in vitro .\n\nHowever , at later times , the complex fate of apoptotic cells in vivo , such as phagocytosis , are not observed in the in vitro studies .", "output": "Resisting cell death" }, { "input": "Cell kinetics and activity of ornithine decarboxylase ( ODC ) were studied during the process of 1-hydroxyanthraquinone ( 1-HA)-induced intestinal carcinogenesis in rats .\n\nStarting at 6 weeks of age , a total of 37 male ACI/N rats were divided into two groups and treated as follows : group I ( 18 rats ) received diet containing 1% 1-HA for 12 months ; group II ( 19 rats ) was given the basal diet alone .\n\nSub-groups of 5-7 rats were sequentially killed at 4 , 8 and 12 months for evaluation of the length , cell numbers and 5-bromo-2'-deoxyuridine ( BrDU ) labeling indices of large bowel crypts together with ODC activity .\n\nAll kinetic and ODC data indicated increased DNA synthesis and proliferation at all time points .\n\nMorphological observation of the intestines also revealed melanosis , crypt abscesses and erosion , becoming more pronounced with length of exposure to the anthraquinone .\n\nThe data thus suggest that cell proliferation in the crypts of the cecum or colon is important for 1-HA-induced intestinal carcinogenesis .", "output": "Sustaining proliferative signaling" }, { "input": "Cells dissociated from spontaneous and transplanted tumours of C3HJax mammary gland have been cultured on polylysine and gelatin substrates .\n\nThe isolated cells proliferated to form monolayers with high degree of organoid structure as indicated by formation of alveolar cavities .\n\nDifferences were observed in the cell attachment , growth pattern , number and size of alveolar cavities , cells which lined the cavity and cell morphology on polylysine and gelatin substrates as compared to conventional cell culture plastic surface .\n\nOn polylysine more than 90% cells attached rapidly , within 15-45 min after plating , with or without serum and formed confluent monolayers marked by presence of large and small alveolar cavities .\n\nMultiple interacting cell types took part in organization of the cavity .\n\nCells lining the cavity constantly proliferated and rearranged to expand it .\n\nOn gelatin , 60-70% cells attached over a period of 6-24 hr in presence of serum and formed confluent monolayers dominated by small alveolar cavities .\n\nCells forming the cavities were epithelial in nature and cavities once formed did not increase in size .\n\nUpon subculture , the cell morphology on these substrates was strikingly different .\n\nOn polylysine , the predominant cell type had numerous irregular microvilli whereas on gelatin , cells had smoother boundaries with a few stunted cytoplasmic extensions .\n\nThe cell attachment on conventional surface was low , 40-50% .\n\nWhen seeded at high cell density , formation of alveolar cavities was suppressed and at low cell density , cultures were marked by contact inhibition of cells and failure to attain confluence .\n\nThese results suggest differential behaviour and interaction of mammary tumour epithelium with the substrates used .", "output": "Evading growth suppressors" }, { "input": "The use of substrates containing well defined adducts at precise sites , is required to perform a careful analysis of the toxic and mutagenic potential of a lesion .\n\nAs a first step in this direction the octamer 5'-d(CCGGCGGT) , containing the sequence of the codons 12 d(GGC) and 13 d(GGT) of the human H-ras gene , was reacted with the antitumoral drug cis-diamminedichloroplatinum(II) .\n\nThe platinated products have been purified by HPLC .\n\nA first set of experiments , including enzymatic digestions with nuclease P1 followed by alkaline phosphatase and acid-catalysed hydrolysis , allowed us to determine which bases were engaged in the cis-DDP lesions .\n\nOur results indicate that only guanine residues were chelated with cisplatin to yield bifunctional adducts .\n\nFurthermore , by performing enzymatic digestions with phosphodiesterases , we have located the adducts with respect to the 5 ' end of the octamer .\n\nAmong the purified and characterized platinated oligonucleotides , three present a particular interest , since we have shown here that the cis-d(GpG) adduct is precisely situated either at the d(GGC) or at the d(GGT) or at both sites of their sequence .", "output": "Genomic instability and mutation" }, { "input": "We have exploited properties of photosensitizers to study an aspect of the packing of chromatin in the cell nucleus .\n\nThe fluorescent photosensitizers mesotetra(3-hydroxyphenyl) porphyrin and Photofrin II were both localized in the nuclear membrane and other membrane structures , but could not be found inside the nuclei .\n\nLight exposure of cells at 1 degrees C in the presence of the sensitizers induced DNA double-strand breaks .\n\nThe length distributions of DNA fragments were determined by pulsed field gel electrophoresis .\n\nBecause DNA damage is produced mainly via singlet oxygen diffusing less than 0.1 microns from the sensitizer , DNA double-strand breaks were supposedly produced within this distance of the nuclear membrane .\n\nConsistent with this , with prolonged illumination and with increasing concentrations of sensitizer the distribution of DNA fragment lengths reached a plateau level .\n\nIn contrast , with the hydrophilic , intranuclear sensitizer meso-tetra(4-sulphonatophenyl)porphyrin , no such plateau level was found .\n\nThe plateau distributions of DNA fragment lengths of different cell types had the same general shape with average fragment lengths ranging from 174 to 194 kilobasepairs .\n\nParticular genes , c-myc , fos and p53 , were found on broad distributions of photocleaved fragment lengths .\n\nThe results indicate that on each side of the genes the locus of the chromatin fibre situated close to the nuclear membrane , varied randomly .", "output": "Genomic instability and mutation" }, { "input": "Using an immunoradiometric assay , Cathepsin D ( Cath D ) levels were measured in the cytosol of 23 normal and 39 neoplastic human laryngeal tissues .\n\nScattered Cath D levels ( from 2.2 to 17.8 pM/mg protein ; median = 7.6 ) were found in normal mucosa specimens .\n\nCath D concentrations range from 2.0 to 29.3 pM/mg protein ( median = 8.5 ) in laryngeal tumors .\n\nWhen a comparison between Cath D levels in normal and neoplastic tissue specimens from the same patient was done , Cath D levels were significantly higher in laryngeal cancers than in their normal counterparts ( P = 0.03 ) .\n\nNo correlation with clinico-pathological parameters and steroid hormone and epidermal growth factor receptor status was found .\n\nFurther studies should investigate whether the production of Cath D by laryngeal tumors could have a clinical relevance for this neoplasia .", "output": "Sustaining proliferative signaling" }, { "input": "To assess the importance of changes in DNA methylation in an X-ray-induced cellular transformation process , methylation patterns of five nuclear protooncogenes in fifteen transformant clones were studied and compared to that of the parental non-transformed cell line m5S/1M .\n\nAll transformants examined revealed an alteration in DNA methylation in some of the genes , although these changes were variable among them .\n\nA comparison of cellular characteristics with corresponding DNA methylation changes in different clones suggested that the loss of contact inhibition and the gain of anchorage independency were associated with increases of methylation in many genes , whereas the acquisition of tumorigenicity was often accompanied by a decrease of methylation in the N-myc and c-myc genes .\n\nResultant data indicate that the alteration of DNA methylation is closely related to transformation process , yet how this involvement occurs is complex and remains unclear .", "output": "Evading growth suppressors" }, { "input": "We compared gadolinium diethylenetriaminepentaacetic acid ( Gd-DTPA ) enhanced T1-weighted images ( T1-Gd ) with the histopathological findings in 13 patients with bone or soft tissue sarcomas .\n\nSignal intensity of the viable tumor tissue was increased in T1-Gd in 92% of the patients .\n\nThe necrotic or cystic areas in the tumor were not enhanced , rendering them distinctly .\n\nThe degree of enhancement of the edematous area around the tumor was similar to or more marked than that of the tumor in 54% of the patients .\n\nArea showing inflammatory cells infiltration and edematous areas in the tumor tissue were also enhanced .\n\nThus , the effect of preoperative chemotherapy in tumor tissues other than necrotic and cystic areas tended to be underestimated in T1-Gd .\n\nIts effect should be comprehensively evaluated based on not only T1-Gd but also T2-weighted images and findings of other imaging techniques .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Prevention of environmentally related cancer will be enhanced by the availability of sensitive early warning systems and by improvements in quantitative assessment of human risks .\n\nAccordingly , we have carried out a series of molecular epidemiologic studies aimed at validating a panel of biologic markers , including carcinogen-DNA and -protein adducts , sister chromatid exchange , micronucleus formation , DNA strand breaks , and DNA repair capacity .\n\nResults from three such studies illustrate the usefulness of these biomarkers in elucidating low-dose-response relationships , correlations between biomarkers , and the range of variation in biomarkers between individuals exposed to similar concentrations of carcinogens .\n\nLow-level workplace or ambient exposures to styrene , ethylene oxide , and polycyclic aromatic hydrocarbons ( PAH ) were associated with significant increases in both molecular dose of carcinogens ( adducts ) and various markers of preclinical effects .\n\nCorrelations between biomarkers varied by exposure .\n\nFor example , in the styrene study , sister chromatid exchange frequency was not correlated with any of the markers , in contrast to the studies of ethylene oxide and PAH .\n\nSignificant molecular effects were observed not only in occupationally exposed people but also in residents of an area in Poland characterized by high levels of air pollution .\n\nFor example , the mean PAH-DNA level in exposed residents ( winter sample ) was 30.4 adducts per 10(8) nucleotides .\n\nThis level was significantly higher than that of adducts seen in summer samples from the same area ( 4.2/10(8) , or in winter samples from residents of a rural area ( 11.01/10(8) .\n\nSignificant seasonal variation in PAH-DNA adduct formation in this group was consistent with recorded fluctuations in air pollution levels .\n\nStriking interindividual variation was observed in all three exposed populations .", "output": "Genomic instability and mutation" }, { "input": "We have extended our studies on the relationship between cisplatin/carboplatin-induced DNA damage in readily accessible tissue(s) and clinical response to therapy .\n\nSuch an approach may assist in the study of cancer drug resistance and in establishing parameters for assessing human populations for sensitivity to DNA damaging agents in the environment .\n\nPlatinum-DNA adduct levels were measured by atomic absorbance spectrometry .\n\nDNA repair capacity was assessed in human T-lymphocytes by the ability to repair cisplatin lesions in cellular DNA or in transfected plasmid DNA .\n\nIn a \" blinded \" study of 21 patients receiving combination cisplatin/carboplatin drug therapy , there was a direct relationship between DNA damage in leukocytes and disease response ( summary two-sided p = 0.00011 ) .\n\nThe cohort of patients had 15 different tumor types , suggesting that blood tissue and tumor tissue of an individual may process platinum-DNA damage similarly regardless of the tissue of origin of the tumor .\n\nIn leukocytes in vivo , persistence and accumulation were prominent features of the cisplatin-DNA adduct profile .\n\nFunctional DNA repair capacity has been studied in eight human leukocyte cell lines in vitro ( three , T-cells ; three , B-cells ; one , monocytic ; one , promyelocytic ) , using a host cell reactivation assay with cisplatin-damaged pRSVcat .\n\nIn the three T cell lines studied , host cell reactivation efficiency was directly related to the cells ' abilities to repair cisplatin-damaged cellular DNA ( correlation coefficient = 0.993).(ABSTRACT TRUNCATED AT 250 WORDS )", "output": "Genomic instability and mutation" }, { "input": "PRL induces quiescent Nb2 rat T-lymphoma cells to undergo mitogenesis .\n\nUpon PRL stimulation , the transcription factor interferon regulatory factor-1 ( IRF-1 ) is induced as a novel T-cell activation gene in Nb2 cells .\n\nSurprisingly , IRF-1 is expressed twice during a single PRL-induced growth cycle : first during the early G1 phase , in an immediate transient peak from 15 min to 2 h , and second during the G1/S phase transition , in a broader peak beginning at 8 h .\n\nThe unusual biphasic expression of IRF-1 mRNA is accompanied both times by de novo IRF-1 protein synthesis .\n\nHowever , the rate of IRF-1 protein turnover appears to be different in G1 and S phases .\n\nIRF-1 protein expressed in G1 exhibits a half-life of about 25 min , whereas in the S phase , the half-life is about 60 min .\n\nBy washing out PRL at various times during G1 , we found a direct correlation among the length of PRL exposure , the second peak of IRF-1 mRNA expression , and DNA synthesis .\n\nOur data suggest that PRL and one putative nuclear mediator , IRF-1 , may be important in two distinct phases of the cell cycle : first in cell cycle activation , and then in S phase progression .", "output": "Sustaining proliferative signaling" }, { "input": "Psoralen photoreaction with DNA produces interstrand crosslinks , which require the activity of excision and recombinational pathways for repair .\n\nYeast replicating plasmids , carrying the HIS3 , TRP1 , and URA3 genes , were photoreacted with psoralen in vitro and transfected into Saccharomyces cerevisiae cells .\n\nRepair was assayed as the relative transformation efficiency .\n\nA recombination-deficient rad52 strain was the least efficient in the repair of psoralen-damaged plasmids ; excision repair-deficient rad1 and rad3 strains had repair efficiencies intermediate between those of rad52 and RAD cells .\n\nThe level of repair also depended on the conditions of transformant selection ; repair was more efficient in medium lacking tryptophan than in medium from which either histidine or uracil was omitted .\n\nThe plasmid repair differential between these selective media was greatest in rad1 cells , and depended on RAD52 .\n\nPlasmid-chromosome recombination was stimulated by psoralen damage , and required RAD52 function .\n\nChromosome to plasmid gene conversion was seen most frequently at the HIS3 locus .\n\nIn RAD and rad3 cells , the majority of the conversions were associated with plasmid integration , while in rad1 cells most were non-crossover events .\n\nPlasmid to chromosome gene conversion was observed most frequently at the TRP1 locus , and was accompanied by plasmid loss .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE To investigate a potential mechanism of invasion and metastasis of oral cancers , and the possibility of the adhesion molecules being markers for evaluating the prognosis of oral cancers .\n\nMETHODS Randomly selecting achieved paraffin blocks , including 60 cases of squamous cell carcinoma(SCC) , 30 cases of verrucous carcinoma(VC) of oral mucosa and 30 cases of basal cell carcinoma ( BCC ) of epidermis .\n\nAll cases were followed up .\n\nUsing immunohistochemistry method ( Envision ) to detect the expression of E-cadherin antibody and observe under light microscope .\n\nRESULTS The studies demonstrated that 38.2% of SCC had recurrence and/or metastases after operation .\n\nHowever , only 0.0% and 0.04% , in VC and BCC , respectively , had recurrence and/or metastases .\n\nThere was significant difference on prognoses between SCC and VC or BCC .\n\nThe immunostaining showed significant difference of E-cad expression between SCC and VC ( P<0.01 ) , SCC and BCC ( P<0.05 ) .\n\nThe former was weaker than VC and BCC .\n\nHowever , no difference was showed in E-cad expression between VC and BCC ( P>0.05 ) .\n\nCONCLUSION E-cad plays an important role in maintaining the phenotype of epithelial cell of SCC and normal oral mucosa .\n\nThe degree of expression of E-cad was correlated with the ability of invasion and/or metastasis of SCC , VC and BCC .\n\nIt indicates that E-cad may be as a helpful marker to evaluate the prognosis of oral cancers .", "output": "Activating invasion and metastasis" }, { "input": "In our study we investigated the level of apoptosis in PBMCs and the serological level of sFas ( CD95/APO-1 ) in 22 patients with malignant melanoma ( 12 patients with unique cutaneous primary tumour and 10 patients with unique brain metastasis ) .\n\nThe first determination was performed before tumour excision and the second at 6-7 months after excision .\n\nResults in patients with primary tumour in the first determination : 6 patients with over normal values in PBMCs apoptosis and 5 patients with increased values of sFas .\n\nIn the second determination : apoptosis was increased in 5 patients and sFas level was increased in 4 cases .\n\nIn patients with metastases in the first determination apoptosis of PBMC was increased in 7 cases and sFas in 5 cases .\n\nIn the second determination apoptosis was increased in 4 cases and sFas was increased in 4 cases .\n\nOur results show that half of the investigated patients presented elevated values of PBMCs apoptosis and Fas receptor both before and 6-7 months after tumour excision .\n\nApoptosis values for PBMCs and sFas values were with 1/4 higher than normals .\n\nThere was no difference in clinical evolution of the patients with normal or increased values for studied parameters .\n\nClinical evolution was performed for 1 year .\n\nThe presence of increased values for PBMCs and sFas after tumour excision , primary or metastasis is surprising and hard to explain .\n\nIt is possible that tumoral evolution induces a disregulation at PBMCs level or other cells level that persists unexpectedly , after tumour excision or apoptotic processes , in a certain level to be independent and anterior to tumour development .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "Contrary to the paradigm that cancer incidence increases indefinitely with age , significant data now suggest cancer incidence may markedly reduce beyond age 80 years for humans and beyond 800 days for mice , and is not inevitable .\n\nWe show that increasing cellular senescence with age is a possible cause of this reduction , since senescent cells are removed from the pool of cells that retain proliferative ability necessary for cancer .\n\nWe further show that animal interventions appearing to alter senescence , p53 mutation and melatonin dosing , support the prediction that increasing senescence rate reduces cancer while reducing lifespan , and vice versa .\n\nStudies of environmental agents associated with increased cancer might be re-examined to find if there is an association with longevity increases , which may markedly alter our view of such agents .\n\nWe also show that if an agent functions by slowing both senescence and carcinogenesis , longevity is increased while reducing cancer .\n\nDietary restriction is the only known intervention that accomplishes this , but there may be others .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Epidermal growth factor ( EGF ) receptor is inversely related to expression of estrogen receptor ( ER ) and progesterone receptor in primary breast tumors and is a negative predictor for response to endocrine therapy .\n\nTo investigate a possible causal role of EGF receptor expression in breast cancer progression to hormone independence , we have created an experimental cell system .\n\nEpidermal growth factor receptor complementary DNA was introduced in estrogen-dependent ZR-75-1 breast cancer cells , and the resulting ZR/HERc cells exhibited a mitogenic response to epidermal growth factor , thus bypassing estrogen dependence .\n\nThis EGF-induced proliferation could not be inhibited by antiestrogens .\n\nIn addition , we noted changes in cell morphology and keratin expression of EGF-stimulated ZR/HERc cells , suggestive of an altered differentiation state .\n\nFurthermore , intolerance of functional ER and EGF receptor signal transduction pathways in ZR/HERc cells was observed during simultaneous activation , which possibly explains the inverse relationship of ER and EGF receptor expression in primary tumors .\n\nIn contrast to the parental cells , ZR/HERc cells rapidly progressed to a stable ER-negative phenotype when cultured in the presence of the antiestrogen hydroxy-tamoxifen .\n\nThese results suggest a possible role for EGF receptor in progression of breast cancer to hormone independence .", "output": "Sustaining proliferative signaling" }, { "input": "In mice fed a diet supplemented with red clover isoflavones the prostatic epithelium displays a significant increase in the production of estrogen receptor beta and the adhesion protein E-cadherin but a decrease in transforming growth factor beta1 .\n\nThese proteins are estrogenically-induced markers of proliferation , maintenance of histological architecture , preservation of cell phenotype and reduction of the potential for neoplastic and metastatic transformation .\n\nThis study suggests that red clover isoflavones represent a non-toxic dietary treatment for prostatic hyperplasia and a reduction in the potential for neoplastic transformation .", "output": "Sustaining proliferative signaling" }, { "input": "Limited options for the treatment of prostate cancer have spurred the search for new therapies .\n\nOne innovative approach is the use of targeted alpha therapy ( TAT ) to inhibit cancer growth , using an alpha particle emitting radioisotope such as ( 213)Bi .\n\nBecause of its short range and high linear energy transfer ( LET ) , alpha-particles may be particularly effective in the treatment of cancer , especially in inhibiting the development of metastatic tumors from micro-metastases .\n\nProstate-specific membrane antigen ( PSMA ) is expressed in prostate cancer cells and the neovasculature of a wide variety of malignant neoplasms including lung , colon , breast and others , but not in normal vascular endothelium .\n\nThe expression is further increased in higher-grade cancers , metastatic disease and hormone-refractory prostate cancer ( PCA ) .\n\nJ591 is one of several monoclonal antibodies ( mabs ) to the extracellular domain of PSMA .\n\nChelation of J591 mab with ( 213)Bi forms the alpha-radioimmunoconjugate ( AIC ) .\n\nThe objective of this preclinical study was to design an injectable AIC to treat human prostate tumors growing subcutaneously in mice .\n\nThe anti-proliferative effects of AIC against prostate cancer were tested in vitro using the MTS assay and in vivo with the nude mice model .\n\nApoptosis was documented using terminal deoxynucleotidyl transferase [ TdT]-mediated deoxyuridinetriphosphate [ dUTP ] nick end-labeling ( TUNEL ) assay , while proliferative index was assessed using the Ki-67 marker .\n\nWe show that a very high density of PSMA is expressed in an androgen-dependent human PCA cell line ( LNCaP-LN3 ) and in tumor xenografts from nude mice .\n\nWe also demonstrate that the AIC extensively inhibits the growth of LN3 cells in vitro in a concentration-dependent fashion , causing the cells to undergo apoptosis .\n\nOur in vivo studies showed that a local AIC injection of 50 microCi at 2 days post-cell inoculation gave complete inhibition of tumor growth , whereas results for a non-specific AIC were similar to those for untreated mice .\n\nFurther , after 1 and 3 weeks post-tumor appearance , a single ( 100 microCi/100 microl ) intra-lesional injection of AIC can inhibit the growth of LN3 tumor xenografts ( volume<100 mm(3) ) in nude mice .\n\nTumors treated with AIC decreased in volume from a mean 46+/-14 mm(3) in the first week or 71+/-15 mm(3) in the third week to non-palpable , while in control mice treated with a non-specific AIC using the same dose , tumor volume increased from 42 to 590 mm(3) .\n\nThere were no observed side effects of the treatment .\n\nBecause of its in vitro cytotoxicity and these anti-proliferative properties in vivo , the ( 213)Bi-J591 conjugate has considerable potential as a new therapeutic agent for the treatment of prostate cancer .", "output": "Resisting cell death" }, { "input": "We analyzed the clinico-pathological features of the initial tumors in 205 patients with superficial bladder cancer , admitted to Kyoto University Hospital between 1974 and 1988 , to investigate the prognostic factors for progression to the muscle invasive disease or metastasis .\n\nOf 205 patients , 35 ( 17% ) exhibited muscular invasion alone ( 12 patients ) and/or metastasis ( 23 patients ) .\n\nTumor multiplicity , higher grade and positive urinary cytology were the significant risk factors for later malignant progression .\n\nExpression of A , B , H-blood group isoantigens in the bladder tumor were significantly decreased from the onset in the patients with initially T1 tumor but not in those with Ta tumor .\n\nSignificant loss of expression was also found at the time of progression in the initially Ta cases .\n\nThus , loss of A , B , H-blood group antigen expression seems to be correlated with the malignant potential of superficial bladder cancer .\n\nHowever , more feasible and reliable diagnostic markers such as molecular genetical and biochemical markers remain to be developed to predict the malignant potential of the superficial bladder cancer .", "output": "Activating invasion and metastasis" }, { "input": "Although it has been demonstrated that discrete origins of DNA replication exist in eukaryotic cellular chromosomes , the detailed organization of a eukaryotic cellular origin remains to be determined .\n\nLinker substitution mutations were constructed across the entire Saccharomyces cerevisiae chromosomal origin , ARS1 .\n\nFunctional studies of these mutants revealed one essential element ( A ) , which includes a match to the ARS consensus sequence , and three additional elements ( B1 , B2 , and B3 ) , which collectively are also essential for origin function .\n\nThese four elements arranged exactly as in ARS1 , but surrounded by completely unrelated sequence , functioned as an efficient origin .\n\nElement B3 is the binding site for the transcription factor-origin binding protein ABF1 .\n\nOther transcription factor binding sites substitute for the B3 element and a trans-acting transcriptional activation domain is required .\n\nThe multipartite nature of a chromosomal replication origin and the role of transcriptional activators in its function present a striking similarity to the organization of eukaryotic promoters .", "output": "Genomic instability and mutation" }, { "input": "Glutathione S-transferase ( GST ) class Mu activity was determined in 145 unrelated hospital patients in Berlin by measuring their conjugation activity towards the specific substrate trans-stilbene oxide ( TSO ) with two substrate concentrations ( 50 and 250 microM ) in homogenates prepared from lymphocytes .\n\nEighty individuals ( 55.2% ) had an activity lower than 10 pmol/min/10(6) lymphocytes and were classified as GST class Mu deficient .\n\nIn 142 of 145 cases , phenotype was confirmed by the results of a genotyping procedure using the polymerase chain reaction technique .\n\nTwo fragments of 273 and about 650 bp including one and two introns , respectively , could always be amplified from genomic DNA in individuals with high GST class Mu activity and could not be amplified in persons with impaired glutathione-TSO conjugation activity .\n\nThis indicates that persons with low activity carry a large deletion mutation within the GST class Mu gene .\n\nThe enzymatically determined antimode between low and high activity determined as 10 pmol/min/1 million lymphocytes in the assay with 50 microM TSO could be clearly confirmed by genotyping .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE To evaluate the difference of angiogenic factors PDGF/dThdPase,VEGF expression and microvessel density ( MVD ) in primary hypopharyngeal tumor and metastasis lymph nodes .\n\nMETHOD The author studied immunohistochemically a series of 48 primary hypopharyngeal carcinoma patients and metastasis lymph nodes were calculated .\n\nRESULT The percentage of VEGF was 25.38% in primary tumor and 21.52% in lymph nodes .\n\nNo significant difference was found .\n\nThe percentage of PDGF/dThdPase was 29.59% in primary tumor and and 21.2% in lymph nodes .\n\nThis showed significent difference .\n\nVEGF showed significent difference between live and death group(P < 0.05 ) and among differentiation group ( P < 0.05 ) .\n\nMVD showed significant difference between live and death group , early and late stage group , and T1-2 and T3-4 group ( P < 0.05 ) .\n\nThere were statistically significant correlations between the score of PDGF/ dThdPase , or VEGF and the score of MVD respectively .\n\nCONCLUSION The present study suggests that there was a correlation between VEGF or PDGF and MVD .\n\nVEGF and MVD were possible to be prognostic discriminators in hypopharyngeal carcinoma .\n\nPDGF expression in lymph nodes was significant higher than in primary tumors , and MVD expression in primary tumor was significant higher than in lymph nodes .", "output": "Inducing angiogenesis" }, { "input": "BACKGROUND Ovarian surface epithelial cells undergo several rounds of division to repair the wound created by follicular rupture at the time of ovulation .\n\nThis cyclical requirement for cell division , when not interrupted by the long anovulatory rest periods that occur during pregnancy and lactation , may contribute to the development of ovarian cancer .\n\nPURPOSE AND METHODS To test this hypothesis , we isolated rat ovarian surface epithelial cells from 10 adult female Fisher rats , initiated two mixed-population and seven clonal cell lines , and repeatedly subcultured these cells in vitro for more than 20 passages .\n\nWe then tested them for the acquisition of the following four features associated with transformation : 1 ) the loss of contact inhibition , 2 ) the capacity for substrate-independent growth , 3 ) the ability to form tumors when injected subcutaneously and/or intraperitoneally into athymic mice , and 4 ) cytogenetic abnormalities .\n\nRESULTS Loss of contact inhibition was observed in all nine late-passage cell lines .\n\nSix of the nine late-passage , but none of the early-passage , cell lines tested exhibited a capacity for substrate-independent growth that was augmented in a dose-dependent manner by epidermal growth factor .\n\nTwo late-passage cell lines ( clone 2 and mixed-population 2 ) generated tumors in athymic BALB/c mice within 3 weeks following subcutaneous injection of 5 x 10(6) cells , whereas similar numbers of early-passage cells from the same cell lines failed to generate palpable tumors .\n\nLate-passage clone 7 cells were tumorigenic when 5 x 10(7) cells were injected intraperitoneally .\n\nTwo of the cell lines analyzed exhibited alterations involving losses of part or all of one member of the chromosome 5 pair .\n\nClone 2 possessed an interstitial deletion , del(5)(q21.3q24) , consistent with the loss of an uncloned putative tumor suppressor gene at 5q22q23 previously reported to reside near the loci for the interferon alpha , interferon beta , and c-jun genes .\n\nEarly-passage clone 7 cells exhibited chromosome 5 monosomy , while late-passage cells contained one normal chromosome 5 and a derivative ( 5q12q ) .\n\nSouthern analysis of the three cell lines revealed no consistent loss of loci for the interferon and c-jun genes , although early-passage clone 7 cells had one half the gene copy number for the interferon beta and c-jun genes and both early- and late-passage clone 7 cells lacked DNA sequences hybridizing with the probe for interferon alpha .\n\nCONCLUSION This pattern of passage-dependent spontaneous transformation of rat ovarian surface epithelial cells in vitro supports the hypothesis that repetitious ovulation contributes to the etiology of human ovarian cancer .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The hypothesis that rodent cells can be immortalized by the direct induction of a single mutation-like event was tested by initiating cultures of benzo(a)pyrene treated Syrian hamster embryo cells with low inocula and expanding these few cells maximally until senescence prevented further culturing or immortalization took place .\n\nAccording to the mutation hypothesis immortalization is hardly to be expected under these conditions .\n\nHowever , immortalization was frequently observed .\n\nTherefore the induction of immortalization appears indirect .\n\nThe progeny of benzo(a)pyrene treated cells immortalized with a rate of 3.9 x 10(-8)/cell/generation , which is 64 times higher than the spontaneous rate .\n\nThe results are in line with the probabilistic theory developed in 1980 by both Fernandez et al .\n\n( Proc .\n\nNatl .\n\nAcad .\n\nSci .\n\nUSA , 77 : 7272-7276 , 1980 ) and Kennedy et al .\n\n( Proc .\n\nNatl .\n\nAcad .\n\nSci .\n\nUSA , 77 : 7262-7266 , 1980 ) , which states that treatment of cells with a carcinogen can result in a so-called activated state of the treated cells which is transmitted to the progeny and which results in an enhanced rate of transforming events .", "output": "Enabling replicative immortality" }, { "input": "The protein toxins ricin , abrin , Shiga toxin , and diphtheria toxin were found to induce lysis of several cell lines in a manner characteristic for programmed cell death or apoptosis .\n\nThe toxins induced DNA degradation , and light and electron microscopical studies revealed that lysis was preceded by reorganization of intracellular vacuoles , cell blebbing , and chromatin condensation both in Vero and in MDCK cells .\n\nCell lysis was efficiently inhibited by cycloheximide and 3-methyladenine ( 3MA ) , a specific inhibitor of autophagy .\n\nCycloheximide , which like 3MA inhibits autophagy , protected even when added at a time when the protein synthesis had been blocked by ricin , suggesting that the effect of cycloheximide on cell lysis is independent of its ability to inhibit protein synthesis .\n\nAlso theophylline and dibutyryl-cGMP had some protective effect , whereas a number of compounds reported to protect against apoptosis in other systems were without protective effects .\n\nThe data suggest that autophagy is important for the toxin-induced cell lysis .", "output": "Resisting cell death" }, { "input": "Cellular senescence is the genetically programmed cessation of cellular proliferation .\n\nWe have recently mapped a putative senescence gene(s) on the X chromosome of Chinese hamster embryo ( CHE ) cells .\n\nIn the present study , we have utilized microcell-mediated chromosome transfer ( microcell fusion ) to test whether : ( i ) the human X chromosome exhibits similar genetic potential to induce senescence and ( ii ) the deletion or inactivation of the X-linked senescence gene(s) in CHE cells is associated with nickel-induced immortalization .\n\nA normal CHE or human X chromosome was first introduced into mouse-cell hybrids , then transferred by microcell fusion into a nickel-transformed , immortal male CHE cell line ( Ni-2/TGR ) with an X deletion ( Xq1 ) .\n\nMicrocell fusion of the normal CHE X chromosome into tumorigenic Ni-2/TGR cells yielded senescence of all X recipient clones .\n\nThe normal human X chromosome induced dominant senescence of tumorigenic Ni-2/TGR cells in only 17% of the resulting microcell hybrids ( 14/81 ) .\n\nKaryotypic analyses of 13 non-senescing human X chromosome-derived microcell hybrid clones revealed that none of these clones retained the complete X. A normal CHE X chromosome induced senescence of 75% of hybrids obtained with another immortal and tumorigenic nickel-transformed male CHE cell line ( Ni-6/TGR ) , which exhibited no visible deletion of the X chromosome , while the normal human X chromosome , only induced senescence in 19% of these hybrids .\n\nTransfer of the normal CHE or human X chromosome into spontaneously transformed and tumorigenic cell lines , CHO/TGR or V79/TGR , had little or no effect on their growth .\n\nThese data suggest that both human and CHE cells possess similar X-linked genetic activities that regulate the process of cellular senescence , and that in Chinese hamster cells nickel-induced immortalization but not that of CHO or V79 cells is associated with inactivation of an X-linked senescence gene .", "output": "Enabling replicative immortality" }, { "input": "Clones of mortal chicken fibroblasts and erythroblasts transformed by temperature-sensitive v-src and v-erb B oncoproteins have been developed into immortal cell lines that retain the conditional transformed phenotype .\n\nThe expressions of two tumor suppressor genes , the retinoblastoma ( Rb ) gene and the p53 gene , were investigated during senescence , crisis , and cell line establishment .\n\nIn temperature-sensitive ( ts)-v-erb B erythroblasts and ts-v-src fibroblasts ( as well as in v-myc macrophages ) , loss of p53 mRNA or expression of a mutated p53 gene invariably occurred in the early phase of immortalization .\n\nIn contrast , expression of the Rb gene was unchanged at all stages of immortalization .\n\nInactivation of the original temperature-sensitive oncogene led to loss of the transformed phenotype in fibroblasts and to differentiation in erythroblasts , even in lines that were immortal and lacked p53 .\n\nThe results demonstrate that the process of immortalization is distinct from cell transformation , probably requiring different mutational events .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "We assayed the estrogen and progesterone cytosolic receptors by using the enzyme immunoassay method , the epidermal growth factor ( EGF ) cell surface receptors by using 125I-labeled hormone , and the levels of polyamines ( putrescine , spermine , and spermidine ) by using a high-pressure liquid chromatography ( HPLC ) procedure in neoplastic and surrounding normal tissues of patients with colorectal cancer .\n\nOur findings show that mean polyamine levels in neoplastic tissue were approximately two-fold greater than the levels in normal colonic mucosa .\n\nEstrogen and progesterone receptorial content in normal mucosa were twofold greater than those in neoplastic tissue .\n\nNo significant differences in EGF receptors were found between colonic cancer tissue and the surrounding normal tissues .\n\nThe correlations we found between 1 ) estrogen and polyamine levels and 2 ) estrogen and EGF binding suggest the existence of a modulation of the estrogens on colonic mucosa cell proliferation .\n\nFurthermore , there was no significant dependency of polyamine and receptor concentrations from the tumor site , the histologic differentiation , or the age and sex of patients .", "output": "Sustaining proliferative signaling" }, { "input": "Loss of telomeric DNA during cell proliferation may play a role in ageing and cancer .\n\nSince telomeres permit complete replication of eukaryotic chromosomes and protect their ends from recombination , we have measured telomere length , telomerase activity and chromosome rearrangements in human cells before and after transformation with SV40 or Ad5 .\n\nIn all mortal populations , telomeres shortened by approximately 65 bp/generation during the lifespan of the cultures .\n\nWhen transformed cells reached crisis , the length of the telomeric TTAGGG repeats was only approximately 1.5 kbp and many dicentric chromosomes were observed .\n\nIn immortal cells , telomere length and frequency of dicentric chromosomes stabilized after crisis .\n\nTelomerase activity was not detectable in control or extended lifespan populations but was present in immortal populations .\n\nThese results suggest that chromosomes with short ( TTAGGG)n tracts are recombinogenic , critically shortened telomeres may be incompatible with cell proliferation and stabilization of telomere length by telomerase may be required for immortalization .", "output": "Enabling replicative immortality" }, { "input": "Findings of increased numbers of epidermal growth factor receptors ( EGF-R ) and increased expression of transforming growth factor alpha ( TGF-alpha ) in surgical specimens of human renal cell carcinoma have led to the proposal that growth of these tumors may be regulated by TGF-alpha in an autocrine manner .\n\nIn the studies presented here , we have examined this hypothesis using two human renal carcinoma cell lines , SKRC-4 and SKRC-29 .\n\nWe demonstrated that both SKRC-4 and SKRC-29 cells were growth stimulated by greater than 35% when cultured in the presence of TGF-alpha or EGF and were inhibited by 29% to 46% if cultured in the presence of anti-EGF-R monoclonal antibody 225 .\n\nTreatment of cells with TGF-alpha enhanced the levels of expression of EGF-R mRNA and TGF-alpha mRNA .\n\nIn addition , incubation of cells with monoclonal antibody 225 significantly elevated the levels of excreted TGF-alpha species in the culture medium .\n\nOur findings suggest that proliferation of human renal carcinoma cells may be regulated by endogenously produced TGF-alpha and that this regulatory pathway can be interrupted using antibody to its receptor , EGF-R .", "output": "Sustaining proliferative signaling" }, { "input": "The immune system has an important role in tumor appearance and spreading .\n\nOne of the most efficient subpopulations of cytotoxic cells in the destruction of tumors are NK cells .\n\nNK cells are activated and increase their cytotoxic potential and modulate their cytokine production after treatment with IFNgamma , IL-12 , TNFalpha and IL-2 .\n\nThe investigation of the activity of NK cells was performed on peripheral blood lymphocytes ( PBL ) of 16 healthy controls and of 40 patients with metastatic breast carcinoma .\n\nModulation of NK cells was performed with IL-2 , IL-7 , IL-12 , TNFalpha , monoclonal antibodies ( mAb ) for TNFalpha and TNFalpha receptors type I and II , as well as with sera of healthy controls and patients with breast cancer in different clinical stages .\n\nModulating effect of the applied factors after in vitro treatment of PBL was evaluated by the cytotoxic assay using 51chromium .\n\nOur results indicate that IL-2 significantly increased the activity of NK cells of controls and breast cancer patients .\n\nThe sera of patients with advanced breast cancer significantly reduced NK cell activity .\n\nIL-7 , IL-12 and mAb for TNFalpha do not significantly change the activity of NK cells .\n\nThe presence of anti-TNFalpha mAb did not change the inhibitory effect of the sera of breast cancer patients with advanced disease on the activity of NK cells of controls and patients with breast cancer .\n\nBlocking of TNFalpha Rcs with mAbs decrease the reactivity of NK cells for IL-2 .\n\nThe treatment of breast cancer patients with advanced clinical stage of breast cancer with IL-2 , as an additional therapy , could be advantageous , as NK cells after this treatment increase their cytotoxic activity against tumor cells and can improve therapeutical results .", "output": "Avoiding immune destruction" }, { "input": "Considerable debate centers on the optimal treatment for vulvar melanoma , as well as those clinicopathological factors influencing prognosis .\n\nWe reviewed 80 patients with vulvar melanoma seen between 1949 and 1990 .\n\nPrimary tumors were assessed according to Chung ( 47 patients ) and Breslow ( 65 patients ) microstaging systems .\n\nFifty-nine patients ( 76% ) underwent radical vulvectomy , ten patients ( 13% ) had a partial vulvectomy , and nine patients ( 12% ) had a wide local excision .\n\nFifty-six also underwent inguinal node dissection .\n\nMedian follow-up was 193 months .\n\nMedian survival was 63 months .\n\nTen-year survival by Chung level was as follows : I 100% ; II , 81% ; III , 87% ; IV , 11% ; V , 33% .\n\nTen-year survival by tumor thickness was as follows : 0.75 mm , 48% ; 0.75-1.5 mm , 68% ; 1.51-3.0 mm , 44% ; greater than 3.0 mm , 22% .\n\nIncreased depth of invasion was associated with increased incidence of inguinal node metastasis .\n\nCox regression analysis demonstrated prognostic significance for tumor thickness ( P less than 0.001 ) , inguinal node metastasis ( P less than 0.001 ) , and older age at diagnosis ( P less than 0.001 ) .\n\nRadical vulvectomy did not seem to improve survival over less radical procedures .\n\nBased on this experience , we recommend radical local excision for patients with malignant melanoma of the vulva .\n\nPatients who have more than a superficially invasive melanoma should also have inguinal lymph node dissection .", "output": "Activating invasion and metastasis" }, { "input": "Many human cellular and tissue compartments are supersaturated with respect to calcium oxyanion salts .\n\nIn order to prevent the formation of injurious crystals efficient anti-crystallization protective mechanisms must be necessary .\n\nWe suggest that depletion of such systems , particularly in ageing organisms and under conditions of oxidative stress , plays an important role in degenerative and inflammatory diseases , including cancer .", "output": "Tumor promoting inflammation" }, { "input": "Tricyclic antidepressants , such as amitriptyline ( Elavil ) , and the nontricyclic agent , fluoxetine ( Prozac ) , bind to growth-regulatory intracellular histamine receptors , associated with anti-estrogen binding sites in microsomes and nuclei .\n\nThe prototype anti-estrogen binding site/intracellular histamine receptor ligand , N,N-diethyl-2-[4-(phenylmethyl)phenoxy]ethanamine HCl , inhibits normal cell proliferation in vitro but stimulates tumor growth in vivo .\n\nBecause of their structural similarity to N,N-diethyl-2-[4-(phenylmethyl)phenoxy]ethanamine HCl , we carried out studies to determine whether amitriptyline and fluoxetine stimulate tumor growth and/or development in rodents at concentrations relevant to the treatment of human depression ( equivalent human dose range , approximately 100-150 mg/day for amitriptyline and approximately 20-80 mg/day for fluoxetine ) .\n\nAll experiments were performed blinded .\n\nIn studies of growth stimulation of transplantable syngeneic tumors , groups of mice were inoculated s.c. with C-3 fibrosarcoma cells or given i.v. or s.c. injections of B16f10 melanoma cells , followed 24 h later by daily i.p. injections of saline , amitriptyline , or fluoxetine .\n\nTumor latency ( fibrosarcoma ) , aggregate tumor weight ( s.c. injected melanoma ) , or time to death from pulmonary metastasis ( i.v. injected melanoma ) was determined ; drug-induced stimulation of DNA synthesis in C-3 fibrosarcoma cells in vitro was correlated with tumor growth acceleration in vivo .\n\nIn a mammary carcinogenesis model , the effects of chronic saline , amitriptyline , or fluoxetine administration on the rate and frequency of development of mammary tumors in rats fed dimethylbenzanthracene ( DMBA ) were compared .\n\nEight of 20 amitriptyline- or fluoxetine-treated mice developed fibrosarcoma tumors by day 5 , as compared to none of 20 saline controls ( P less than 0.002 ) .\n\nSimilarly , 20 of 21 DMBA-treated rats receiving the antidepressant drugs developed 33 mammary tumors by week 15 as compared to 5 tumors in 4 of 7 DMBA-treated rats receiving saline ( P less than 0.001 ) .\n\nFor both models , tumor latency decreased 30-40% and , in the DMBA model , tumor frequency increased greater than 2-fold in the antidepressant-treated rats as compared to controls .\n\nStimulation of fibrosarcoma growth in vivo correlated with a corresponding bell-shaped drug-induced increase in DNA synthesis in vitro .\n\nWhile the median time to death from pulmonary metastases did not differ among groups given i.v. injections of melanoma cells , a significant ( P less than 0.01 ) stimulation of growth of s.c. injected melanoma was observed in mice receiving the antidepressants.(ABSTRACT TRUNCATED AT 400 WORDS )", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Twelve postmenopausal women ( 54-93 years ) with primary breast carcinoma were treated with tamoxifen due to infirmity or refusal to undergo surgery .\n\nSeven premenopausal patients ( 32-50 years ) were given preoperative chemotherapy because of large tumors or inflammatory carcinoma .\n\nFine-needle aspiration biopsy was used to procure tumor cells for diagnosis , hormone receptor determination and analysis of proliferation fraction .\n\nAspirations were repeated every 3 months in the tamoxifen group and each month in patients receiving chemotherapy .\n\nTwo patients who responded to tamoxifen had tumors with more than 75% estrogen receptor positive cells .\n\nA decreased proliferation fraction was observed in two tumors responding to tamoxifen .\n\nEight patients , all with estrogen receptor positive tumors , had stable disease .\n\nProgressive disease was observed in two patients with less than 25% receptor positive cells .\n\nIn these tumors the percentage of proliferating cells remained high during therapy .\n\nObjective response was recorded for six patients treated with chemotherapy .\n\nThe clinical response was reflected in a decreased proliferation fraction .\n\nNo correlation was observed between response and percentage of proliferating cells in the untreated tumor .\n\nThe results suggest that analysis of tumor cell characteristics such as hormone receptor content and proliferation fraction can be used to predict and monitor response to endocrine treatment and chemotherapy in breast carcinomas .", "output": "Sustaining proliferative signaling" }, { "input": "Because PRL has growth factor activities in several tissues , we have asked whether it also has autocrine growth factor activity in pituitary GH3 cells .\n\nGH3 cells were grown at increasing densities in the presence or absence of antirat PRL ( polyclonal and monoclonal ) or nonspecific antibodies .\n\nCell proliferation increased with increasing cell density , as did the concentration of PRL in the medium .\n\nAntirat PRL , but not control antibody , markedly inhibited but did not eliminate cell proliferation , and this effect was diminished with increasing PRL concentration in the medium .\n\nPRL receptors were demonstrated on 40-50% of the cells by indirect immunofluorescence using a specific antirat PRL receptor monoclonal antibody .\n\nCell surface PRL was colocalized to the same 40-50% of the cells and copatched or cocapped along with the receptors .\n\nAbsence or presence of PRL receptors did not correlate with stage of the cell cycle , as judged by ethidium bromide dual labeling .\n\nCell surface PRL was found to be on PRL-containing cells .\n\nThese data have fulfilled four criteria necessary for establishment of a substance as a secreted autocrine growth factor : 1 ) the factor must be secreted ; 2 ) in log growth phase , increased cell proliferation should occur at increased cell densities ; 3 ) the cells must display a receptor for the factor ; and 4 ) there must be a growth response to the factor .\n\nThus we have established that PRL is an autocrine growth factor for at least 40-50% of the GH3 cell population .\n\nThis , to our knowledge , is the first example of autocrine growth factor activity of a major hormone normotopically expressed .", "output": "Sustaining proliferative signaling" }, { "input": "The effects of human interferon ( IFN)-alpha , -beta , and -gamma on the immortalization of human and rabbit lymphocytes by human T-lymphotropic virus type-I ( HTLV-I ) have been investigated .\n\nThe immortalization of human peripheral-blood lymphocytes co-cultured with lethally X-ray-irradiated HTLV-I-producer cells , MT-2 , was blocked in the presence of more than 40 u/ml human recombinant IFN-alpha or more than 200 u/ml human natural type IFN-beta .\n\nHowever , rhIFN-gamma did not block immortalization by HTLV-I even at higher doses .\n\nOn the other hand , the presence of high doses of hIFN-alpha , -beta , or -gamma did not exhibit any biological effect on the immortalization of rabbit peripheral-blood lymphocytes co-cultured with lethally X-ray-irradiated MT-2 cells .\n\nIntegration of the full length of HTLV-I genome was detected in every transformant by Southern blot analysis .\n\nAll cell lines established were CD4+/CD8 divided by T-lymphocytes , except for one cell line of CD4+/CD8+ .\n\nMorphologically intact HTLV-I production was observed by electron microscopy in these cells .\n\nOur results indicate that HTLV-I released under the strongly suppressed condition in the presence of IFNs remains active and able to immortalize T lymphocytes .\n\nIt is also suggested that immortalization of human T lymphocytes by HTLV-I can be inhibited by the antiviral state induced by the treatment with low doses of hIFN-alpha and -beta , whereas immortalization of rabbit T lymphocytes is not inhibited because of the species specificity of hIFNs .", "output": "Enabling replicative immortality, Avoiding immune destruction" }, { "input": "The study focuses on the effects of combined oral contraceptives ( COCs ) on the onset of cervical dysplasia among Zimbabwean women .\n\nWomen who had used COCs for at least 2 years and were in continued use were compared to non-users of COCs ( control group ) .\n\nIt was difficult to establish the average period of contraceptive use because in most instances there was no proper documentation on the exact dates as to when the subjects started using COCs .\n\nThe number of subjects with each condition was noted from each of the following age groups ; <20 years , 20-29years , 30-39years , 40-49years and >50years .\n\nIt was found that the percentage of the control group with benign conditions was higher than that of COC users in all age groups .\n\nSignificant differences at 95 percent confidence level were noted for the 20-29 years age group ( z= -2.21 ) and 40-49 years age group ( z= -2.53).The number of subjects in the <20 years and >50 years age groups were too small for z-score computation .\n\nNo significant differences were noted for mild to moderate cervical inflammation in all age groups .\n\nThere was a higher percentage of COC users with severe cervical inflammation compared to the control group in all age groups .\n\nSignificant differences were noted in the 30-39 years age group ( z=3.45 ) and 40-49 years age group ( z= 1.98 ) .\n\nA higher percentage of CIN I was noted among pooled COC users compared to the control group ( z= 2.00 ) although no significant differences were obtained within different age groups .\n\nIn conclusion , severe cervical inflammation and CIN I are more frequent among Zimbabwean women who use COCs as compared to non-users of COCs .\n\nFrequencies of advanced CIN are low among women who undergo routine cytological screening because this enables early detection and subsequent treatment .", "output": "Tumor promoting inflammation" }, { "input": "We have constructed plasmids pS3G-1 and pSG4 that contain single acetylaminofluorene adducts within contiguous runs of three ( 5'-CCCG1G2G3-3' ) and four ( 5'-CG1GGG4T-3' ) guanine residues , respectively .\n\nIn Escherichia coli , the frequency of induced -1 frameshift mutations was strongly dependent on the position of modification : pS3G-G3 was approximately 100-fold and 10-fold more mutagenic than pS3G-G1 and pS3G-G2 , respectively ; pSG4-G4 was approximately 600-fold more mutagenic than pSG4-G1 .\n\nMutagenesis was SOS-dependent and was markedly reduced in bacteria that were proficient in nucleotide excision repair as compared to a repair-deficient uvrA6 mutant .\n\nDNA sequencing showed that -1 frameshift events in pS3G-1 consisted of either targeted mutations ( greater than 90% of induced mutations ) within the guanine sequence or semitargeted mutations ( greater than 10% ) in the 5 ' flanking repetitive cytosine sequence .\n\nSemitargeted events , which were observed when acetylaminofluorene modification was at G1 and G2 , show that a lesion can reduce the fidelity of replication at positions 5 ' to its location on the template strand .\n\nNo semitargeted frameshifts were observed in plasmid pSG4 , which lacks a repetitive sequence 5 ' to the adduct .\n\nOur results are consistent with a model for frameshift mutagenesis in which the acetylaminofluorene adduct ( i ) allows accurate incorporation of cytosine opposite the bulky lesion during DNA synthesis and ( ii ) impedes elongation of primer/template termini formed opposite the adduct or 5 ' to the adduct on the template strand , providing increased opportunity for the formation of slipped frameshift intermediates .", "output": "Genomic instability and mutation" }, { "input": "Novel photosensitizers Hypocrellin A ( HA ) and Hypocrellin B ( HB ) , lipid soluble perylquinone derivatives of the genus Hypericum have a strong photodynamic effect on tumors and viruses .\n\nHowever , the mechanisms of tumor cell death induced by HA and HB are still unclear .\n\nIn this study , we attempt to elucidate the photodynamic effects of HA and HB compounds in poorly differentiated ( CNE2 ) and moderately differentiated ( TW0-1 ) human nasopharyngeal carcinoma ( NPC ) cells as well as human mucosal colon ( CCL-220.1 ) and bladder ( SD ) cells .\n\nUsing these cell lines we investigated few hall marks of apoptotic commitments in a drug and light dose dependent manner .\n\nTumor cells photoactivated with HA and HB showed cell size shrinkage and an increase in the sub-diploid DNA content .\n\nA loss of membrane phospholipid asymmetry associated with apoptosis was induced by all tumor cell lines as evidenced by the externalization of phosphatidylserine .\n\nWestern blot analysis of poly ( ADP-ribose ) polymerase , a caspases substrate , showed the classical cleavage pattern ( 116 to 85kDa ) associated with apoptosis in HA and HB-treated cell lysates .\n\nIn addition , PARP cleavage was blocked by using tetrapepdide caspases inhibitors such as DEVD or z-VAD .\n\nThese results demonstrate that tumor cell death induced by HB and HA is mediated by caspase proteases .\n\nThis study also identifies both colon and bladder cells were more sensitive cell lines than NPC ( CNE2 and TWO-1 ) cell lines .", "output": "Resisting cell death" }, { "input": "We have investigated the cytotoxic activity , the induction of apoptosis , and the interstrand cross-linking efficiency in the A2780cisR ovarian tumor cell line , after replacement of the two NH3 nonleaving groups in trans-[PtCl2(NH3)2] ( trans-DDP ) by dimethylamine and isopropylamine .\n\nThe data show that trans-[PtCl2(NH(CH)2)(NHCH(CH3)2)] is able to circumvent resistance to cis-[PtCl2(NH3)2] ( cis-DDP , cisplatin ) in A2780cisR cells .\n\nIn fact , trans-[PtCl2(NH(CH3)2)(NHCH(CH3)2)] shows a cytotoxic potency higher than that of cis-DDP and trans-DDP , with the mean IC50 values being 11 , 58 , and 300 microM , respectively .\n\nIn addition , at equitoxic doses ( concentrations of the platinum drugs equal to their IC50 values ) and after 24 hours of drug treatment , the level of induction of apoptosis by trans-[PtCl2(NH(CH3)2)(NHCH(CH3)2)] is twice that produced by cis-DDP .\n\nUnder the same experimental conditions , trans-DDP does not induce significant levels of apoptosis in A2780cisR cells .\n\nAfter 24 hours of incubation of A2780cisR cells at concentrations equal to the IC0o value of the platinum drugs , the level of DNA interstrand cross-links ( ICLs ) induced by trans-[PtCI2(NH(CH)2)(NHCH(CH3)] is two and three times higher , respectively , than those induced by cis-DDP and trans-DDP .\n\nWe also found that trans-[PtCl2(NH(CH3)2)(NHCH(CH3)2)] formed DNA ICLs between guanine and complementary cytosine .\n\nWe propose that , in A2780cisR cells , the induction of apoptosis by trans-[PtCl2(NH(CH3)2)(NHCH(CH3)2)] is related to its greater ability ( relative to cis-DDP and trans-DDP ) to form DNA ICLs .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "The morphology characteristics of cell apoptosis of the malignant tumour cells in magnetic field-treated mouse was observed for the first time .\n\nThe apoptotic cancer cell contracted , became rounder and divorced from adjacent cells ; the heterochromatin condensed and coagulated together along the inner side of the nuclear membrane ; the endoplasmic reticulums ( ER ) expanded and fused with the cellular membrane ; many apoptotic bodies which were packed by the cellular membrane appeared and were devoured by some lymphocytes and plasma .\n\nApoptosis of cancer cells was detected by terminal deoxynucleotidyl transferase mediatedin situ nick end labeling ( TUNEL ) .\n\nIt was found that the number of apoptosis cancer cells of the sample treated by the magnetic field is more than that of the control sample .\n\nThe growth of malignant tumour in mice was inhibited and the ability of immune cell to dissolve cancer cells was improved by ultralow frequency ( ULF ) pulsed gradient magnetic field ; the nuclei DNA contents decreased , indicating that magnetic field can block DNA replication and inhibit mitosis of cancer cells .\n\nIt was suggested that magnetic field could inhibit the metabolism of cancer cell , lower its malignancy , and restrain its rapid and heteromorphic growth .\n\nSince ULF pulsed gradient magnetic field can induce apoptosis of cancer cells and inhibit the growth of malignant tumour , it could be used as a new method to treat cancer .", "output": "Resisting cell death" }, { "input": "ABSTRACT Partial DNA fragments of Botrytis cinerea field isolates encoding the putative osmosensor histidine kinase gene ( BcOS1 ) were cloned by polymerase chain reaction amplification and the predicted amino acid sequences were compared between dicarboximide-sensitive and resistant field isolates .\n\nThe predicted BcOS1p is highly homologous to osmosensor histidine kinase OS1p from Neurospora crassa including the N-terminal six tandem repeats of approximately 90 amino acids .\n\nFour dicarboximide-resistant isolates of B. cinerea ( Bc-19 , Bc-45 , Bc-682 , and Bc-RKR ) contained a single base pair mutation in their BcOS1 gene that resulted in an amino acid substitution in the predicted protein .\n\nIn these resistant isolates , codon 86 of the second repeat , which encodes an isoleucine residue in sensitive strains , was converted to a codon for serine .\n\nThe mutation of Botrytis field resistant isolates was located on the second unit of tandem amino acid repeats of BcOS1p , whereas the point mutations of the fifth repeat of OS1p confer resistance to both dicarboximides and phenylpyrroles and also osmotic sensitivity in Neurospora crassa .\n\nThese results suggest that an amino acid substitution within the second repeat of BcOS1p is responsible for phenotypes of field resistant isolates ( resistant to dicarboximides but sensitive to phenylpyrroles , and normal osmotic sensitivity ) in B. cinerea .", "output": "Genomic instability and mutation" }, { "input": "An aqueous extract of Kefir , fermented milk originally produced in the Caucasus mountains , suppressed morphological changes of human melanoma HMV-1 and SK-MEL cells and human normal fibroblastTIG-1 cells caused by UVC-irradiation , suggesting that UV damage can be suppressed by the Kefir extract .\n\nThe addition of the Kefir extract after UVC-irradiation of HVM-1 cells resulted in a remarkable decrease in intracellular reactive oxygen species ( ROS ) which had been increased by UVC irradiation .\n\nThe Kefir extract also stimulated unscheduled DNA synthesis and suppressed UVC-induced apoptosis of HMV-1 cells .\n\nA colony formation assay revealed that the Kefir extract rescued HMV-1 cells from cell death caused by UVC irradiation .\n\nThe Kefir extract , as well as methyl methanethiosulfonate which is known to enhance the nucleotide excision repair ( NER ) activity , exhibited strong thymine dimer repair-enhancing activity .\n\nEpigalocatechin exhibited a weak NER activity but vitamins A , C , and E and catechin showed no NER activity .\n\nThe thymine dimer repair-enhancing factors in the Kefir extract were heat-stable and assumed to be molecules with a molecular weight of less than 5000 .\n\nThe treatment of HMV-1 cells with the Kefir extract during or before UVC- irradiation also prevented the generation of ROS and thymine dimmer , and suppressed the apoptosis of HMV-1 cells , suggesting that application of Kefir can prevent UV damage .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Resisting cell death" }, { "input": "Biomaterials such as polyetherurethans ( PEUs ) are the scaffolding , which is indispensable for the development of the bio-artificial organs .\n\nHowever , PEUs can induce tumors in subcutaneous implantation sites in rat .\n\nWe have shown that the different inhibitory potential of gap junctional intercellular communication ( GJIC ) on the surface of the biomaterials , including PEUs , is a key step in determining the tumorigenic potential .\n\nHere we show that suppression of a gap junctional protein connexin 43 ( Cx43 ) plays an important role in in vivo tumorigenesis induced by PEUs for the first time and that Cx43 transfection may be an effective strategy for preventing tumorigenesis induced by biomaterials .\n\nRat tumor cell line U41 is derived from tumors in the subcutaneous implantation of PEU films .\n\nThe GJIC and the expression of Cx43 were suppressed in U41 .\n\nThe restoration of normal phenotype , such as reduction of growth rate , recovery of contact inhibition and loss of colony formation ability in soft agar , was achieved by Cx43 transfection .\n\nThese results strongly suggest that suppression of Cx43 expression plays an important role in the development of rat malignant fibrous histiocytoma ( MFHC ) caused by PEUs and that Cx43 transfection is effective for prevention of tumorigenesis induced by PEUs .", "output": "Evading growth suppressors" }, { "input": "The involvement of iron and inflammation parameters on overall survival in non-small-cell lung cancer ( NSCLC ) patients was studied .\n\nFurthermore , transferrin receptors 1 ( TfR1 ) and ferritin expression in tumor tissue , tumor stroma , and normal lung tissue were analyzed .\n\nIron metabolism and inflammation parameters were determined by automated laboratory measurements at the time of diagnosis .\n\nTfR1 and ferritin expression were determined by immuno-histochemical methods .\n\nAbout 50% of patients survived 12 months only .\n\nAt the time of diagnosis more than half of the patients had anemia and significantly elevated serum ferritin .\n\nIron content of serum ferritin ( ICF ) was below the reference values in 90% of patients .\n\nFurthermore , ICF showed positive correlation with iron metabolic parameters and survival but negative correlation with serum ferritin and ESR .\n\nThe expression of TfR1 and ferritin in tumor cells was observed in 88% or 62% of patients , respectively .\n\nTumor stroma was TfR1 negative and sporadically ferritin positive .\n\nTumor tissue ferritin expression showed negative correlation with serum iron and hematokrit ( Ht ) , and positive correlation with ferritin , erythrocyte sedimentation rate ( ESR ) , alpha-1 globulin , and alpha-2 globulin .\n\nPositive correlation was found between TfR1 expression in tumor tissue and alpha-globulin .\n\nThe correlation between TfR1/ferritin expression in tumor tissue and ICF or survival was not observed .\n\nTherefore , we conclude that elevated serum ferritin in sera of NSCLC patients is the result of inflammation and oxidative stress rather than body iron overload .\n\nHigher expression of ferritin in tumor tissue may be the consequence of iron deficiency or local toxicity induced by environmental factors .", "output": "Tumor promoting inflammation" }, { "input": "The glycolytic phenotype is a widespread phenomenon in solid cancer forms , including breast cancer .\n\nDichloroacetate ( DCA ) has recently been proposed as a novel and relatively non-toxic anti-cancer agent that can reverse the glycolytic phenotype in cancer cells through the inhibition of pyruvate dehydrogenase kinase .\n\nWe have examined the effect of DCA against breast cancer cells , including in a highly metastatic in vivo model .\n\nThe growth of several breast cancer cell lines was found to be inhibited by DCA in vitro .\n\nFurther examination of 13762 MAT rat mammary adenocarcinoma cells found that reversal of the glycolytic phenotype by DCA correlated with the inhibition of proliferation without any increase in cell death .\n\nThis was despite a small but significant increase in caspase 3/7 activity , which may sensitize cancer cells to other apoptotic triggers .\n\nIn vivo , DCA caused a 58% reduction in the number of lung metastases observed macroscopically after injection of 13762 MAT cells into the tail vein of rats ( P = 0.0001 , n > or = 9 per group ) .\n\nThese results demonstrate that DCA has anti-proliferative properties in addition to pro-apoptotic properties , and can be effective against highly metastatic disease in vivo , highlighting its potential for clinical use .", "output": "Activating invasion and metastasis, Cellular energetics, Resisting cell death" }, { "input": "Hexavalent chromium ( Cr(VI) ) compounds are known human carcinogens associated with the incidence of lung cancer .\n\nAlthough a direct correlation between Cr(VI) exposure and lung cancer has been established , several studies aimed at generating animal models for Cr(VI) have yielded inconsistent data that do not affirmatively support findings from epidemiologic studies .\n\nBecause the lack of a good animal model has hindered the identification of molecular mechanisms involved in Cr(VI) exposure , we developed an in vitro model that facilitates mechanistic studies of Cr(VI)-induced carcinogenesis .\n\nWe report here that long-term exposure to Cr(VI) leads to the malignant transformation of nontumorigenic human lung epithelial cells .\n\nCr(VI)-transformed cells exhibited loss of contact inhibition , colony formation , and increased rates of cell invasion , migration , and proliferation , as compared with passage-matched control cells .\n\nCr(VI)-transformed cells evaded apoptosis by a mechanism involving S-nitrosylation and stabilization of Bcl-2 protein in a nitric oxide-dependent manner .\n\nThis study establishes an important in vitro model that facilitates mechanistic studies of Cr(VI)-induced carcinogenesis , and elucidates a novel mechanism that causes apoptosis-resistant malignant transformation of nontumorigenic lung cells in response to a human carcinogen .", "output": "Resisting cell death, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Ferric nitrilotriacetate ( Fe-NTA ) is a potent nephrotoxicant and a renal carcinogen that induces its effect by causing oxidative stress .\n\nThe present study was undertaken to explore protective effect of silymarin , a flavonolignan from milk thistle ( Silybum marianum ) , against Fe-NTA mediated renal oxidative stress , inflammation and tumor promotion response along with elucidation of the implicated mechanism(s) .\n\nAdministration of Fe-NTA ( 10 mg/kg bd wt , i.p. ) to Swiss albino mice induced marked oxidative stress in kidney , evident from augmentation in renal metallothionein ( MT ) expression , depletion of glutathione content and activities of antioxidant and phase II metabolizing enzymes , and enhancement in production of aldehyde products such as 4-hydroxy-2-nonenal .\n\nFe-NTA also significantly activated nuclear factor kappa B ( NFkappaB ) and upregulated the expression of downstream genes : cyclooxygenase 2 and inducible nitric oxide synthase and enhancing the production of proinflammatory cytokines : tumor necrosis factor alpha ( TNF-alpha ) and interleukin-6 ( IL-6 ) .\n\nHowever , feeding of 0.5% and 1% silymarin diet conferred a significant protection against Fe-NTA induced oxidative stress and inflammation .\n\nIt further augmented MT expression , restored the antioxidant armory , ameliorated NFkappaB activation and decreased the expression of proinflammatory mediators .\n\nSilymarin also suppressed Fe-NTA induced hyperproliferation in kidney , ameliorating renal ornithine decarboxylase activity and DNA synthesis .\n\nFrom these results , it could be concluded that silymarin markedly protects against chemically induced renal cancer and acts plausibly by virtue of its antioxidant , anti-inflammatory and antiproliferative activities .", "output": "Tumor promoting inflammation" }, { "input": "Nucling is a stress-inducible protein associated with apoptosomes .\n\nThe cytochrome c-triggered formation of apoptosomes represents a key-initiating event in apoptosis .\n\nWe have recently reported that Nucling regulates the apoptotic pathway by controlling the activation of NF-kappaB as well .\n\nHere we show that hepatocellular carcinoma ( HCC ) arising spontaneously against a background of hepatitis occurred more frequently in Nucling-knockout ( KO ) mice than wild-type ( WT ) mice .\n\nBiochemical serum testing revealed potential liver dysfunction with hypercholesterolemia in Nucling-KO males .\n\nIn the background of Nucling-KO mice , we observed the up-regulation of TNFalpha , spontaneous NF-kappaB-activation and the induction of galectin-3 expression in liver .\n\nIn addition , we observed a decrease in the number of Kupffer cells ( KCs ) in the KO mice .\n\nKCs are important for the hepatic immune system , acting as phagocytes or antigen-presenting cells ( APCs ) .\n\nWe found that KCs in Nucling-KO mice were apoptotic possibly through the up-regulation of TNFalpha .\n\nThese observations indicate that Nucling is important for the regulation of NF-kappaB signals in liver .\n\nWe propose that Nucling deficiency could be a powerful tool to reveal the NF-kappaB-related molecular networks leading to hepatitis and HCC development .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "PURPOSE Metabolic dependence on glucose utilisation has been described for different tumours characterised by activation of Akt , upregulation of GLUT1 , M2PK and TKTL1 .\n\nTo date , however , little is known about glucose metabolism in breast cancer tissue .\n\nMETHODS We analysed 55 breast cancer specimens , 26 adjacent ductal carcinomas in situ ( DCIS ) and 23 adjacent normal breast tissues for expression of glycolytic markers by immunohistochemistry .\n\nRESULTS We found expression of pAkt in 49% , GLUT1 in 25% , M2PK in 68% and TKTL1 in 31% of the tumours investigated .\n\nExpression of pAkt and Her2neu are positively correlated with borderline significance ( P = 0.055 ) .\n\nExpression of pAkt , GLUT1 and TKTL1 were higher in breast cancer and DCIS than in normal tissue .\n\nSurprisingly , M2PK expression was highest in normal breast tissue .\n\nCONCLUSIONS We found a glycolytic phenotype in a high percentage of breast cancer samples .\n\nInhibition of glycolysis might evolve as a future option for breast cancer therapy .", "output": "Cellular energetics" }, { "input": "BACKGROUND & AIMS Hepatocellular carcinoma ( HCC ) is an aggressive malignancy with few treatment options .\n\nAs the status of the tumour immune microenvironment can affect progression of established tumours , we evaluated potential immune mechanisms associated with survival in HCC .\n\nMETHODS Immune gene expression profiles were analyzed in tumour and non-tumour liver tissues from resected HCC patients using quantitative PCR and immunohistochemistry .\n\nTumour-infiltrating leukocytes ( TILs ) were isolated to verify the expression of immune genes and to identify proliferating TILs .\n\nThese parameters were analyzed statistically in relation with patient survival and tumour phenotype ( apoptosis and proliferation ) .\n\nRESULTS The immune microenvironment within tumours was found to be heterogeneous , although globally more inert compared to the adjacent non-tumour liver tissue .\n\nUnivariate analysis in 61 patients identified a group of innate immune genes whose expression within tumours is positively associated with patient survival .\n\nTNF , IL6 and CCL2 are the most significant genes , with TNF being an independent predictor of survival in multivariate analysis .\n\nThe gene set includes macrophage and NK-associated molecules such as TLR4 , TLR3 , CCR2 , NCR3 .\n\nMost of these molecules are expressed by TILs .\n\nImportantly , proliferating immune cells , predominantly NK and T cells , are present in tumours of patients with longer survival , and exclusively in areas devoid of proliferating tumour cells .\n\nNK and CD8(+) T cell densities are correlated positively with tumour apoptosis , and negatively with tumour proliferation .\n\nCONCLUSIONS Hence , an inflammatory immune microenvironment within HCC tumours could be an important means to control tumour progression via TIL activation and proliferation .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "BACKGROUND & AIMS GUCY2C is the intestinal receptor for the paracrine hormones guanylin and uroguanylin that converts guanosine-5'-triphosphate to cyclic guanosine monophosphate ( cGMP ) .\n\nIt functions as a tumor suppressor ; its loss disrupts intestinal homeostasis and promotes tumorigenesis .\n\nWe investigated the effects of GUCY2C loss on intestinal cell proliferation , metabolism , signaling , and tumorigenesis in mice .\n\nMETHODS Intestinal cell proliferation and metabolism were examined in Gucy2c(-/-) and colon cancer cells by microscopy , immunoblot , and functional analyses .\n\nMicroarray analyses compared gene expression profiles of intestine cell from Gucy2c(-/-) and wild-type mice. v akt murine thymoma viral oncogene homolog ( AKT ) regulation and signaling were examined , and the role of AKT in GUCY2C-dependent tumorigenesis was defined in Gucy2c(-/-)Akt1(-/-) mice .\n\nRESULTS The size and number of intestinal crypts increased in Gucy2c(-/-) mice ; the associated epithelial cells showed accelerated proliferation , increased glycolysis , and reduced oxidative phosphorylation , which was reversed by oral administration of cGMP .\n\nConversely , activating guanylyl cyclase C in human colon cancer cells delayed cell-cycle progression , decreased DNA synthesis and colony formation , reduced glycolysis , and increased mitochondrial adenosine triphosphate production .\n\nAKT signaling pathways were activated in intestines of Gucy2c(-/-) mice , associated with increased AKT phosphorylation .\n\nDisruption of AKT activity , pharmacologically or genetically , reduced DNA synthesis , proliferation , and glycolysis , and increased mitochondrial biogenesis .\n\nIntestinal tumorigenesis increased after administration of azoxymethane to Gucy2c(-/-) mice , compared with wild-type mice , but was eliminated in Gucy2c(-/-)Akt1(-/-) mice .\n\nCONCLUSIONS GUCY2C is a tumor suppressor that controls proliferation and metabolism of intestinal epithelial cells by inactivating AKT signaling .\n\nThis receptor and its ligands , which are paracrine hormones , might be novel candidates for anticolorectal cancer therapy .", "output": "Cellular energetics" }, { "input": "Inflammatory response plays an important role not only in the normal physiology but also in the pathology such as cancers .\n\nAs chronic inflammations are associated with malignancies , it is important to prevent inflammation-mediated neoplastic formation , promotion and/or progression .\n\nOne possible intervention will be using cancer chemopreventive agents such as curcumin ( CUR ) , a potent anti-inflammatory and anti-oxidative stress compound .\n\nPolyunsaturated fatty acids ( PUFA ) such as docosahexaenoic acid ( DHA ) or eicosapentaenoic acid ( EPA ) are potent anti-inflammatory agents by decreasing the production of inflammatory eicosanoids , cytokines , and reactive oxygen species ( ROS ) .\n\nThe present study aims at examining whether CUR with DHA or EPA would have synergistic anti-inflammatory effects in RAW 264.7 cells .\n\nNon-toxic concentrations of single and combination of the compounds were investigated at 6 , 12 and 24h .\n\nThe nitric oxide ( NO ) suppression effects were most prominent at 24h .\n\nAll the combinations of CUR and DHA or EPA with lower concentrations of CUR 5 microM and 25 microM of DHA or EPA were found to have synergistic effects in suppressing LPS-stimulated NO and endogenous NO levels .\n\nImportantly , very low doses of CUR 2.5 microM and DHA or EPA of 0.78 microM could synergistically suppress the LPS-induced prostaglandin E(2) ( PGE(2) ) .\n\nThe combinations were also found to suppress iNOS , COX-2 , 5-lipoxygenase ( 5-LOX ) and cPLA(2) but induce HO-1 .\n\nTaken together , the present study clearly shows the synergistic anti-inflammatory as well as anti-oxidative stress effects of CUR and PUFA .", "output": "Tumor promoting inflammation" }, { "input": "PTEN loss of function enhances proliferation , but effects on cellular energy metabolism are less well characterized .\n\nWe used an inducible PTEN expression vector in a PTEN-null glioma cell line to examine this issue .\n\nWhile proliferation of PTEN-positive cells was insensitive to increases in glucose concentration beyond 2.5mM , PTEN-null cells significantly increased proliferation with increasing glucose concentration across the normal physiologic range to approximately 10mM , coinciding with a shift to glycolysis and \" glucose addiction \" .\n\nThis demonstrates that the impact of loss of function of PTEN is modified by glucose concentration , and may be relevant to epidemiologic results linking hyperglycemia to cancer risk and cancer mortality .", "output": "Cellular energetics" }, { "input": "Earlier studies indicated that density-arrested cancer cells released an unidentified growth inhibitor whose secretion was prevented by overexpression of the lysosomal protease cathepsin D ( cath D ) .\n\nIn this study , this growth inhibitor was purified by affinity chromatography and identified as the heat shock cognate 70 protein ( hsc70 ) based on its peptide microsequencing and specific antibody recognition .\n\nAmong intracellular proteins , including other heat shock proteins , only constitutive hsc70 was secreted in response to the high-cell density .\n\nMoreover , hsc70 secretion from cancer cells was generated by serum deprivation , whereas its cellular concentration did not change .\n\nPrevention of Hsc70 secretion by cath D overexpression was associated with the formation of multilayer cell cultures , thus indicating a loss of contact inhibition .\n\nIn addition , we showed that supplementing the culture medium with purified hsc70 inhibited cell proliferation in the nanomolar range .\n\nConversely , removal of this extracellular hsc70 from the medium by either retention on ADP-agarose or competition at the Hsc70 binding site restored cell proliferation .\n\nHsc70 appears active in human breast cancer cells and hypersecreted by direct cath D inhibition .\n\nThese results suggest a new role of this secreted hsc70 chaperone in cell proliferation that might account for the higher tumor growth of cancer cells overexpressing cath D .", "output": "Evading growth suppressors" }, { "input": "Manganese , an essential trace nutrient in human beings , has been widely used in the steel industry to improve hardness , stiffness , and strength .\n\nWith the increased applications of manganese compounds , discharge into the environment has rapidly increased and may exert adverse effects on human health .\n\nIn this study , manganese toxicity was investigated using cultured T98G cells , which are derived from human glioblasts with the ability to differentiate into several different types of neuroglia .\n\nCytotoxicity was shown in manganese-treated groups ( 100 , 200 , 400 , and 800microM of MnCl(2) ) , and cell viability was decreased to 58.8% of the control group at 2days after treatment with 800microM of MnCl(2) .\n\nWhen cells were treated with manganese for 24h , ROS dose-dependently increased while antioxidant intracellular GSH decreased .\n\nWith the generation of ROS , the increased activity of caspase-3 was shown , and was followed by chromatin condensation and breakage , which is an indication of the cellular apoptotic process .\n\nROS also triggered pro-inflammatory responses in cultured T98G cells , which were demonstrated by the increased gene expression and protein levels of IL-6 and IL-8 .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Gliomas are aggressive and almost incurable glial brain tumors which frequently display abnormal platelet-derived growth factor ( PDGF ) signaling .\n\nEvidence gained from studies on several in vivo animal models has firmly established a causal connection between aberrant PDGF signaling and the formation of some gliomas .\n\nHowever , only recently has significant knowledge been gained regarding crucial issues such as the glioma cell of origin and the relationship between the transforming stimulus and the cellular characteristics of the resulting tumor .\n\nBased on recent evidence , we propose that PDGF can bias cell-fate decisions , driving the acquisition of cell type-specific features by the progeny of multipotent neural progenitors , thus determining the shape and direction of the transformation path .\n\nFurthermore , recent data about the cellular mechanisms of PDGF-driven glioma progression and maintenance indicate that PDGF may be required , unexpectedly , to override cell contact inhibition and promote glioma cell infiltration rather than to stimulate cell proliferation .", "output": "Evading growth suppressors" }, { "input": "Inflammation which is an indispensable participant in tumor progression is intricately linked with redox modulation .\n\nThe pro-inflammatory cytokine Tumor Necrosis Factor ( TNFalpha ) elevates reactive oxygen species ( ROS ) in glioblastoma multiforme ( GBM ) .\n\nAs both TNFalpha and oxidative stress independently play role in regulating cytoskeletal organization and cell survival pathways we investigated whether TNFalpha mediated oxidative stress regulates responses that offer survival advantages to glioblastoma cells .\n\nTreatment with TNFalpha elevated Akt phosphorylation in glioma cells .\n\nIncreased in Akt phosphorylation was concurrent with the decrease in ROS scavenger SOD-1 levels .\n\nTNFalpha mediated increase in Akt phosphorylation was dependent on oxidative stress as Akt phosphorylation was abrogated in the presence of ROS inhibitor and elevated in cells transfected with SOD-1 siRNA .\n\nTNFalpha altered actin cytoskeletal organization and increased Cdc42 levels .\n\nThis increase in Cdc42 was concomitant with its increased interaction with scaffold protein IQGAP-1 .\n\nAlso , we report for the first time a ROS dependent interaction between pAkt and IQGAP-1 in TNFalpha treated cells .\n\nImportantly , Akt inhibition not only reversed TNFalpha mediated changes in actin cytoskeletal organization but also abrogated anchorage independent growth .\n\nTogether , these results suggest that TNFalpha induced oxidative stress affects Akt activation to regulate actin organization and growth of glioma cells .", "output": "Tumor promoting inflammation" }, { "input": "Polyphenols are widely distributed in plants and known for antioxidant and anti-inflammatory properties .\n\nAreca nut , rich in polyphenols , is the major component of betel quid and we have previously shown that the extract of areca nut can induce oxidative stress in vitro .\n\nIn this study , we have further pinpointed that areca nut extract ( ANE ) contains catechin based procyanidins which range from dimers to decamers and polymers ; this was carried out by HPLC and electrospray ionization/mass spectrometry ( ESI/MS ) .\n\nTo quantify their antioxidant potential , oligomeric and polymeric procyanidins of ANE were separated and evaluated using the Trolox equivalent antioxidant capacity ( TEAC ) assay .\n\nThe results clearly demonstrated that the antioxidant capacity of the ANE procyanidins increased with the degree of polymerization .\n\nThe anti-inflammatory potential of ANE was also tested using 12-O-tetradecanoylphorbol-13-acetate ( TPA)-treated human oral cancer SAS cells .\n\nANE inhibited TPA-induced cyclooxygenase-2 ( COX-2 ) protein expression at low doses , which correlated with the inhibition of ERK phosphorylation in the SAS cells .\n\nFurthermore , feeding rats with ANE at 1 and 10mg/kg/day for 5days significantly repressed carrageenan-induced inflammatory exudates and PGE(2) formation .\n\nIn conclusion , ANE , which contains catechins based oligomeric and polymeric procyanidins , regulates COX-2 expression in vitro and possess anti-inflammatory potential in vivo .", "output": "Tumor promoting inflammation" }, { "input": "Tumor endothelial marker ( TEM ) 5 is an adhesion G-protein-coupled receptor upregulated in endothelial cells during tumor and physiologic angiogenesis .\n\nSo far , the mechanisms leading to upregulation of TEM5 and its function during angiogenesis have not been identified .\n\nHere , we report that TEM5 expression in endothelial cells is induced during capillary-like network formation on Matrigel , during capillary morphogenesis in a three-dimensional collagen I matrix , and upon confluence on a two-dimensional matrix .\n\nTEM5 expression was not induced by a variety of soluble angiogenic factors , including VEGF and bFGF , in subconfluent endothelial cells .\n\nTEM5 upregulation was blocked by toxin B from Clostridium difficile , an inhibitor of the small GTPases Rho , Rac , and Cdc42 .\n\nThe Rho inhibitor C3 transferase from Clostridium botulinum did not affect TEM5 expression , whereas the Rac inhibitor NSC23766 suppressed TEM5 upregulation .\n\nAn excess of the soluble TEM5 extracellular domain or an inhibitory monoclonal TEM5 antibody blocked contact inhibition of endothelial cell proliferation resulting in multilayered islands within the endothelial monolayer and increased vessel density during capillary formation .\n\nBased on our results we conclude that TEM5 expression during capillary morphogenesis is induced by the small GTPase Rac and mediates contact inhibition of proliferation in endothelial cells .", "output": "Inducing angiogenesis, Evading growth suppressors" }, { "input": "In cancer cells , glucose is often converted into lactic acid , which is known as the ' Warburg effect ' .\n\nThe reason that cancer cells have a higher rate of aerobic glycolysis , but not oxidative phosphorylation , remains largely unclear .\n\nHerein , we proposed an epigenetic mechanism of the Warburg effect .\n\nFructose-1,6-bisphosphatase-1 ( FBP1 ) , which functions to antagonize glycolysis was downregulated through NF-kappaB pathway in Ras-transformed NIH3T3 cells .\n\nRestoration of FBP1 expression suppressed anchorage-independent growth , indicating the relevance of FBP1 downregulation in carcinogenesis .\n\nIndeed , FBP1 was downregulated in gastric carcinomas ( P<0.01 , n=22 ) and gastric cancer cell lines ( 57% , 4/7 ) .\n\nRestoration of FBP1 expression reduced growth and glycolysis in gastric cancer cells .\n\nMoreover , FBP1 downregulation was reversed by pharmacological demethylation .\n\nIts promoter was hypermethylated in gastric cancer cell lines ( 57% , 4/7 ) and gastric carcinomas ( 33% , 33/101 ) .\n\nInhibition of NF-kappaB restored FBP1 expression , partially through demethylation of FBP1 promoter .\n\nNotably , Cox regression analysis revealed FBP1 promoter methylation as an independent prognosis predicator for gastric cancer ( hazard ratio : 3.60 , P=0.010 ) .\n\nIn summary , we found that NF-kappaB functions downstream of Ras to promote epigenetic downregulation of FBP1 .\n\nPromoter methylation of FBP1 can be used as a new biomarker for prognosis prediction of gastric cancer .\n\nSuch an important epigenetic link between glycolysis and carcinogenesis partly explains the Warburg effect .", "output": "Cellular energetics" }, { "input": "Chromated copper arsenate , which is used worldwide as a wood preservative , can adversely affect human health .\n\nAccumulating evidence suggests that chromium ( Cr ) and arsenic ( As ) can potentially disrupt the redox balance and cause respiratory diseases and cancer in humans .\n\nThe present study was designed to determine the combined toxic effects of these metals in the lungs and to clarify the specific molecules that are stimulated by combined exposure to both metals .\n\nMale C57BL/6J mice were intratracheally instilled with arsenate [ As(V) ] , hexavalent chromium [ Cr(VI) ] , or a combination of both metals .\n\nMice were sacrificed 2 days after treatment to collect bronchoalveolar lavage fluid and lung tissue samples .\n\nInflammation , cytotoxicity , apoptosis , and oxidative stress markers were measured .\n\nOur results indicated that administration of Cr(VI) alone or in combination with As(V) induced neutrophil-dominant inflammation as well as phosphorylation of mitogen-activated protein kinases ; effects of treatment with As(V) alone were comparatively less potent .\n\nBy analyzing the production of interleukin-6 and activity of lactate dehydrogenase and caspase , we confirmed that co-treatment intensified pulmonary injury and that it was accompanied by oxidative stress , as confirmed by marked increases in the production of reactive oxygen species , reduced glutathione content , and thioredoxin reductase ( TRXRD ) activity .\n\nExpressed mRNA levels of heme oxygenase-1 , glutamylcysteine ligase , glutathione peroxidase 2 , thioredoxin ( TRX ) 1 , and TRXRD1 were also enhanced by co-treatment , whereas treatment with As(V) alone reduced the mRNA expression level of TRX2 .\n\nOur data suggest that co-treatment with As(V) exacerbated Cr(VI)-induced pulmonary injury and that this effect may be exerted through a disruption in the balance among several antioxidant genes .", "output": "Tumor promoting inflammation" }, { "input": "OBJECTIVES Recently , it was revealed that carnosine inhibits growth of cells isolated from human malignant glioma .\n\nIn order to understand how this effect is mediated , experiments were performed that addressed a possible influence of carnosine on energy metabolism .\n\nMETHODS Cells from the glioma line T98G and primary cultured cells from human malignant glioma were cultivated in the presence of carnosine and inhibitors of cellular energy metabolism .\n\nAs a specific inhibitor for anaerobic glycolysis , oxamate , and as an inhibitor for mitochondrial oxidative phosphorylation , potassium cyanide , were used , and the influence on ATP production was determined using cell-based assays .\n\nRESULTS The experiments identified glycolysis as crucial for ATP production in gliomas .\n\nIn addition , ATP production by mitochondrial activity did not significantly contribute to ATP production and carnosine was identified to be an inhibitor of the vital anaerobic glycolysis .\n\nDISCUSSION Carnosine might be considered as a potential drug for the treatment of malignant glioma or other tumors since it inhibits the glycolytic energy metabolism that is crucial for cancer cells and malignant gliomas as shown in the current study .\n\nThis is especially interesting since the dipeptide is a naturally occurring substance that should be well tolerated .", "output": "Cellular energetics" }, { "input": "The aim of the present study was to evaluate immunomodulator ginsan , a polysaccharide extracted from Panax ginseng , on carbon tetrachloride ( CCl(4))-induced liver injury .\n\nBALB/c mice were injected i.p. with ginsan 24 h prior to CCl(4) administration .\n\nSerum liver enzyme levels , histology , expression of antioxidant enzymes , and several cytokines/chemokines were subsequently evaluated .\n\nGinsan treatment markedly suppressed the serum alanine aminotransferase ( ALT ) and aspartate aminotransferase ( AST ) levels , and hepatic histological necrosis increased by CCl(4) treatment .\n\nGinsan inhibited CCl(4) induced lipid peroxidation through the cytochrome P450 2E1 ( CYP2E1 ) downregulation .\n\nThe hepatoprotective effect of ginsan was attributed to induction of anti-oxidant protein contents , such as superoxide dismutase ( SOD ) , catalase , and glutathione peroxidase ( GPX ) as well as restoration of the hepatic glutathione ( GSH ) concentration .\n\nThe marked increase of proinflammatory cytokines ( IL-1beta , IFN-gamma ) and chemokines ( MCP-1 , MIP-2beta , KC ) in CCl(4) treated mice was additionally attenuated by ginsan , thereby preventing leukocyte infiltration and local inflammation .\n\nOur results suggest that ginsan effectively prevent liver injury , mainly through downregulation of oxidative stress and inflammatory response .", "output": "Tumor promoting inflammation" }, { "input": "A common metabolic change in cancer is the acquisition of glycolytic phenotypes .\n\nIncreased expression of glycolytic enzymes is considered as one contributing factor .\n\nThe role of mitochondrial defects in acquisition of glycolytic phenotypes has been postulated but remains controversial .\n\nHere we show that functional defects in mitochondrial respiration could be induced by oncogenic H-Ras(Q61L) transformation , even though the mitochondrial contents or mass was not reduced in the transformed cells .\n\nFirst , mitochondrial respiration , as measured by mitochondrial oxygen consumption , was suppressed in NIH-3T3 cells transformed with H-Ras(Q61L) .\n\nSecond , oligomycin or rotenone did not reduce the cellular ATP levels in the H-Ras(Q61L) transformed cells , suggesting a diminished role of mitochondrial respiration in the cellular energy metabolism .\n\nThird , inhibition of glycolysis with iodoacetic acid reduced ATP levels at a much faster rate in H-Ras(Q61L) transformed cells than in the vector control cells .\n\nThe reduction of cellular ATP levels was reversed by exogenously added pyruvate in the vector control cells but not in H-Ras(Q61L) transformed cells .\n\nFinally when compared to the HRas(Q61L) transformed cells , the vector control cells had increased resistance toward glucose deprivation .\n\nThe increased resistance was dependent on mitochondrial oxidative phosphorylation since rotenone or oligomycin abolished the increased survival of the vector control cells under glucose deprivation .\n\nThe results also suggest an inability of the H-Ras(Q61L) transformed cells to reactivate mitochondrial respiration under glucose deprivation .\n\nTaken together , the data suggest that mitochondrial respiration can be impaired during transformation of NIH-3T3 cells by oncogeneic H-Ras(Q61L) .", "output": "Cellular energetics" }, { "input": "Inhibition of Notch signaling is effective in inhibiting colon tumorigenesis , but targeting specific components of the pathway may provide more effective strategies .\n\nHere we show that the expression of Jagged1 , a ligand for canonical Notch signaling , was restricted to enteroendocrine cells or undetectable in the mucosa of the human small and large intestine , respectively .\n\nIn contrast , increased expression characterized half of human colon tumors , although not all tumors with elevated Wnt signaling displayed elevated Jagged1 .\n\nIncreased Jagged1 was also present in intestinal tumors of Apc(1638N/+) and Apc(Min/+) mice , but to a higher level and more frequently in the former , and in 90% of mouse tumors Notch signaling was elevated when Jagged1 was elevated .\n\nIn the human HT29Cl16E colonic carcinoma cell line , induction of goblet cell differentiation by contact inhibition of growth depended on the loss of Jagged1-mediated Notch activation , with signaling through Notch1 and Notch2 acting redundantly .\n\nTherefore , targeting of Jagged1 could be effective in downregulating Notch signaling in a subset of tumors , but may avoid the limiting gastrointestinal toxicity caused by pharmacological inhibition of Notch signaling .", "output": "Evading growth suppressors" }, { "input": "UNLABELLED ABC transporters like P-glycoprotein ( P-gp/ABCB1 ) are membrane proteins responsible for the transport of toxic compounds out of non-malignant cells and tumor tissue .\n\nAIM To investigate the effect of glycolysis and the tissue redox state on P-gp expression in multicellular tumor spheroids derived from prostate adenocarcinoma cells ( DU-145 ) , glioma cells ( Gli36 ) , and the human cervix carcinoma cell line KB-3-1 transfected with a P-gp-EGFP fusion gene that allows monitoring of P-gp expression in living cells .\n\nDuring cell culture of DU-145 , Gli36 , and KB-3-1 tumor spheroids P-gp expression was observed as well as increased lactate and decreased pyruvate levels and expression of glycolytic enzymes .\n\nInhibition of glycolysis for 24 h by either iodoacetate ( IA ) or 2-deoxy-D-glucose ( 2-DDG ) downregulated P-gp expression which was reversed upon coincubation with the radical scavenger ebselen as shown by semi-quantitative immunohistochemisty in DU-145 and Gli36 tumor spheroids , and by EGFP fluorescence in KB-3-1 tumor spheroids .\n\nConsequently endogenous ROS generation in DU-145 tumor spheroids was increased in the presence of either IA or 2-DDG , which was abolished upon coincubation with ebselen .\n\nExogenous addition of pyruvate significantly reduced ROS generation , increased P-gp expression as well as efflux of the P-gp substrate doxorubicin .\n\nDoxorubicin transport was significantly blunted by 2-DDG and IA , indicating that inhibition of glycolysis reversed the multidrug resistance phenotype .\n\nIn summary our data demonstrate that P-gp expression in tumor spheroids is closely related to the glycolytic metabolism of tumor cells and can be downregulated by glycolysis inhibitors via mechanisms that involve changes in the cellular redox state .", "output": "Cellular energetics" }, { "input": "Reactive oxygen species ( ROS ) such as hydrogen peroxide ( H(2)O(2) ) , O(*-)(2) and OH(*) participate in the pathogenesis of ischemia/reperfusion injury , inflammation and atherosclerosis .\n\nOur previous studies have suggested that increased angiotensin II ( Ang II)-forming chymase may be involved in the development of atherosclerosis .\n\nHowever , the regulatory mechanism of chymase expression has not yet been clarified .\n\nIn this study , we tested whether oxidative stress upregulates mouse mast cell proteinase chymase , mouse mast cell proteinase ( MMCP)-5 or MMCP-4 .\n\nWe also examined the expression and activity of these proteins after treatment .\n\nCultured mouse mastocytoma cells ( MMC ) displaying chymase-dependent Ang II-forming activity were treated with H(2)O(2) and several aminothiols with or without anti-oxidants .\n\nThe levels of MMCP-5 and MMCP-4 expression were determined by quantitative RT-PCR ; the level of chymase-dependent Ang II-forming activity was measured by high performance liquid chromatography using Ang I as a substrate .\n\nTreatment of MMC with homocysteine ( 0.1-3 mmol l(-1) ) significantly increased MMCP-5 and MMCP-4 expression , as well as Ang II-forming activity .\n\nThese effects were significantly inhibited by the addition of catalase and further suppressed by the combination of catalase and superoxide dismutase .\n\nIncubation with hydrogen peroxide alone caused a significant increase in Ang II-forming activity , which was completely suppressed by co-treatment with catalase .\n\nFurthermore , MMCP-5 and MMCP-4 expression levels were drastically suppressed and chymase induction by homocysteine was diminished under the GATA-inhibited condition .\n\nHomocysteine increased mast cell chymase expression and activity through the mechanism of oxidative stress .\n\nOur results suggest that there is a biochemical link between oxidative stress and the local Ang II-forming system .", "output": "Tumor promoting inflammation" }, { "input": "Energy deregulation and abnormalities of tumor cell metabolism are critical issues in understanding cancer .\n\nHereditary leiomyomatosis renal cell carcinoma ( HLRCC ) is an aggressive form of RCC characterized by germline mutation of the Krebs cycle enzyme fumarate hydratase ( FH ) , and one known to be highly metastatic and unusually lethal .\n\nThere is considerable utility in establishing preclinical cell and xenograft models for study of disorders of energy metabolism , as well as in development of new therapeutic approaches targeting of tricarboxylic acid ( TCA ) cycle enzyme-deficient human cancers .\n\nHere we describe a new immortalized cell line , UOK 262 , derived from a patient having aggressive HLRCC-associated recurring kidney cancer .\n\nWe investigated gene expression , chromosome profiles , efflux bioenergetic analysis , mitochondrial ultrastructure , FH catabolic activity , invasiveness , and optimal glucose requirements for in vitro growth .\n\nUOK 262 cells have an isochromosome 1q recurring chromosome abnormality , i(1)(q10) , and exhibit compromised oxidative phosphorylation and in vitro dependence on anaerobic glycolysis consistent with the clinical manifestation of HLRCC .\n\nThe cells also display glucose-dependent growth , an elevated rate of lactate efflux , and overexpression of the glucose transporter GLUT1 and of lactate dehydrogenase A ( LDHA ) .\n\nMutant FH protein was present primarily in edematous mitochondria , but with catalytic activity nearly undetectable .\n\nUOK 262 xenografts retain the characteristics of HLRCC histopathology .\n\nOur findings indicate that the severe compromise of oxidative phosphorylation and rapid glycolytic flux in UOK 262 are an essential feature of this TCA cycle enzyme-deficient form of kidney cancer .\n\nThis tumor model is the embodiment of the Warburg effect .\n\nUOK 262 provides a unique in vitro and in vivo preclinical model for studying the bioenergetics of the Warburg effect in human cancer .", "output": "Cellular energetics" }, { "input": "Most cancer cells exhibit increased glycolysis for generation of their energy supply .\n\nThis specificity could be used to preferentially kill these cells .\n\nIn this study , we identified the signaling pathway initiated by glycolysis inhibition that results in sensitization to death receptor ( DR)-induced apoptosis .\n\nWe showed , in several human cancer cell lines ( such as Jurkat , HeLa , U937 ) , that glucose removal or the use of nonmetabolizable form of glucose ( 2-deoxyglucose ) dramatically enhances apoptosis induced by Fas or by tumor necrosis factor-related apoptosis-inducing ligand .\n\nThis sensitization is controlled through the adenosine monophosphate ( AMP)-activated protein kinase ( AMPK ) , which is the central energy-sensing system of the cell .\n\nWe established the fact that AMPK is activated upon glycolysis block resulting in mammalian target of rapamycin ( mTOR ) inhibition leading to Mcl-1 decrease , but no other Bcl-2 anti-apoptotic members .\n\nInterestingly , we determined that , upon glycolysis inhibition , the AMPK-mTOR pathway controlled Mcl-1 levels neither through transcriptional nor through posttranslational mechanism but rather by controlling its translation .\n\nTherefore , our results show a novel mechanism for the sensitization to DR-induced apoptosis linking glucose metabolism to Mcl-1 downexpression .\n\nIn addition , this study provides a rationale for the combined use of DR ligands with AMPK activators or mTOR inhibitors in the treatment of human cancers .", "output": "Cellular energetics, Resisting cell death" }, { "input": "Depleted uranium ( DU ) is commonly used in military armor and munitions , and thus , exposure of soldiers and noncombatants is frequent and widespread .\n\nPrevious studies have shown that DU has both chemical and radiological toxicity and that the primary route of exposure of DU to humans is through inhalation and ingestion .\n\nHowever , there is limited research information on the potential carcinogenicity of DU in human bronchial cells .\n\nAccordingly , we determined the neoplastic transforming ability of particulate DU to human bronchial epithelial cells ( BEP2D ) .\n\nWe observed the loss of contact inhibition and anchorage independent growth in cells exposed to DU after 24 h .\n\nWe also characterized these DU-induced transformed cell lines and found that 40% of the cell lines exhibit alterations in plating efficiency and no significant changes in the cytotoxic response to DU .\n\nCytogenetic analyses showed that 53% of the DU-transformed cell lines possess a hypodiploid phenotype .\n\nThese data indicate that human bronchial cells are transformed by DU and exhibit significant chromosome instability consistent with a neoplastic phenotype .", "output": "Evading growth suppressors" }, { "input": "Odontogenic tumors originate from the remains of migrating enamel epithelium after the completion of normal tooth genesis .\n\nThese enamel epithelium remnants exhibit the ability to recapitulate the events that occur during tooth formation .\n\nSeveral lines of evidence suggest that aberrance in the signaling pathways similar to the ones that are used during tooth development , including the WNT pathway , might be the cause of odontogenic tumorigenesis and maintenance .\n\nIn this study we demonstrated that WNT5A expression was intense in both the epithelial component of ameloblastomas , the most common epithelial odontogenic tumor , and in this tumor's likely precursor cell , the enamel epithelium located at the cervical loop of normal developing human tooth buds .\n\nAdditionally , when WNT5A was overexpressed in enamel epithelium cells ( LS-8 ) , the clones expressing high levels of WNT5A ( S ) exhibited characteristics of tumorigenic cells , including growth factor independence , loss of anchorage dependence , loss of contact inhibition , and tumor formation in immunocompromised mice .\n\nMoreover , overexpression of WNT5A drastically increased LS-8 cell migration and actin reorganization when compared with controls .\n\nSuppression of endogenous WNT5A in LS-8 cells ( AS ) greatly impaired their migration and AS cells failed to form significant actin reorganization and membrane protrusion was rarely seen .\n\nTaken together , our data indicate that WNT5A signaling is important in modulating tumorigenic behaviors of enamel epithelium cells in ameloblastomas .", "output": "Evading growth suppressors" }, { "input": "A fully intact immune system would be expected to hinder the efficacy of oncolytic virotherapy by inhibiting viral replication .\n\nSimultaneously , however , it may also enhance antitumor therapy through initiation of proinflammatory , antiviral cytokine responses at the tumor site .\n\nThe aim of this study was to investigate the role of a fully intact immune system on the antitumor efficacy of an oncolytic virus .\n\nIn this respect , injection of oncolytic vesicular stomatitis virus ( VSV ) into subcutaneous B16ova melanomas in C57Bl/6 mice leads to tumor regression , but it is not associated with viral replicative burst in the tumor .\n\nIn contrast , intratumoral delivery of VSV induces an acute proinflammatory reaction , which quickly resolves concomitantly with virus clearance .\n\nConsistent with the hypothesis that therapy may not be dependent on the ability of VSV to undergo progressive rounds of replication , a single-cycle VSV is equally effective as a fully replication-competent VSV , whereas inactivated viruses do not generate therapy .\n\nEven though therapy is dependent on host CD8+ and natural killer cells , these effects are not associated with interferon-gamma-dependent responses against either the virus or tumor .\n\nThere is , however , a strong correlation between viral gene expression , induction of proinflammatory reaction in the tumor and in vivo therapy .\n\nOverall , our results suggest that acute innate antiviral immune response , which rapidly clears VSV from B16ova tumors , is associated with the therapy observed in this model .\n\nTherefore , the antiviral immune response to an oncolytic virus mediates an intricate balance between safety , restriction of oncolysis and , potentially , significant immune-mediated antitumor therapy .", "output": "Tumor promoting inflammation" }, { "input": "The high glucose consumption of tumor cells even in an oxygen-rich environment , referred to as the Warburg effect , has been noted as a nearly universal biochemical characteristic of cancer cells .\n\nTargeting the glycolysis pathway has been explored as an anti-cancer therapeutic strategy to eradicate cancer based on this fundamental biochemical property of cancer cells .\n\nOncoproteins such as Akt and c-Myc regulate cell metabolism .\n\nAccumulating studies have uncovered various molecular mechanisms by which oncoproteins affect cellular metabolism , raising a concern as to whether targeting glycolysis will be equally effective in treating cancers arising from different oncogenic activities .\n\nHere , we established a dual-regulatable FL5.12 pre-B cell line in which myristoylated Akt is expressed under the control of doxycycline , and c-Myc , fused to the hormone-binding domain of the human estrogen receptor , is activated by 4-hydroxytamoxifen .\n\nUsing this system , we directly compared the effect of these oncoproteins on cell metabolism in an isogenic background .\n\nActivation of either Akt or c-Myc leads to the Warburg effect as indicated by increased cellular glucose uptake , glycolysis , and lactate generation .\n\nWhen cells are treated with glycolysis inhibitors , Akt sensitizes cells to apoptosis , whereas c-Myc does not .\n\nIn contrast , c-Myc but not Akt sensitizes cells to the inhibition of mitochondrial function .\n\nThis is correlated with enhanced mitochondrial activities in c-Myc cells .\n\nHence , although both Akt and c-Myc promote aerobic glycolysis , they differentially affect mitochondrial functions and render cells susceptible to the perturbation of cellular metabolic programs .", "output": "Cellular energetics" }, { "input": "IL-15 is a pluripotent antiapoptotic cytokine that signals to cells of both the innate and adaptive immune system and is regarded as a highly promising immunomodulatory agent in cancer therapy .\n\nSepsis is a lethal condition in which apoptosis-induced depletion of immune cells and subsequent immunosuppression are thought to contribute to morbidity and mortality .\n\nThis study tested the ability of IL-15 to block apoptosis , prevent immunosuppression , and improve survival in sepsis .\n\nMice were made septic using cecal ligation and puncture or Pseudomonas aeruginosa pneumonia .\n\nThe experiments comprised a 2 x 2 full factorial design with surgical sepsis versus sham and IL-15 versus vehicle .\n\nIn addition to survival studies , splenic cellularity , canonical markers of activation and proliferation , intracellular pro- and antiapoptotic Bcl-2 family protein expression , and markers of immune cell apoptosis were evaluated by flow cytometry .\n\nCytokine production was examined both in plasma of treated mice and splenocytes that were stimulated ex vivo .\n\nIL-15 blocked sepsis-induced apoptosis of NK cells , dendritic cells , and CD8 T cells .\n\nIL-15 also decreased sepsis-induced gut epithelial apoptosis .\n\nIL-15 therapy increased the abundance of antiapoptotic Bcl-2 while decreasing proapoptotic Bim and PUMA .\n\nIL-15 increased both circulating IFN-gamma , as well as the percentage of NK cells that produced IFN-gamma .\n\nFinally , IL-15 increased survival in both cecal ligation and puncture and P. aeruginosa pneumonia .\n\nIn conclusion , IL-15 prevents two immunopathologic hallmarks of sepsis , namely , apoptosis and immunosuppression , and improves survival in two different models of sepsis .\n\nIL-15 represents a potentially novel therapy of this highly lethal disorder .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "High concentrations of lactic acid ( LA ) are found under various pathophysiological conditions and are accompanied by an acidification of the environment .\n\nTo study the impact of LA on TNF secretion , human LPS-stimulated monocytes were cultured with or without LA or the corresponding pH control .\n\nTNF secretion was significantly suppressed by low concentrations of LA ( < or = 10 mM ) , whereas only strong acidification had a similar effect .\n\nThis result was confirmed in a coculture model of human monocytes with multicellular tumor spheroids .\n\nBlocking synthesis of tumor-derived lactate by oxamic acid , an inhibitor of lactate dehydrogenase , reversed the suppression of TNF secretion in this coculture model .\n\nWe then investigated possible mechanisms underlying the suppression .\n\nUptake of [ 3-(13)C]lactate by monocytes was shown by hyphenated mass spectrometry .\n\nAs lactate might interfere with glycolysis , the glycolytic flux of monocytes was determined .\n\nWe added [ 1,2-(13)C(2)]glucose to the culture medium and measured glucose uptake and conversion into [ 2,3-(13)C(2)]lactate .\n\nActivation of monocytes increased the glycolytic flux and the secretion of lactate , whereas oxygen consumption was decreased .\n\nAddition of unlabeled LA resulted in a highly significant decrease in [ 2,3-(13)C(2)]lactate secretion , whereas a mere corresponding decrease in pH exerted a less pronounced effect .\n\nBoth treatments increased intracellular [ 2,3-(13)C(2)]lactate levels .\n\nBlocking of glycolysis by 2-deoxyglucose strongly inhibited TNF secretion , whereas suppression of oxidative phosphorylation by rotenone had little effect .\n\nThese results support the hypothesis that TNF secretion by human monocytes depends on glycolysis and suggest that LA and acidification may be involved in the suppression of TNF secretion in the tumor environment .", "output": "Cellular energetics" }, { "input": "BACKGROUND & AIMS Hepatocytes are considered an exception of the paradigmatic inverse correlation between cell proliferation and terminal differentiation .\n\nIn fact , hepatic vital functions are guaranteed by proliferating parenchymal cells during liver regeneration .\n\nHowever , a fine molecular characterization of the relationship between proliferation and differentiation in hepatocytes has been hampered by the lack of reliable in vivo or in vitro models .\n\nMETHODS The hepatocyte terminal differentiation program was characterized in the immortalized , untransformed and differentiated hepatocytic cell line MMH , using several techniques .\n\nParticularly , two-dimensional difference gel electrophoresis combined to tandem mass spectrometry proteomic approach was used .\n\nCell cycle and cell adhesion properties of MMH have been altered using either myc-overexpression and MEK1/2 inhibition or a constitutive active beta-catenin mutant , respectively .\n\nRESULTS The hepatocyte terminal differentiation program is stimulated by the exit from the cell cycle induced by cell-cell contact .\n\nComparative proteomic analysis of proliferating versus quiescent hepatocytes validated the importance of contact inhibition , identifying 68 differently expressed gene products , representing 49 unique proteins .\n\nNotably , enzymes involved in important liver functions such as detoxification processes , lipid metabolism , iron and vitamin A storage and secretion , anti-inflammatory response and exocytosis were found significantly up-regulated in quiescent hepatocytes .\n\nFinally , we found that : ( i ) cell cycle arrest induced by MEK1/2 inhibition is not sufficient to induce hepatic product expression ; ( ii ) constitutive activation of beta-catenin counteracts the contact inhibition-induced terminal differentiation .\n\nCONCLUSION The hepatocyte terminal differentiation program requires a quiescent state maintained by cell-cell contact through the E-cadherin/beta-catenin pathway , rather than the inhibition of proliferation .", "output": "Evading growth suppressors" }, { "input": "Candida albicans infections are very frequent in cancer patients , whose immune system is often compromised , but whether this fungal pathogen affects cancer progression is unknown .\n\nC. albicans infection involves endogenous production of inflammatory cytokines such as tumour necrosis factor alpha ( TNF-alpha ) and interleukin-18 ( IL-18 ) .\n\nIncreased levels of these cytokines have already been correlated with metastasis of most common cancer types .\n\nIn this study , a well-established model of IL-18-dependent hepatic melanoma metastasis was used to study whether C. albicans can alter the ability of murine B16 melanoma ( B16M ) cells to colonize the liver .\n\nFirst , we determined the ability of intrasplenically ( IS ) injected B16M cells to metastasize into the liver of mice challenged with 5 x 10(4) C. albicans cells by three different routes ( intravenous , IV ; intrasplenic , IS ; or intraperitoneal , IP ) 12 h prior to injection of B16M cells .\n\nWe demonstrated that C. albicans significantly increased metastasis of B16M cells with all three fungal injection routes .\n\nPro-metastatic effects occurred when hepatic colonization with B16M cells place after the peak of TNF-alpha and IL-18 levels had been reached in the hepatic blood of fungal challenged mice .\n\nIn a second set of experiments , mice were fungal challenged 4 days after injection of B16M cells .\n\nIn these mice , C. albicans also potentiated the growth of established micro-metastases .\n\nSignificantly , the fungal challenge had pro-metastatic effects without the C. albicans being able to reach the liver , suggesting that soluble factors can promote metastasis in remote sites .\n\nMouse treatment with antifungal ketoconazol abrogated hepatic TNF-alpha stimulation by C. albicans and prevented the enhancement of hepatic metastasis in fungal challenged-mice .\n\nTherefore , the pro-inflammatory microenvironment generated by the host's systemic response to C. albicans stimulates circulating cancer cells to metastasize in the liver .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "Mutations in the tumor suppressor tuberin ( TSC2 ) are a common factor in the development of lymphangioleiomyomatosis ( LAM ) .\n\nLAM is a cystic lung disease that is characterized by the infiltration of smooth muscle-like cells into the pulmonary parenchyma .\n\nThe mechanism by which the loss of tuberin promotes the development of LAM has yet to be elucidated , although several lines of evidence suggest it is due to the metastasis of tuberin-deficient cells .\n\nHere we show that tuberin-null cells become nonadherent and invasive .\n\nThese nonadherent cells express cleaved forms of \u03b2-catenin .\n\nIn reporter assays , the \u03b2-catenin products are transcriptionally active and promote MMP7 expression .\n\nInvasion by the tuberin-null cells is mediated by MMP7 .\n\nExamination of LAM tissues shows the expression of cleaved \u03b2-catenin products and MMP7 consistent with a model that tuberin-deficient cells acquire invasive properties through a \u03b2-catenin-dependent mechanism , which may underlie the development of LAM .", "output": "Activating invasion and metastasis" }, { "input": "The present study was designed to investigate the protective efficacy of eugenol against skin cancer and probe into the mechanistic aspects .\n\nSkin tumors were initiated by applying 160 nmol DMBA and promoted by twice weekly applications of 8.5 nmol TPA for 28 wk .\n\nAll mice developed tumors by 13 wk of promotion .\n\nHowever , in mice pretreated with 30 microL eugenol , no tumors were detected until 8 wk ( following anti-initiation protocol ) and until 14 wk ( following antipromotion protocol ) of tumor promotion .\n\nPCNA and TUNEL immunohistochemistry of tumors revealed eugenol to ameliorate cell proliferation and elevate apoptosis respectively .\n\nThe effect of eugenol was assessed on specific stages of carcinogenesis .\n\nInitiation with DMBA led to a significant upregulation of p53 expression with a concomitant increase in p21(WAF1) levels in epidermal cells indicating induction of damage to the DNA .\n\nHowever , pretreatment with eugenol led to overexpression of these genes , which probably helped stimulate apoptosis of the initiated cells .\n\nTo ascertain the molecular mechanisms implicated in the antitumor promoting activity of eugenol , its effect was investigated on markers of tumor promotion and inflammation : ODC activity and iNOS and COX-2 expression , and on levels of proinflammatory cytokines ( IL-6 , TNF-alpha , and PGE(2) ) .\n\nEugenol markedly inhibited all .\n\nEugenol also inhibited the upstream signaling molecule : NF-kappaB , which regulates the expression of these genes .\n\nTPA-induced depletion of cutaneous GSH and antioxidant enzymes armory was also precluded by eugenol .\n\nFrom these results , it could be concluded that eugenol markedly protects against chemically induced skin cancer and acts possibly by virtue of its antiproliferative , anti-inflammatory , and antioxidant activities .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors, Tumor promoting inflammation, Resisting cell death" }, { "input": "Histone H3 methylation at lysine 4 ( K4 ) is associated with euchromatic regions and is thought to be important for the transcriptional activation of genes during differentiation .\n\nIn this study , we found that di- and tri-methylation of histone H3 at K4 and acetylation of histones H3 and H4 from the promoter/enhancer to the transcribed region close to the transcription initiation site of the solute carrier family 2 , member 5 ( SLC2A5 ) gene , and its expression , were induced by differentiation of intestine-like Caco-2 cells .\n\nThese effects were accompanied by contact inhibition of cell growth of these cells .\n\nFurthermore , these modifications were induced by co-treatment with a synthetic glucocorticoid hormone dexamethasone and a p44/42 mitogen-activated protein kinase inhibitor PD89059 .\n\nOur results suggest that methylation of histone H3 at K4 and acetylation of histones H3 and H4 are involved in SLC2A5 gene induction associated with intestinal differentiation of Caco-2 cells .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND Inorganic arsenic is a ubiquitous environmental carcinogen affecting millions of people worldwide .\n\nEvolving theory predicts that normal stem cells ( NSCs ) are transformed into cancer stem cells ( CSCs ) that then drive oncogenesis .\n\nIn humans , arsenic is carcinogenic in the urogenital system ( UGS ) , including the bladder and potentially the prostate , whereas in mice arsenic induces multi-organ UGS cancers , indicating that UGS NSCs may represent targets for carcino-genic initiation .\n\nHowever , proof of emergence of CSCs induced by arsenic in a stem cell population is not available .\n\nMETHODS We continuously exposed the human prostate epithelial stem/progenitor cell line WPE-stem to an environmentally relevant level of arsenic ( 5 microM ) in vitro and determined the acquired cancer phenotype .\n\nRESULTS WPE-stem cells rapidly acquired a malignant CSC-like phenotype by 18 weeks of exposure , becoming highly invasive , losing contact inhibition , and hyper-secreting matrix metalloproteinase-9 .\n\nWhen hetero-transplanted , these cells ( designated As-CSC ) formed highly pleomorphic , aggressive tumors with immature epithelial- and mesenchymal-like cells , suggesting a highly pluripotent cell of origin .\n\nConsistent with tumor-derived CSCs , As-CSCs formed abundant free-floating spheres enriched in CSC-like cells , as confirmed by molecular analysis and the fact that only these floating cells formed xeno-graft tumors .\n\nAn early loss of NSC self-renewal gene expression ( p63 , ABCG2 , BMI-1 , SHH , OCT-4 , NOTCH-1 ) during arsenite exposure was sub-sequently reversed as the tumor suppressor gene PTEN was progressively suppressed and the CSC-like phenotype acquired .\n\nCONCLUSIONS Arsenite transforms prostate epithelial stem/progenitor cells into CSC-like cells , indicating that it can produce CSCs from a model NSC population .", "output": "Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Polycyclic aromatic hydrocarbons ( PAHs ) are widely distributed immunotoxic and carcinogenic environmental contaminants , known to affect macrophages .\n\nIn order to identify their molecular targets in such cells , we have analyzed gene expression profile of primary human macrophages treated by the prototypical PAH benzo(a)pyrene ( BaP ) , using pangenomic oligonucleotides microarrays .\n\nExposure of macrophages to BaP for 8 and 24 h resulted in 96 and 1100 genes , differentially expressed by at least a twofold change factor , respectively .\n\nSome of these targets , including the chemokine receptor CXCR5 , the G protein-coupled receptor 35 ( GPR35 ) , and the Ras regulator RASAL1 , have not been previously shown to be affected by PAHs , in contrast to others , such as interleukin-1beta and the aryl hydrocarbon receptor ( AhR ) repressor .\n\nThese BaP-mediated gene regulations were fully validated by reverse transcription-quantitative polymerase chain reaction assays for some selected genes .\n\nTheir bioinformatic analysis indicated that biological functions linked to immunity , inflammation , and cell death were among the most affected by BaP in human macrophages and that the AhR and p53 signaling pathways were the most significant canonical pathways activated by the PAH .\n\nAhR and p53 implications were moreover fully confirmed by the prevention of BaP-related upregulation of some selected target genes by AhR silencing or the use of pifithrin-alpha , an inhibitor of PAH bioactivation-related DNA damage/p53 pathways .\n\nOverall , these data , through identifying genes and signaling pathways targeted by PAHs in human macrophages , may contribute to better understand the molecular basis of the immunotoxicity of these environmental contaminants .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Avoiding immune destruction" }, { "input": "Mitochondrial biogenesis , which depends on nuclear as well as mitochondrial genes , occurs in response to increased cellular ATP demand .\n\nThe nuclear transcriptional factors , estrogen-related receptor alpha ( ERRalpha ) and nuclear respiratory factors 1 and 2 , are associated with the coordination of the transcriptional machinery governing mitochondrial biogenesis , whereas coactivators of the peroxisome proliferator-activated receptor gamma coactivator-1 ( PGC-1 ) family serve as mediators between the environment and this machinery .\n\nIn the context of proliferating cells , PGC-1-related coactivator ( PRC ) is a member of the PGC-1 family , which is known to act in partnership with nuclear respiratory factors , but no functional interference between PRC and ERRalpha has been described so far .\n\nWe explored three thyroid cell lines , FTC-133 , XTC.UC1 and RO 82 W-1 , each characterized by a different mitochondrial content , and studied their behavior towards PRC and ERRalpha in terms of respiratory efficiency .\n\nOverexpression of PRC and ERRalpha led to increased respiratory chain capacity and mitochondrial mass .\n\nThe inhibition of ERRalpha decreased cell growth and respiratory chain capacity in all three cell lines .\n\nHowever , the inhibition of PRC and ERRalpha produced a greater effect in the oxidative cell model , decreasing the mitochondrial mass and the phosphorylating respiration , whereas the nonphosphorylating respiration remained unchanged .\n\nWe therefore hypothesize that the ERRalpha-PRC complex plays a role in arresting the cell cycle through the regulation of oxidative phosphorylation in oxidative cells , and through some other pathway in glycolytic cells .", "output": "Cellular energetics" }, { "input": "BACKGROUND Prostate cancer is the second leading cause of cancer mortality among US men .\n\nEpidemiological evidence suggests that high vitamin D status protects men from prostate cancer and the active form of vitamin D , 1alpha,25 dihydroxyvitamin D3 ( 1,25(OH)2D ) has anti-cancer effects in cultured prostate cells .\n\nStill , the molecular mechanisms and the gene targets for vitamin D-mediated prostate cancer prevention are unknown .\n\nRESULTS We examined the effect of 1,25(OH)2D ( +/- 100 nM , 6 , 24 , 48 h ) on the transcript profile of proliferating RWPE1 cells , an immortalized , non-tumorigenic prostate epithelial cell line that is growth arrested by 1,25(OH)2D ( Affymetrix U133 Plus 2.0 , n = 4/treatment per time and dose ) .\n\nOur analysis revealed many transcript level changes at a 5% false detection rate : 6 h , 1571 ( 61% up ) , 24 h , 1816 ( 60% up ) , 48 h , 3566 ( 38% up). 288 transcripts were regulated similarly at all time points ( 182 up , 80 down ) and many of the promoters for these transcripts contained putative vitamin D response elements .\n\nFunctional analysis by pathway or Gene Set Analysis revealed early suppression of WNT , Notch , NF-kB , and IGF1 signaling .\n\nTranscripts related to inflammation were suppressed at 6 h ( e.g .\n\nIL-1 pathway ) and suppression of proinflammatory pathways continued at later time points ( e.g .\n\nIL-17 and IL-6 pathways ) .\n\nThere was also evidence for induction of anti-angiogenic pathways and induction of transcripts for protection from oxidative stress or maintenance of cell redox homeostasis at 6 h .\n\nCONCLUSIONS Our data reveal of large number of potential new , direct vitamin D target genes relevant to prostate cancer prevention .\n\nIn addition , our data suggests that rather than having a single strong regulatory effect , vitamin D orchestrates a pattern of changes within prostate epithelial cells that limit or slow carcinogenesis .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "BACKGROUND AND AIMS Hepatitis C virus ( HCV)-induced chronic inflammation may induce oxidative stress which could compromise the repair of damaged DNA , rendering cells more susceptible to spontaneous or mutagen-induced alterations , the underlying cause of liver cirrhosis and hepatocellular carcinoma .\n\nIn the current study we examined the induction of reactive oxygen species ( ROS ) resulting from HCV infection and evaluated its effect on the host DNA damage and repair machinery .\n\nMETHODS HCV infected human hepatoma cells were analyzed to determine ( i ) ROS , ( ii ) 8-oxoG and ( iii ) DNA glycosylases NEIL1 , NEIL2 , OGG1 .\n\nLiver biopsies were analyzed for NEIL1 .\n\nRESULTS Human hepatoma cells infected with HCV JFH-1 showed 30-60-fold increases in ROS levels compared to uninfected cells .\n\nLevels of the oxidatively modified guanosine base 8-oxoguanine ( 8-oxoG ) were significantly increased sixfold in the HCV-infected cells .\n\nBecause DNA glycosylases are the enzymes that remove oxidized nucleotides , their expression in HCV-infected cells was analyzed .\n\nNEIL1 but not OGG1 or NEIL2 gene expression was impaired in HCV-infected cells .\n\nIn accordance , we found reduced glycosylase ( NEIL1-specific ) activity in HCV-infected cells .\n\nThe antioxidant N-acetyl cystein ( NAC ) efficiently reversed the NEIL1 repression by inhibiting ROS induction by HCV .\n\nNEIL1 expression was also partly restored when virus-infected cells were treated with interferon ( IFN ) .\n\nHCV core and to a lesser extent NS3-4a and NS5A induced ROS , and downregulated NEIL1 expression .\n\nLiver biopsy specimens showed significant impairment of NEIL1 levels in HCV-infected patients with advanced liver disease compared to patients with no disease .\n\nCONCLUSION Collectively , the data indicate that HCV induction of ROS and perturbation of NEIL1 expression may be mechanistically involved in progression of liver disease and suggest that antioxidant and antiviral therapies can reverse these deleterious effects of HCV in part by restoring function of the DNA repair enzyme/s .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "RAD51 is a key enzyme of homologous recombination and repair of DNA double-strand breaks .\n\nRAD51 mRNA expression levels are significantly increased in laser-microdissected mammary simple carcinomas and their lymph node metastases when compared to adenomas or nonneoplastic mammary gland of the same dog .\n\nHere , RAD51 protein expression was analyzed by immunohistochemistry in paraffin-embedded mammary carcinomas and their lymph node metastases of 40 dogs , adenomas of 48 dogs , and nonneoplastic mammary gland of 88 dogs .\n\nNumber of cells with nuclear RAD51 expression was significantly ( P < or = .05 ) increased in carcinomas when compared to adenomas and metastases .\n\nIn contrast , no significant differences in the number of RAD51-expressing cells were detected when metastases were compared with adenomas and nonneoplastic gland .\n\nRAD51 expression in carcinomas was correlated with expression in metastases but not with histologic grade .\n\nIn conclusion , the increased number of RAD51-expressing cells in carcinomas might indicate genomic instability in these cells .\n\nNevertheless , the increased RAD51 mRNA expression in metastases could not be confirmed by immunohistochemistry .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "Ataxia-telangiectasia mutated ( ATM ) is a high molecular weight protein serine/threonine kinase that plays a central role in the maintenance of genomic integrity by activating cell cycle checkpoints and promoting repair of DNA double-strand breaks .\n\nLittle is known about the regulatory mechanisms for ATM expression itself .\n\nMicroRNAs are naturally existing regulators that modulate gene expression in a sequence-specific manner .\n\nHere , we show that a human microRNA , miR-421 , suppresses ATM expression by targeting the 3'-untranslated region ( 3'UTR ) of ATM transcripts .\n\nEctopic expression of miR-421 resulted in S-phase cell cycle checkpoint changes and an increased sensitivity to ionizing radiation , creating a cellular phenotype similar to that of cells derived from ataxia-telangiectasia ( A-T ) patients .\n\nBlocking the interaction between miR-421 and ATM 3'UTR with an antisense morpholino oligonucleotide rescued the defective phenotype caused by miR-421 overexpression , indicating that ATM mediates the effect of miR-421 on cell cycle checkpoint and radiosensitivity .\n\nOverexpression of the N-Myc transcription factor , an oncogene frequently amplified in neuroblastoma , induced miR-421 expression , which , in turn , down-regulated ATM expression , establishing a linear signaling pathway that may contribute to N-Myc-induced tumorigenesis in neuroblastoma .\n\nTaken together , our findings implicate a previously undescribed regulatory mechanism for ATM expression and ATM-dependent DNA damage response and provide several potential targets for treating neuroblastoma and perhaps A-T .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "Cell proliferation is known to be accompanied by activation of glycolysis .\n\nWe have recently discovered that the glycolysis-promoting enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase , isoform 3 ( PFKFB3 ) , is degraded by the E3 ubiquitin ligase APC/C-Cdh1 , which also degrades cell-cycle proteins .\n\nWe now show in two different cell types ( neoplastic and nonneoplastic ) that both proliferation and aerobic glycolysis are prevented by overexpression of Cdh1 and enhanced by its silencing .\n\nFurthermore , we have coexpressed Cdh1 with PFKFB3--either wild-type or a mutant form resistant to ubiquitylation by APC/C-Cdh1--or with the glycolytic enzyme 6-phosphofructo-1-kinase and demonstrated that whereas glycolysis is essential for cell proliferation , its initiation in the presence of active Cdh1 does not result in proliferation .\n\nOur experiments indicate that the proliferative response , regardless of whether it occurs in normal or neoplastic cells , is dependent on a decrease in the activity of APC/C-Cdh1 , which activates both proliferation and glycolysis .\n\nThese observations have implications for cell proliferation , neoplastic transformation , and the prevention and treatment of cancer .", "output": "Cellular energetics" }, { "input": "The contribution that mitochondrial bioenergetics could have in cancer development is debated .\n\nHere , we have generated HCT116-derived colocarcinoma cell lines expressing different levels of the beta catalytic subunit of the mitochondrial H+-adenosine triphosphate synthase to assess the contribution of mitochondrial bioenergetics in colon cancer progression .\n\nThe generated cells exhibit large ultrastructural , transcriptomic , proteomic and functional differences in their mitochondria and in their in vivo tumor forming capacity .\n\nWe show that the activity of oxidative phosphorylation defines the rate of glucose utilization by aerobic glycolysis .\n\nThe aggressive cellular phenotype , which is highly glycolytic , is bound to the deregulated expression of genes involved in metabolic processes , the regulation of the cell cycle , apoptosis , angiogenesis and cell adhesion .\n\nRemarkably , the molecular and ultrastructural analysis of the tumors derived from the three HCT116 cell lines under study highlight that tumor promotion inevitably requires the selection of cancer cells with a repressed biogenesis and functional activity of mitochondria , i.e. the highly glycolytic phenotype is selected for tumor development .\n\nThe tumor forming potential of the cells is a non-genetically acquired condition that provides the cancer cell with a cell-death resistant phenotype .\n\nAn abrogated mitochondrial respiration contributes to a diminished potential for reactive oxygen species signaling in response to 5-fluorouracil treatment .\n\nTreatment of cancer cells with dichloroacetate partially restores the functional differentiation of mitochondria and promotes tumor regression , emphasizing the reversible nature of the metabolic trait of cancer .", "output": "Cellular energetics" }, { "input": "DNA double-strand breaks ( DSBs ) trigger ATM ( ataxia telangiectasia mutated ) signalling and elicit genomic rearrangements and chromosomal fragmentation if misrepaired or unrepaired .\n\nAlthough most DSB repair is ATM-independent , approximately 15% of ionizing radiation ( IR)-induced breaks persist in the absence of ATM-signalling. 53BP1 ( p53-binding protein 1 ) facilitates ATM-dependent DSB repair but is largely dispensable for ATM activation or checkpoint arrest .\n\nATM promotes DSB repair within heterochromatin by phosphorylating KAP-1 ( KRAB-associated protein 1 , also known as TIF1beta , TRIM28 or KRIP-1 ; ref. 2 ) .\n\nHere , we show that the ATM signalling mediator proteins MDC1 , RNF8 , RNF168 and 53BP1 are also required for heterochromatic DSB repair .\n\nAlthough KAP-1 phosphorylation is critical for 53BP1-mediated repair , overall phosphorylated KAP-1 ( pKAP-1 ) levels are only modestly affected by 53BP1 loss. pKAP-1 is transiently pan-nuclear but also forms foci overlapping with gammaH2AX in heterochromatin .\n\nCells that do not form 53BP1 foci , including human RIDDLE ( radiosensitivity , immunodeficiency , dysmorphic features and learning difficulties ) syndrome cells , fail to form pKAP-1 foci. 53BP1 amplifies Mre11-NBS1 accumulation at late-repairing DSBs , concentrating active ATM and leading to robust , localized pKAP-1 .\n\nWe propose that ionizing-radiation induced foci ( IRIF ) spatially concentrate ATM activity to promote localized alterations in regions of chromatin otherwise inhibitory to repair .", "output": "Genomic instability and mutation" }, { "input": "Diets rich in fruits and vegetables are associated with lower risk of cancer which may be conferred in part by the antioxidant properties of these foods .\n\nHowever , antioxidant supplementation or increased consumption of antioxidant-rich foods has been reported to have inconsistent effects on DNA damage .\n\nThe present work ( the DART study ) investigated the extent of inter-individual variation in DNA damage , the capacity for base excision repair ( BER ) and the responses of both variables to supplementation with an antioxidant supplement for 6 weeks .\n\nThere was a wide inter-individual variation in endogenous lymphocyte DNA strand breaks ( 8-fold variation ) , in damage after a challenge with H2O2 ( 16-fold variation ) and in DNA repair ( 41-fold variation ) measured using the comet assay .\n\nWhen stratified into tertiles according to the pre-supplementation level of endogenous DNA damage , there was a statistically significant decrease in DNA damage after supplementation in the tertile with the highest pre-supplementation level of damage .\n\nThere was no effect of supplementation on BER .\n\nEndogenous DNA damage level before supplementation was significantly different ( P = 0.037 ) between the three genotypes for the Val16Ala single nucleotide polymorphism in manganese superoxide dismutase ( rs4880 ) with individuals homozygous/wild type showing less damage than those carrying the alanine variant .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE Genomic complexity is present in approximately 15% to 30% of all chronic lymphocytic leukemia ( CLL ) and has emerged as a strong independent predictor of rapid disease progression and short remission duration in CLL .\n\nWe conducted this study to advance our understanding of the causes of genomic complexity in CLL .\n\nEXPERIMENTAL DESIGN We have obtained quantitative measurements of radiation-induced apoptosis and radiation-induced ATM autophosphorylation in purified CLL cells from 158 and 140 patients , respectively , and have used multivariate analysis to identify independent contributions of various biological variables on genomic complexity in CLL .\n\nRESULTS Here , we identify a strong independent effect of radiation resistance on elevated genomic complexity in CLL and describe radiation resistance as a predictor for shortened CLL survival .\n\nFurthermore , using multivariate analysis , we identify del17p/p53 aberrations , del11q , del13q14 type II ( invariably resulting in Rb loss ) , and CD38 expression as independent predictors of genomic complexity in CLL , with aberrant p53 as a predictor of approximately 50% of genomic complexity in CLL .\n\nFocusing on del11q , we determined that normalized ATM activity was a modest predictor of genomic complexity but was not independent of del11q .\n\nThrough single nucleotide polymorphism array-based fine mapping of del11q , we identified frequent monoallelic loss of Mre11 and H2AFX in addition to ATM , indicative of compound del11q-resident gene defects in the DNA double-strand break response .\n\nCONCLUSIONS Our quantitative analysis links multiple molecular defects , including for the first time del11q and large 13q14 deletions ( type II ) , to elevated genomic complexity in CLL , thereby suggesting mechanisms for the observed clinical aggressiveness of CLL in patients with unstable genomes .", "output": "Genomic instability and mutation" }, { "input": "Acute endothelial cell apoptosis and microvascular compromise couple gastrointestinal tract irradiation to reproductive death of intestinal crypt stem cell clonogens ( SCCs ) following high-dose radiation .\n\nGenetic or pharmacologic inhibition of endothelial apoptosis prevents intestinal damage , but as the radiation dose is escalated , SCCs become directly susceptible to an alternate cell death mechanism , mediated via ceramide synthase ( CS)-stimulated de novo synthesis of the proapoptotic sphingolipid ceramide , and p53-independent apoptosis of crypt SCCs .\n\nWe previously reported that ataxia-telangiectasia mutated deficiency resets the primary radiation lethal pathway , allowing CS-mediated apoptosis at the low-dose range of radiation .\n\nThe mechanism for this event , termed target reordering , remains unknown .\n\nHere , we show that inactivation of DNA damage repair pathways signals CS-mediated apoptosis in crypt SCCs , presumably via persistent unrepaired DNA double-strand breaks ( DSBs ) .\n\nGenetic loss of function of sensors and transducers of DNA DSB repair confers the CS-mediated lethal pathway in intestines of sv129/B6Mre11(ATLD1/ATLD1) and C57BL/6(Prkdc/SCID) ( severe combined immunodeficient ) mice exposed to low-dose radiation .\n\nIn contrast , CS-mediated SCC lethality was mitigated in irradiated gain-of-function Rad50(s/s) mice , and epistasis studies order Rad50 upstream of Mre11 .\n\nThese studies suggest unrepaired DNA DSBs as causative in target reordering in intestinal SCCs .\n\nAs such , we provide an in vivo model of DNA damage repair that is standardized , can be exploited to understand allele-specific regulation in intact tissue , and is pharmacologically tractable .", "output": "Genomic instability and mutation" }, { "input": "CEACAM1 , CEA/CEACAM5 , and CEACAM6 are cell adhesion molecules ( CAMs ) of the carcinoembryonic antigen ( CEA ) family that have been shown to be deregulated in lung cancer and in up to 50% of all human cancers .\n\nHowever , little is known about the functional impact of these molecules on undifferentiated cell growth and tumor progression .\n\nHere we demonstrate that cell surface expression of CEACAM1 on confluent A549 human lung adenocarcinoma cells plays a critical role in differentiated , contact-inhibited cell growth .\n\nInterestingly , CEACAM1-L , but not CEACAM1-S , negatively regulates proliferation via its ITIM domain , while in proliferating cells no CEACAM expression is detectable .\n\nFurthermore , we show for the first time that CEACAM6 acts as an inducer of cellular proliferation in A549 cells , likely by interfering with the contact-inhibiting signal triggered by CEACAM1-4L , leading to undifferentiated anchorage-independent cell growth .\n\nWe also found that A549 cells expressed significant amounts of non-membrane anchored variants of CEACAM5 and CEACAM6 , representing a putative source for the increased CEACAM5/6 serum levels frequently found in lung cancer patients .\n\nTaken together , our data suggest that post-confluent contact inhibition is established and maintained by CEACAM1-4L , but disturbances of CEACAM1 signalling by CEACAM1-4S and other CEACAMs lead to undifferentiated cell growth and malignant transformation .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND Folate ( vitamin B9 ) is essential for cellular proliferation as it is involved in the biosynthesis of deoxythymidine monophosphate ( dTMP ) and s-adenosylmethionine ( AdoMet ) .\n\nThe link between folate depletion and the genesis and progression of cancers of epithelial origin is of high clinical relevance , but still unclear .\n\nWe recently demonstrated that sensitivity to low folate availability is affected by the rate of polyamine biosynthesis , which is prominent in prostate cells .\n\nWe , therefore , hypothesized that prostate cells might be highly susceptible to genetic , epigenetic and phenotypic changes consequent to folate restriction .\n\nRESULTS We studied the consequences of long-term , mild folate depletion in a model comprised of three syngenic cell lines derived from the transgenic adenoma of the mouse prostate ( TRAMP ) model , recapitulating different stages of prostate cancer ; benign , transformed and metastatic .\n\nHigh-performance liquid chromatography analysis demonstrated that mild folate depletion ( 100 nM ) sufficed to induce imbalance in both the nucleotide and AdoMet pools in all prostate cell lines .\n\nRandom oligonucleotide-primed synthesis ( ROPS ) revealed a significant increase in uracil misincorporation and DNA single strand breaks , while spectral karyotype analysis ( SKY ) identified five novel chromosomal rearrangements in cells grown with mild folate depletion .\n\nUsing global approaches , we identified an increase in CpG island and histone methylation upon folate depletion despite unchanged levels of total 5-methylcytosine , indicating a broad effect of folate depletion on epigenetic regulation .\n\nThese genomic changes coincided with phenotype changes in the prostate cells including increased anchorage-independent growth and reduced sensitivity to folate depletion .\n\nCONCLUSIONS This study demonstrates that prostate cells are highly susceptible to genetic and epigenetic changes consequent to mild folate depletion as compared to cells grown with supraphysiological amounts of folate ( 2 microM ) routinely used in tissue culture .\n\nIn addition , we elucidate for the first time the contribution of these aspects to consequent phenotype changes in epithelial cells .\n\nThese results provide a strong rationale for studying the effects of folate manipulation on the prostate in vivo , where cells might be more sensitive to changes in folate status resulting from folate supplementation or antifolate therapeutic approaches .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE Radiation therapy appears to kill cells mainly by inducing DNA double-strand breaks .\n\nWe investigated whether the DNA repair gene expression status might influence the risk of locoregional recurrence ( LRR ) in breast cancer patients .\n\nMETHODS AND MATERIALS We used a quantitative reverse transcriptase PCR-based approach to measure messenger RNA levels of 20 selected DNA repair genes in tumor samples from 97 breast cancer patients enrolled in a phase III trial ( Centre Ren\u00e9 Huguenin cohort ) .\n\nNormalized mRNA levels were tested for an association with LRR-free survival ( LRR-FS ) and overall survival ( OS ) .\n\nThe findings were validated in comparison with those of an independent cohort ( Netherlands Cancer Institute ( NKI ) cohort ) .\n\nMultivariate analysis encompassing known prognostic factors was used to assess the association between DNA repair gene expression and patient outcome .\n\nRESULTS RAD51 was the only gene associated with LRR in both cohorts .\n\nWith a median follow-up of 126 months in the CRH cohort , the 5-year LRR-FS and OS rates were 100% and 95% in the 61 patients with low RAD51 expression , compared with 70% and 69% in the 36 patients with high RAD51 expression , respectively ( p < 0.001 ) .\n\nRAD51 overexpression was associated with a higher risk of LRR ( hazard ratio [ HR ] , 12.83 ; 95% confidence interval [ CI ] , 3.6-45.6 ) and death ( HR , 4.10 ; 95% CI , 1.7-9.7 ) .\n\nRAD51 overexpression was also significantly associated with shorter LRR-FS and OS in the NKI cohort .\n\nCONCLUSIONS Overexpression of RAD51 , a key component of the homologous DNA repair pathway , is associated with poor breast cancer outcome .\n\nThis finding warrants prospective studies of RAD51 as a prognosticator and therapeutic target .", "output": "Genomic instability and mutation" }, { "input": "ATM(Tel1) and ATR(Rad3) checkpoint kinases phosphorylate the C-terminus of histone H2AX ( H2A in yeasts ) in chromatin flanking DNA damage , establishing a recruitment platform for checkpoint and repair proteins .\n\nPhospho-H2A/X ( gammaH2A/X)-binding proteins at double-strand breaks ( DSBs ) have been characterized , but those required for replication stress responses are unknown .\n\nHere , we present genetic , biochemical , small angle X-ray scattering ( SAXS ) , and X-ray structural studies of the Schizosaccharomyces pombe Brc1 , a 6-BRCT-domain protein that is structurally related to Saccharomyces cerevisiae Rtt107 and mammalian PTIP .\n\nBrc1 binds gammaH2A to form spontaneous and DNA damage-induced nuclear foci .\n\nSpontaneous Brc1 foci colocalize with ribosomal DNA repeats , a region prone to fork pausing and genomic instability , whereas DNA damage-induced Brc1 foci colocalize with DSB response factors. gammaH2A binding is critical for Brc1 function .\n\nThe 1.45 A resolution crystal structure of Brc1-gammaH2A complex shows how variable BRCT insertion loops sculpt tandem-BRCT phosphoprotein-binding pockets to facilitate unique phosphoprotein-interaction specificities , and unveils an acidic DNA-mimicking Brc1 surface .\n\nFrom these results , Brc1 docking to gammaH2A emerges as a critical chromatin-specific response to replication-associated DNA damage .", "output": "Genomic instability and mutation" }, { "input": "Epidemiological studies exploring the connection between hypertension and cancer incidence find a higher cancer mortality in hypertensive patients , particularly elevated in hypertension associated with a stimulation of the renin-angiotensin-aldosterone system .\n\nPrimary aldosteronism , with plasma aldosterone levels between 0.5 and 1 nM ( 18-36 ng/dL ) and local aldosterone levels up to 500 nM ( 18,000 ng/dL ) , is now recognised as a more common cause for hypertension .\n\nWe recently found angiotensin II to be genotoxic due to its induction of oxidative stress .\n\nSince aldosterone in higher concentrations also has oxidative effects , its potential genotoxic action in pig LLC-PK1 cells with properties of proximal tubules was analysed .\n\nDNA damage was evaluated by two test systems : the comet assay , and the micronucleus frequency test .\n\nThe results showed that aldosterone concentrations starting from 10 nM ( 360 ng/dL ) caused a significant increase of DNA damage monitored with the comet assay in LLC-PK1 , while there was no change in cell vitality and proliferation .\n\nThe micronucleus frequency test revealed that 10 nM aldosterone also leads to the formation of micronuclei .\n\nFurthermore , the formation of superoxide radicals in the cells by this aldosterone concentration could be detected with the superoxide-specific stain dihydroethidium .\n\nFurther evidence for oxidative stress-induced DNA damage was its reversibility by the antioxidants tempol and catalase .\n\nAddition of the steroidal mineralocorticoid receptor antagonist spironolactone or the novel selective nonsteroidal antagonist ( R)-BR-4628 reduced the DNA damage and the amount of superoxide radicals indicating a receptor-dependent process .", "output": "Genomic instability and mutation" }, { "input": "Mutations in the gene encoding cytosolic Cu,Zn-superoxide dismutase ( SOD1 ) have been linked to familial amyotrophic lateral sclerosis ( FALS ) .\n\nHowever the molecular mechanisms of motor neuron death are multi-factorial and remain unclear .\n\nHere we examined DNA damage , p53 activity and apoptosis in SH-SY5Y human neuroblastoma cells transfected to achieve low-level expression of either wild-type or mutant Gly(93)-->Ala ( G93A ) SOD1 , typical of FALS .\n\nDNA damage was investigated by evaluating the levels of 8-oxo-7,8-dihydro-2'-deoxyguanosine ( 8-oxodGuo ) and DNA strand breaks .\n\nSignificantly higher levels of DNA damage , increased p53 activity , and a greater percentage of apoptotic cells were observed in SH-SY5Y cells transfected with G93A SOD1 when compared to cells overexpressing wild-type SOD1 and untransfected cells .\n\nWestern blot , FACS , and confocal microscopy analysis demonstrated that G93A SOD1 is present in the nucleus in association with DNA .\n\nNuclear G93A SOD1 has identical superoxide dismutase activity but displays increased peroxidase activity when compared to wild-type SOD1 .\n\nThese results indicate that the G93A mutant SOD1 association with DNA might induce DNA damage and trigger the apoptotic response by activating p53 .\n\nThis toxic activity of mutant SOD1 in the nucleus may play an important role in the complex mechanisms associated with motor neuron death observed in ALS pathogenesis .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "During tumorigenesis , cells acquire immortality in association with the development of genomic instability .\n\nHowever , it is still elusive how genomic instability spontaneously generates during the process of tumorigenesis .\n\nHere , we show that precancerous DNA lesions induced by oncogene acceleration , which induce situations identical to the initial stages of cancer development , trigger tetraploidy/aneuploidy generation in association with mitotic aberration .\n\nAlthough oncogene acceleration primarily induces DNA replication stress and the resulting lesions in the S phase , these lesions are carried over into the M phase and cause cytokinesis failure and genomic instability .\n\nUnlike directly induced DNA double-strand breaks , DNA replication stress-associated lesions are cryptogenic and pass through cell-cycle checkpoints due to limited and ineffective activation of checkpoint factors .\n\nFurthermore , since damaged M-phase cells still progress in mitotic steps , these cells result in chromosomal mis-segregation , cytokinesis failure and the resulting tetraploidy generation .\n\nThus , our results reveal a process of genomic instability generation triggered by precancerous DNA replication stress .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "The purpose of this study was to combine a three-dimensional NMR-compatible bioreactor with hyperpolarized ( 13)C NMR spectroscopy in order to probe cellular metabolism in real time .\n\nJM1 ( immortalized rat hepatoma ) cells were cultured in a three-dimensional NMR-compatible fluidized bioreactor .\n\n( 31)P spectra were acquired before and after each injection of hyperpolarized [ 1-(13)C ] pyruvate and subsequent ( 13)C spectroscopy at 11.7 T .\n\n( 1)H and two-dimensional ( 1)H-(1)H-total correlation spectroscopy spectra were acquired from extracts of cells grown in uniformly labeled ( 13)C-glucose , on a 16.4 T , to determine ( 13)C fractional enrichment and distribution of ( 13)C label .\n\nJM1 cells were found to have a high rate of aerobic glycolysis in both two-dimensional culture and in the bioreactor , with 85% of the ( 13)C label from uniformly labeled ( 13)C-glucose being present as either lactate or alanine after 23 h .\n\nFlux measurements of pyruvate through lactate dehydrogenase and alanine aminotransferase in the bioreactor system were 12.18 +/- 0.49 nmols/sec/10(8) cells and 2.39 +/- 0.30 nmols/sec/10(8) cells , respectively , were reproducible in the same bioreactor , and were not significantly different over the course of 2 days .\n\nAlthough this preliminary study involved immortalized cells , this combination of technologies can be extended to the real-time metabolic exploration of primary benign and cancerous cells and tissues prior to and after therapy .", "output": "Cellular energetics" }, { "input": "Multivitamin preparation ( MVP ) is part of total parenteral nutrition given to premature infants .\n\nPhotoactivated MVP carries an important load in peroxides , but their cellular effects have not yet been determined .\n\nWe hypothesized that these peroxides may elicit a DNA-damage response .\n\nWe found that photoactivation of MVP and the resulting peroxide production were time-dependent and required the simultaneous presence of ascorbic acid and riboflavin .\n\nCells treated with photoactivated MVP showed strongly stimulated poly(ADP-ribosyl)ation , an early DNA-damage response in mammals .\n\nPoly(ADP-ribosyl)ation stimulation was dependent on the presence of ascorbic acid and riboflavin in the photoactivated MVP .\n\nIt did not occur in the presence of a specific PARP inhibitor nor in mouse fibroblasts deficient in PARP-1 .\n\nPhotoactivated MVP was able to induce single- and double-strand breaks in DNA , with a predominance of single-stand breaks .\n\nThe presence of double-strand breaks was further confirmed using a 53PB1 focus analysis .\n\nFinally , photoactivated MVP was shown to be toxic to human cells and induced caspase-independent cell death .\n\nThese results suggest that photoactivated MVP carries an important toxic load able to damage DNA and induce cell death .\n\nThis study also emphasizes the importance of protecting MVP solution from light before use in preterm infants .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "It is thought that high linear energy transfer ( LET ) radiation induces more complex DNA damage than low-LET particles , specifically clustered DNA damage that causes cells to repair DNA double strand breaks ( DSB ) more slowly and leads to severe biological consequences .\n\nThe present study aimed to investigate the role of exogenously added glutathione ( GSH ) on ( 12)C-beam ( 287keV/mum ) and ( 7)Li-beam ( 60keV/mum ) induced chromosome aberration ( CA ) formation , particularly on exchange aberration formation .\n\nIn order to characterize the role of GSH in the joining of DNA DSBs , we induced DNA lesions with bleomycin ( Blem ) in conjunction with either high- or low-LET radiation ( X-rays ) since the chemistry of the free DNA ends created by Blem and X-rays is similar .\n\nCHO cells were exposed to reduced GSH at a concentration of 2mM for 3h before radiation .\n\nTreatment with Blem ( 20mug/ml ) was carried out for 2h before the cells were exposed to radiation .\n\nOur results show that the frequency of chromosomal aberration increases with increased LET .\n\nHeavy ion exposed cells show a higher frequency of CA over time than do X-irradiated cells .\n\nAn analysis of the first post-irradiation mitosis of exposed CHO cells shows that high-LET radiation induces more breaks than exchange-type aberrations and exogenous GSH has no influence on high-LET radiation-induced DNA damage .\n\nThe DNA lesions induced by low-LET radiation interact relatively strongly with Blem-induced lesions whereas interaction between Blem and high-LET radiations was poor .\n\nThis could be attributed to differences in repair kinetics and qualitative differences in the DNA lesions induced by Blem and high-LET radiation .", "output": "Genomic instability and mutation" }, { "input": "p32/gC1qR/C1QBP/HABP1 is a mitochondrial/cell surface protein overexpressed in certain cancer cells .\n\nHere we show that knocking down p32 expression in human cancer cells strongly shifts their metabolism from oxidative phosphorylation ( OXPHOS ) to glycolysis .\n\nThe p32 knockdown cells exhibited reduced synthesis of the mitochondrial-DNA-encoded OXPHOS polypeptides and were less tumorigenic in vivo .\n\nExpression of exogenous p32 in the knockdown cells restored the wild-type cellular phenotype and tumorigenicity .\n\nIncreased glucose consumption and lactate production , known as the Warburg effect , are almost universal hallmarks of solid tumors and are thought to favor tumor growth .\n\nHowever , here we show that a protein regularly overexpressed in some cancers is capable of promoting OXPHOS .\n\nOur results indicate that high levels of glycolysis , in the absence of adequate OXPHOS , may not be as beneficial for tumor growth as generally thought and suggest that tumor cells use p32 to regulate the balance between OXPHOS and glycolysis .", "output": "Cellular energetics" }, { "input": "Recently , the use of gold nanoparticles as potential tumor selective radiosensitizers has been proposed as a breakthrough in radiotherapy .\n\nExperiments in living cells and in vivo have demonstrated the efficiency of the metal nanoparticles when combined with low energy x-ray radiations ( below conventional 1 MeV Linac radiation ) .\n\nFurther studies on DNA have been performed in order to better understand the fundamental processes of sensitization and to further improve the method .\n\nIn this work , we propose a new strategy based on the combination of platinum nanoparticles with irradiation by fast ions effectively used in hadron therapy .\n\nIt is observed in particular that nanoparticles enhance strongly lethal damage in DNA , with an efficiency factor close to 2 for double strand breaks .\n\nIn order to disentangle the effect of the nano-design architecture , a comparison with the effects of dispersed metal atoms at the same concentration has been performed .\n\nIt is thus shown that the sensitization in nanoparticles is enhanced due to auto-amplified electronic cascades inside the nanoparticles , which reinforces the energy deposition in the close vicinity of the metal .\n\nFinally , the combination of fast ion radiation ( hadron therapy ) with platinum nanoparticles should strongly improve cancer therapy protocols .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE This study aims to investigate the role of the aberrant expression of Transkelolase-like 1 ( TKTL1 ) in head and neck squamous cell carcinoma ( HNSCC ) tumorigenesis and to characterize TKTL1 contribution to HNSCC tumorigenesis through aerobic glycolysis and HIF1alpha stabilization .\n\nEXPERIMENTAL DESIGN TKTL1 promoter hypomethylation and mRNA/protein aberrant expression were studied in human HNSCC tumor samples and normal mucosas .\n\nOncogenic functions of TKTL1 were examined in HNSCC cell line panels and tumor xenograft models with TKTL1 expression construct .\n\nThe metabolite levels of fructose-6-phosphate , glyceraldehydes-3-phosphate , pyruvate , lactate , and the levels of HIF1alpha protein and its downsteam glycolytic targets were compared between the TKTL1-expressing and vehicle-expressing HNSCC cells .\n\nMeanwhile , the effects of HIF1alpha/glycolytic inhibitors were evaluated on the TKTL1 transfectants .\n\nRESULTS TKTL1 exhibits high frequency of promoter hypomethylation in HNSCC tumors compared with the normal mucosas , correlating with its overexpression in HNSCC .\n\nOverexpression of TKTL1 in HNSCC cells promoted cellular proliferation and enhanced tumor growth in vitro and in vivo .\n\nOverexpression of TKTL1 increased the production of fructose-6-phosphate and glyceraldehyde-3-phosphate , in turn elevating the production of pyruvate and lactate , resulting in the normoxic stabilization of the malignancy-promoting transcription factor HIF1alpha and the upregulation of downstream glycolytic enzymes .\n\nNotably , the reduction of TKTL1 expression decreased HIF1alpha accumulation and inhibition with HIF1alpha and/or the glycolysis inhibitor could abrogate the growth effects mediated by TKTL1 overexpression .\n\nCONCLUSION TKTL1 is a novel candidate oncogene that is epigenetically activated by aberrant hypomethlation and contributes to a malignant phenotype through altered glycolytic metabolism and HIF1alpha accumulation .", "output": "Cellular energetics" }, { "input": "In eukaryotic cells , multiple DNA repair mechanisms respond to a wide variety of DNA lesions .\n\nHomologous recombination-dependent repair provides a pathway for dealing with DNA double-strand breaks and replication fork demise .\n\nA key step in this process is the resolution of recombination intermediates such as Holliday junctions ( HJs ) .\n\nRecently , nucleases from yeast ( Yen1 ) and human cells ( GEN1 ) were identified that can resolve HJ intermediates , in a manner analogous to the E. coli HJ resolvase RuvC .\n\nHere , we have analyzed the role of Yen1 in DNA repair in S. cerevisiae , and show that while yen1Delta mutants are repair-proficient , yen1Delta mus81Delta double mutants are exquisitely sensitive to a variety of DNA-damaging agents that disturb replication fork progression .\n\nThis phenotype is dependent upon RAD52 , indicating that toxic recombination intermediates accumulate in the absence of Yen1 and Mus81 .\n\nAfter MMS treatment , yen1Delta mus81Delta double mutants arrest with a G2 DNA content and unsegregated chromosomes .\n\nThese findings indicate that Yen1 can act upon recombination/repair intermediates that arise in MUS81-defective cells following replication fork damage .", "output": "Genomic instability and mutation" }, { "input": "BRIT1 protein ( also known as MCPH1 ) contains 3 BRCT domains which are conserved in BRCA1 , BRCA2 , and other important molecules involved in DNA damage signaling , DNA repair , and tumor suppression .\n\nBRIT1 mutations or aberrant expression are found in primary microcephaly patients as well as in cancer patients .\n\nRecent in vitro studies suggest that BRIT1/MCPH1 functions as a novel key regulator in the DNA damage response pathways .\n\nTo investigate its physiological role and dissect the underlying mechanisms , we generated BRIT1(-/-) mice and identified its essential roles in mitotic and meiotic recombination DNA repair and in maintaining genomic stability .\n\nBoth BRIT1(-/-) mice and mouse embryonic fibroblasts ( MEFs ) were hypersensitive to gamma-irradiation .\n\nBRIT1(-/-) MEFs and T lymphocytes exhibited severe chromatid breaks and reduced RAD51 foci formation after irradiation .\n\nNotably , BRIT1(-/-) mice were infertile and meiotic homologous recombination was impaired .\n\nBRIT1-deficient spermatocytes exhibited a failure of chromosomal synapsis , and meiosis was arrested at late zygotene of prophase I accompanied by apoptosis .\n\nIn mutant spermatocytes , DNA double-strand breaks ( DSBs ) were formed , but localization of RAD51 or BRCA2 to meiotic chromosomes was severely impaired .\n\nIn addition , we found that BRIT1 could bind to RAD51/BRCA2 complexes and that , in the absence of BRIT1 , recruitment of RAD51 and BRCA2 to chromatin was reduced while their protein levels were not altered , indicating that BRIT1 is involved in mediating recruitment of RAD51/BRCA2 to the damage site .\n\nCollectively , our BRIT1-null mouse model demonstrates that BRIT1 is essential for maintaining genomic stability in vivo to protect the hosts from both programmed and irradiation-induced DNA damages , and its depletion causes a failure in both mitotic and meiotic recombination DNA repair via impairing RAD51/BRCA2's function and as a result leads to infertility and genomic instability in mice .", "output": "Genomic instability and mutation" }, { "input": "Topoisomerase inhibitors such as camptothecin and etoposide are used as anti-cancer drugs and induce double-strand breaks ( DSBs ) in genomic DNA in cycling cells .\n\nThese DSBs are often covalently bound with polypeptides at the 3 ' and 5 ' ends .\n\nSuch modifications must be eliminated before DSB repair can take place , but it remains elusive which nucleases are involved in this process .\n\nPrevious studies show that CtIP plays a critical role in the generation of 3 ' single-strand overhang at \" clean \" DSBs , thus initiating homologous recombination ( HR)-dependent DSB repair .\n\nTo analyze the function of CtIP in detail , we conditionally disrupted the CtIP gene in the chicken DT40 cell line .\n\nWe found that CtIP is essential for cellular proliferation as well as for the formation of 3 ' single-strand overhang , similar to what is observed in DT40 cells deficient in the Mre11/Rad50/Nbs1 complex .\n\nWe also generated DT40 cell line harboring CtIP with an alanine substitution at residue Ser332 , which is required for interaction with BRCA1 .\n\nAlthough the resulting CtIP(S332A/-/-) cells exhibited accumulation of RPA and Rad51 upon DNA damage , and were proficient in HR , they showed a marked hypersensitivity to camptothecin and etoposide in comparison with CtIP(+/-/-) cells .\n\nFinally , CtIP(S332A/-/-)BRCA1(-/-) and CtIP(+/-/-)BRCA1(-/-) showed similar sensitivities to these reagents .\n\nTaken together , our data indicate that , in addition to its function in HR , CtIP plays a role in cellular tolerance to topoisomerase inhibitors .\n\nWe propose that the BRCA1-CtIP complex plays a role in the nuclease-mediated elimination of oligonucleotides covalently bound to polypeptides from DSBs , thereby facilitating subsequent DSB repair .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE Monoclonal anti-CD33 antibodies conjugated with toxic calicheamicin derivative ( gemtuzumab ozogamicin , GO ) are a novel therapy option for acute myeloid leukaemia ( AML ) .\n\nKey prognostic factors for patients with AML are high CD33 expression on the leukaemic cells and the ability to overcome mechanisms of resistance to cytotoxic chemotherapies , including drug efflux or other mechanisms decreasing apoptosis .\n\nAlpha particle-emitting radionuclides overwhelm such anti-apoptotic mechanisms by producing numerous DNA double-stranded breaks ( DSBs ) accompanied by decreased DNA repair .\n\nMETHODS We labelled anti-CD33 antibodies with the alpha-emitter ( 211)At and compared survival of leukaemic HL-60 and K-562 cells treated with the ( 211)At-labelled antibodies , GO or unlabelled antibodies as controls .\n\nWe also measured caspase-3/7 activity , DNA fragmentation and necrosis in HL-60 cells after treatment with the different antibodies or with free ( 211)At .\n\nRESULTS The mean labelling ratio of ( 211)At-labelled antibodies was 1:1,090 +/- 364 ( range : 1:738-1:1,722 ) in comparison to 2-3:1 for GO .\n\nTumour cell binding of ( 211)At-anti-CD33 was high in the presence of abundant CD33 expression and could be specifically blocked by unlabelled anti-CD33 .\n\n( 211)At-anti-CD33 decreased survival significantly more than did GO at comparable dilution ( 1:1,000 ) .\n\nNo significant differences in induction of apoptosis or necrosis or DNA DSB or in decreased survival were observed after ( 211)At-anti-CD33 ( 1:1,090 ) versus GO ( 1:1 ) treatment .\n\nCONCLUSION Our results suggest that ( 211)At is a promising , highly cytotoxic radioimmunotherapy in CD33-positive leukaemia and kills tumour cells more efficiently than does calicheamicin-conjugated antibody .\n\nLabelling techniques leading to higher chemical yield and specific activities must be developed to increase ( 211)At-anti-CD33 therapeutic effects .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "PURPOSE Anthracyclines have been widely used as antitumor agents , playing a crucial role in the successful treatment of many types of cancer , despite some side effects related to cardiotoxicity .\n\nNew anthracyclines have been designed and tested , but the first ones discovered , doxorubicin and daunorubicin , continue to be the drugs of choice .\n\nDespite their extensive use in chemotherapy , little is known about the DNA repair mechanisms involved in the removal of lesions caused by anthracyclines .\n\nThe anthracycline cosmomycin D is the main product isolated from Streptomyces olindensis , characterized by a peculiar pattern of glycosylation with two trisaccharide rings attached to the A ring of the tetrahydrotetracene .\n\nMETHODS We assessed the induction of apoptosis ( Sub-G1 ) by cosmomycin D in nucleotide excision repair-deficient fibroblasts ( XP-A and XP-C ) as well as the levels of DNA damage ( alkaline comet assay ) .\n\nRESULTS Treatment of XP-A and XP-C cells with cosmomycin D resulted in apoptosis in a time-dependent manner , with highest apoptosis levels observed 96 h after treatment .\n\nThe effects of cosmomycin D were equivalent to those obtained with doxorubicin .\n\nThe broad caspase inhibitor Z-VAD-FMK strongly inhibited apoptosis in these cells , and DNA damage induced by cosmomycin D was confirmed by alkaline comet assay .\n\nCONCLUSIONS Cosmomycin D induced time-dependent apoptosis in nucleotide excision repair-deficient fibroblasts .\n\nDespite similar apoptosis levels , cosmomycin D caused considerably lower levels of DNA damage compared to doxorubicin .\n\nThis may be related to differences in structure between cosmomycin D and doxorubicin .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "We have developed fluorescent probes for the detection of strand scission in the excision repair of oxidatively damaged bases .\n\nThey were hairpin-shaped oligonucleotides , each containing an isomer of thymine glycol or 5,6-dihydrothymine as a damaged base in the center , with a fluorophore and a quencher at the 5'- and 3'-ends , respectively .\n\nFluorescence was detected when the phosphodiester linkage at the damage site was cleaved by the enzyme , because the short fragment bearing the fluorophore could not remain in a duplex form hybridized to the rest of the molecule at the incubation temperature .\n\nThe substrate specificities of Escherichia coli endonuclease III and its human homolog , NTH1 , determined by using these probes agreed with those determined previously by gel electrophoresis using ( 32)P-labeled substrates .\n\nKinetic parameters have also been determined by this method .\n\nSince different fluorophores were attached to the oligonucleotides containing each lesion , reactions with two types of substrates were analyzed separately in a single tube , by changing the excitation and detection wavelengths .\n\nThese probes were degraded during an incubation with a cell extract .\n\nTherefore , phosphorothioate linkages were incorporated to protect the probes from nonspecific nucleases , and the base excision repair activity was successfully detected in HeLa cells .", "output": "Genomic instability and mutation" }, { "input": "Cancer cells constantly adapt to oxidative phosphorylation ( OXPHOS ) suppression resulting from hypoxia or mitochondria defects .\n\nUnder the OXPHOS suppression , AMP-activated protein kinase ( AMPK ) regulates global metabolism adjustments , but its activation has been found to be transient .\n\nWhether cells can maintain cellular ATP homeostasis and survive beyond the transient AMPK activation is not known .\n\nHere , we study the bioenergetic adaptation to the OXPHOS inhibitor oligomycin in a group of cancer cells .\n\nWe found that oligomycin at 100 ng/ml completely inhibits OXPHOS activity in 1 h and induces various levels of glycolysis gains by 6 h , from which we calculate the bioenergetic organizations of cancer cells .\n\nIn glycolysis-dominant cells , oligomycin does not induce much energy stress as measured by glycolysis acceleration , ATP imbalance , AMPK activation , AMPK substrate acetyl-CoA carboxylase phosphorylation at Ser(79) , and cell growth inhibition .\n\nIn OXPHOS-dependent LKB1 wild type cells , oligomycin induces 5-8% ATP drops and transient AMPK activation during the initial 1-2 h .\n\nAfter AMPK activation is completed , oligomycin-induced increase of acetyl-CoA carboxylase phosphorylation at Ser(79) is still detected , and cellular ATP is back at preoligomycin treatment levels by sustained elevation of glycolysis .\n\nCell growth , however , is inhibited without an increase in cell death and alteration in cell cycle distribution .\n\nIn OXPHOS-dependent LKB1-null cells , no AMPK activation by oligomycin is detected , yet cells still show a similar adaptation .\n\nWe also demonstrate that the adaptation to oligomycin does not invoke activation of hypoxia-induced factor .\n\nOur data suggest that cancer cells may grow and survive persistent OXPHOS suppression through an as yet unidentified regulatory mechanism .", "output": "Cellular energetics" }, { "input": "It is commonly believed that neurons remain in G(0) phase of the cell cycle indefinitely .\n\nCell-cycle re-entry , however , is known to contribute to neuronal apoptosis .\n\nMoreover , recent evidence demonstrates the expression of cell-cycle proteins in differentiated neurons under physiological conditions .\n\nThe functional roles of such expression remain unclear .\n\nSince DNA repair is generally attenuated by differentiation in most cell types , the cell-cycle-associated events in postmitotic cells may reflect the need to re-enter the cell cycle to activate DNA repair .\n\nWe show that cyclin-C-directed , pRb-dependent G(0) exit activates the non-homologous end joining pathway of DNA repair ( NHEJ ) in postmitotic neurons .\n\nUsing RNA interference , we found that abrogation of cyclin-C-mediated exit from G(0) compromised DNA repair but did not initiate apoptosis .\n\nForced G(1) entry combined with prevention of G(1) --> S progression triggered NHEJ activation even in the absence of DNA lesions , but did not induce apoptosis in contrast to unrestricted progression through G(1) --> S. We conclude that G(0) --> G(1) transition is functionally significant for NHEJ repair in postmitotic neurons .\n\nThese findings reveal the importance of cell-cycle activation for controlling both DNA repair and apoptosis in postmitotic neurons , and underline the particular role of G(1) --> S progression in apoptotic signaling , providing new insights into the mechanisms of DNA damage response ( DDR ) in postmitotic neurons .", "output": "Genomic instability and mutation" }, { "input": "Mucoepidermoid carcinoma ( MEC ) , the most common primary salivary malignancy , shows great variability in clinical behaviour , thus demanding investigation to identify of prognostic markers .\n\nSince Warburg's studies , unrestricted cell growth during tumorigenesis has been linked to altered metabolism , implying hypoxic stimulation of glycolysis and diminished contribution of mitochondrial oxidative phosphorylation to cellular ATP supply .\n\nHypothesizing that the study of MEC metabolic status could lead to the discovery of prognostic markers , we investigated by immunohistochemistry the expression of glucose transporter 1 ( Glut-1 ) , mitochondrial antigen and peroxiredoxin I ( Prx I ) in samples of MEC from different histological grades .\n\nOur results showed that mitochondrial antigen and Prx I were expressed in the majority of the MEC cases independent of the histological grade .\n\nIn contrast Glut-1 expression increased significantly as the tumours became more aggressive .\n\nThese results suggested that oxidative phosphorylation may contribute to ATP supply in all stages of MEC progression , and that the relative contribution of glycolysis over mitochondria for cellular ATP supply increases during MEC progression , favouring growth under low oxygen concentration .\n\nIn addition , the observed high Prx I protein levels could provide protection to tumour cells against reactive oxygen species generated as a consequence of mitochondrial function and hypoxia-reoxygenation cycling .\n\nAltogether our findings suggest that upregulation of Glut-1 and Prx I constitute successful adaptive strategies of MEC cells conferring a growth advantage over normal salivary gland cells in the unstable oxygenation tumour environment .", "output": "Cellular energetics" }, { "input": "In response to DNA double strand breaks , the histone variant H2AX at the break site is phosphorylated at serine 139 by DNA damage sensor kinases such as ataxia telangiectasia-mutated , forming gamma-H2AX .\n\nThis phosphorylation event is critical for sustained recruitment of other proteins to repair the break .\n\nAfter repair , restoration of the cell to a prestress state is associated with gamma-H2AX dephosphorylation and dissolution of gamma-H2AX-associated damage foci .\n\nThe phosphatases PP2A and PP4 have previously been shown to dephosphorylate gamma-H2AX .\n\nHere , we demonstrate that the wild-type p53-induced phosphatase 1 ( WIP1 ) also dephosphorylates gamma-H2AX at serine 139 in vitro and in vivo .\n\nOverexpression of WIP1 reduces formation of gamma-H2AX foci in response to ionizing and ultraviolet radiation and blocks recruitment of MDC1 ( mediator of DNA damage checkpoint 1 ) and 53BP1 ( p53 binding protein 1 ) to DNA damage foci .\n\nFinally , these inhibitory effects of WIP1 on gamma-H2AX are accompanied by WIP1 suppression of DNA double strand break repair .\n\nThus , WIP1 has a homeostatic role in reversing the effects of ataxia telangiectasia-mutated phosphorylation of H2AX .", "output": "Genomic instability and mutation" }, { "input": "The anti-apoptotic protein , BAX inhibitor-1 ( BI-1 ) , has a role in cancer/tumor progression .\n\nBI-1-overexpressing HT1080 and B16F10 cells produced higher lung weights and tumor volumes after injection into the tail veins of mice .\n\nTransfection of BI-1 siRNA into cells before injection blocked lung metastasis. in vitro , the overexpression of BI-1 increased cell mobility and invasiveness , with highly increased glucose consumption and cytosolic accumulation of lactate and pyruvate , but decreased mitochondrial O(2) consumption and ATP production .\n\nGlucose metabolism-associated extracellular pH also decreased as cells excreted more H(+) , and sodium hydrogen exchanger ( NHE ) activity increased , probably as a homeostatic mechanism for intracellular pH .\n\nThese alterations activated MMP 2/9 and cell mobility and invasiveness , which were reversed by the NHE inhibitor , 5-(N-ethyl-N-isopropyl) amiloride ( EIPA ) , suggesting a role for NHE in cancer metastasis .\n\nIn both in vitro and in vivo experiments , C-terminal deleted ( CDeltaBI-1 ) cells showed similar results to control cells , suggesting that the C-terminal motif is required for BI-1-associated alterations of glucose metabolism , NHE activation and cancer metastasis .\n\nThese findings strongly suggest that BI-1 reduces extracellular pH and regulates metastasis by altering glucose metabolism and activating NHE , with the C-terminal tail having a pivotal role in these processes .", "output": "Cellular energetics, Activating invasion and metastasis" }, { "input": "We investigated the mechanisms of DNA exit during single-cell gel electrophoresis ( the comet assay ) by measuring the kinetics of the comet tail formation .\n\nIn the neutral comet assay , the rate of DNA exit was found to be dependent on the topological state of DNA , which was influenced by either ethidium bromide or a low radiation dose .\n\nThe results clearly show that the comet tail is formed by extended DNA loops : the loop extension , being reversible when the DNA torsional constraint remains in the loops , is favored when the constraint is relaxed .\n\nThe kinetics of the comet formation in the case of a high radiation dose points out that accumulation of the single-strand breaks causes DNA fragmentation .\n\nIn contrast to the neutral comet assay , the alkaline comet assay is not related to the chromatin loops .\n\nOur results imply that the alkaline treatment induces detachment of the loops from the nuclear matrix , and the comet tail is formed by ssDNA fragments , the ends of which are pulled out from the comet head by electric force .\n\nWe suggest that the kinetic approach can be considered as an important improvement of the comet assay .", "output": "Genomic instability and mutation" }, { "input": "Homologous recombination ( HR ) is essential for repair of meiotic DNA double-strand breaks ( DSBs ) .\n\nAlthough the mechanisms of RAD-51-DNA filament assembly and strand exchange are well characterized , the subsequent steps of HR are less well defined .\n\nHere , we describe a synthetic lethal interaction between the C. elegans helicase helq-1 and RAD-51 paralog rfs-1 , which results in a block to meiotic DSB repair after strand invasion .\n\nWhereas RAD-51-ssDNA filaments assemble at meiotic DSBs with normal kinetics in helq-1 , rfs-1 double mutants , persistence of RAD-51 foci and genetic interactions with rtel-1 suggest a failure to disassemble RAD-51 from strand invasion intermediates .\n\nIndeed , purified HELQ-1 and RFS-1 independently bind to and promote the disassembly of RAD-51 from double-stranded , but not single-stranded , DNA filaments via distinct mechanisms in vitro .\n\nThese results indicate that two compensating activities are required to promote postsynaptic RAD-51 filament disassembly , which are collectively essential for completion of meiotic DSB repair .", "output": "Genomic instability and mutation" }, { "input": "Diphenyl ditelluride ( DPDT ) is a potential prototype for the development of novel biologically active molecules .\n\nThus , it is important to evaluate the toxic effects of this compound .\n\nIn the present study , we evaluated the cytotoxic , genotoxic and mutagenic properties of DPDT in Chinese hamster fibroblast ( V79 ) cells , in strains of the yeast Saccharomyces cerevisiae both proficient and deficient in several DNA repair pathways and in Salmonella typhimurium .\n\nDPDT induced frameshift mutations in both S.typhimurium and a haploid wild-type strain of S.cerevisiae .\n\nMutants of S.cerevisiae defective in base excision repair and recombinational repair were more sensitive to DPDT .\n\nThe results of a lactate dehydrogenase leakage assay suggest that DPDT is cytotoxic to V79 cells .\n\nAt cytotoxic concentrations , this compound increased thiobarbituric reactive species levels and decreased the glutathione:GSSH ratio in yeast and V79 cells .\n\nDPDT generated single- and double-strand DNA breaks in V79 cells , both with and without metabolic activation , as revealed by alkaline and neutral comet assays .\n\nMoreover , an induction of oxidative DNA base damage was indicated by a modified comet assay using formamidopyrimidine DNA glycosylase and endonuclease III .\n\nTreatment with DPDT also induced micronucleus formation in V79 cells .\n\nPre-incubation with N-acetylcysteine reduced DPDT's oxidative , genotoxic and mutagenic effects in yeast and V79 cells .\n\nOur results suggest that the toxic and mutagenic properties of DPDT may stem from its ability to disturb the redox balance of the cell , which leads to oxidative stress and the induction of DNA damage .", "output": "Genomic instability and mutation" }, { "input": "DNA damage checkpoints arrest cell cycle progression to facilitate DNA repair .\n\nThe ability to survive genotoxic insults depends not only on the initiation of cell cycle checkpoints but also on checkpoint maintenance .\n\nWhile activation of DNA damage checkpoints has been studied extensively , molecular mechanisms involved in sustaining and ultimately inactivating cell cycle checkpoints are largely unknown .\n\nHere , we explored feedback mechanisms that control the maintenance and termination of checkpoint function by computationally identifying an evolutionary conserved mitotic phosphorylation network within the DNA damage response .\n\nWe demonstrate that the non-enzymatic checkpoint adaptor protein 53BP1 is an in vivo target of the cell cycle kinases Cyclin-dependent kinase-1 and Polo-like kinase-1 ( Plk1 ) .\n\nWe show that Plk1 binds 53BP1 during mitosis and that this interaction is required for proper inactivation of the DNA damage checkpoint. 53BP1 mutants that are unable to bind Plk1 fail to restart the cell cycle after ionizing radiation-mediated cell cycle arrest .\n\nImportantly , we show that Plk1 also phosphorylates the 53BP1-binding checkpoint kinase Chk2 to inactivate its FHA domain and inhibit its kinase activity in mammalian cells .\n\nThus , a mitotic kinase-mediated negative feedback loop regulates the ATM-Chk2 branch of the DNA damage signaling network by phosphorylating conserved sites in 53BP1 and Chk2 to inactivate checkpoint signaling and control checkpoint duration .", "output": "Evading growth suppressors" }, { "input": "Class switch recombination ( CSR ) in B lymphocytes is initiated by introduction of multiple DNA double-strand breaks ( DSBs ) into switch ( S ) regions that flank immunoglobulin heavy chain ( IgH ) constant region exons .\n\nCSR is completed by joining a DSB in the donor S mu to a DSB in a downstream acceptor S region ( e.g. , S gamma1 ) by end-joining .\n\nIn normal cells , many CSR junctions are mediated by classical nonhomologous end-joining ( C-NHEJ ) , which employs the Ku70/80 complex for DSB recognition and XRCC4/DNA ligase 4 for ligation .\n\nAlternative end-joining ( A-EJ ) mediates CSR , at reduced levels , in the absence of C-NHEJ , even in combined absence of Ku70 and ligase 4 , demonstrating an A-EJ pathway totally distinct from C-NHEJ .\n\nMultiple DSBs are introduced into S mu during CSR , with some being rejoined or joined to each other to generate internal switch deletions ( ISDs ) .\n\nIn addition , S-region DSBs can be joined to other chromosomes to generate translocations , the level of which is increased by absence of a single C-NHEJ component ( e.g. , XRCC4 ) .\n\nWe asked whether ISD and S-region translocations occur in the complete absence of C-NHEJ ( e.g. , in Ku70/ligase 4 double-deficient B cells ) .\n\nWe found , unexpectedly , that B-cell activation for CSR generates substantial ISD in both S mu and S gamma1 and that ISD in both is greatly increased by the absence of C-NHEJ .\n\nIgH chromosomal translocations to the c-myc oncogene also are augmented in the combined absence of Ku70 and ligase 4 .\n\nWe discuss the implications of these findings for A-EJ in normal and abnormal DSB repair .", "output": "Genomic instability and mutation" }, { "input": "Cancer cells preferentially metabolize glucose by aerobic glycolysis , characterized by increased lactate production .\n\nThis distinctive metabolism involves expression of the embryonic M2 isozyme of pyruvate kinase , in contrast to the M1 isozyme normally expressed in differentiated cells , and it confers a proliferative advantage to tumor cells .\n\nThe M1 and M2 pyruvate-kinase isozymes are expressed from a single gene through alternative splicing of a pair of mutually exclusive exons .\n\nWe measured the expression of M1 and M2 mRNA and protein isoforms in mouse tissues , tumor cell lines , and during terminal differentiation of muscle cells , and show that alternative splicing regulation is sufficient to account for the levels of expressed protein isoforms .\n\nWe further show that the M1-specific exon is actively repressed in cancer-cell lines--although some M1 mRNA is expressed in cell lines derived from brain tumors--and demonstrate that the related splicing repressors hnRNP A1 and A2 , as well as the polypyrimidine-tract-binding protein PTB , contribute to this control .\n\nDownregulation of these splicing repressors in cancer-cell lines using shRNAs rescues M1 isoform expression and decreases the extent of lactate production .\n\nThese findings extend the links between alternative splicing and cancer , and begin to define some of the factors responsible for the switch to aerobic glycolysis .", "output": "Cellular energetics" }, { "input": "As the result of genetic alterations and tumor hypoxia , many cancer cells avidly take up glucose and generate lactate through lactate dehydrogenase A ( LDHA ) , which is encoded by a target gene of c-Myc and hypoxia-inducible factor ( HIF-1 ) .\n\nPrevious studies with reduction of LDHA expression indicate that LDHA is involved in tumor initiation , but its role in tumor maintenance and progression has not been established .\n\nFurthermore , how reduction of LDHA expression by interference or antisense RNA inhibits tumorigenesis is not well understood .\n\nHere , we report that reduction of LDHA by siRNA or its inhibition by a small-molecule inhibitor ( FX11 [ 3-dihydroxy-6-methyl-7-(phenylmethyl)-4-propylnaphthalene-1-carboxylic acid] ) reduced ATP levels and induced significant oxidative stress and cell death that could be partially reversed by the antioxidant N-acetylcysteine .\n\nFurthermore , we document that FX11 inhibited the progression of sizable human lymphoma and pancreatic cancer xenografts .\n\nWhen used in combination with the NAD(+) synthesis inhibitor FK866 , FX11 induced lymphoma regression .\n\nHence , inhibition of LDHA with FX11 is an achievable and tolerable treatment for LDHA-dependent tumors .\n\nOur studies document a therapeutical approach to the Warburg effect and demonstrate that oxidative stress and metabolic phenotyping of cancers are critical aspects of cancer biology to consider for the therapeutical targeting of cancer energy metabolism .", "output": "Cellular energetics" }, { "input": "The contribution of a type II restriction-modification system ( R-M system ) to genome integrity and cell viability was investigated .\n\nWe established experimental conditions which enabled the achievement of hemimethylated and unmethylated states for the specific bases of the recognition sequences of the host's DNA .\n\nTo achieve this , we constructed the MboII R-M system containing only one ( i.e .\n\nM2.MboII ) out of two functional MboII methyltransferases found in Moraxella bovis .\n\nUsing the incomplete R-M system we were able to perturb the balance between methylation and restriction in an inducible manner .\n\nWe demonstrate that upon the SOS-induced DNA repair in the mitomycin C treated cells , restriction significantly reduces cell viability .\n\nSimilar results for the well-studied wild type EcoRI R-M system , expressed constitutively in Escherichia coli , were obtained .\n\nOur data provide further insights into the benefits and disadvantages of maintaining of a type II R-M system , highlighting its impact on host cell fitness .", "output": "Genomic instability and mutation" }, { "input": "Acidosis commonly observed in solid tumors like pancreatic cancer promotes genetic instability and selection of a more malignant phenotype of cancer cells .\n\nOverexpression or activation of integral membrane proteins mediating H+ efflux may contribute to extracellular acidification .\n\nNeurotensin ( NT ) induces intracellular alkalinization and stimulates interleukin-8 production in pancreatic cancer cells and , as demonstrated here , the stable NT analog Lys(8)-psi-Lys(9)NT(8-13) enhances the amiloride-sensitive , Na+-dependent transmembrane H+ flux by a factor of 2.05+/-0.28 and 2.69+/-0.07 in BxPC-3 and PANC-1 pancreatic cancer cells , respectively , by phosphorylation of the Na+/H+ exchanger 1 ( NHE1 ) .\n\nHuman genome-wide gene expression analysis was performed to detect effects of Lys(8)-psi-Lys(9)NT(8-13) on BxPC-3 cells .\n\nResults indicated upregulation of genes involved in regulation of NHE1 , hypoxic response and glycolysis in response to Lys(8)-psi-Lys(9)NT(8-13) even under normoxic conditions .\n\nTherefore , our findings suggest that growth factors like NT may be implicated in the early progression of pancreatic cancer by localized acidification and induction of an aerobic glycolytic phenotype with higher metastatic potential in small cell aggregates .", "output": "Cellular energetics" }, { "input": "Aerobic glycolysis and mitochondrial dysfunction are common features of aggressive cancer growth .\n\nWe observed promoter methylation and loss of expression in neurofilament heavy polypeptide ( NEFH ) in a significant proportion of primary esophageal squamous cell carcinoma ( ESCC ) samples that were of a high tumor grade and advanced stage .\n\nRNA interference-mediated knockdown of NEFH accelerated ESCC cell growth in culture and increased tumorigenicity in vivo , whereas forced expression of NEFH significantly inhibited cell growth and colony formation .\n\nLoss of NEFH caused up-regulation of pyruvate kinase-M2 type and down-regulation of pyruvate dehydrogenase , via activation of the Akt/beta-catenin pathway , resulting in enhanced aerobic glycolysis and mitochondrial dysfunction .\n\nThe acceleration of glycolysis and mitochondrial dysfunction in NEFH-knockdown cells was suppressed in the absence of beta-catenin expression , and was decreased by the treatment of 2-Deoxyglucose , a glycolytic inhibitor , or API-2 , an Akt inhibitor .\n\nLoss of NEFH activates the Akt/beta-catenin pathway and increases glycolysis and mitochondrial dysfunction .\n\nCancer cells with methylated NEFH can be targeted for destruction with specific inhibitors of deregulated downstream pathways .", "output": "Cellular energetics" }, { "input": "BACKGROUND Taxol is one of the most effective chemotherapeutic agents for the treatment of patients with breast cancer .\n\nDespite impressive clinical responses initially , the majority of patients eventually develop resistance to Taxol .\n\nLactate dehydrogenase-A ( LDH-A ) is one of the predominant isoforms of LDH expressed in breast tissue , which controls the conversion of pyruvate to lactate and plays an important role in glucose metabolism .\n\nIn this study we investigated the role of LDH-A in mediating Taxol resistance in human breast cancer cells .\n\nRESULTS Taxol-resistant subclones , derived from the cancer cell line MDA-MB-435 , sustained continuous growth in high concentrations of Taxol while the Taxol-sensitive cells could not .\n\nThe increased expression and activity of LDH-A were detected in Taxol-resistant cells when compared with their parental cells .\n\nThe downregulation of LDH-A by siRNA significantly increased the sensitivity of Taxol-resistant cells to Taxol .\n\nA higher sensitivity to the specific LDH inhibitor , oxamate , was found in the Taxol-resistant cells .\n\nFurthermore , treating cells with the combination of Taxol and oxamate showed a synergistical inhibitory effect on Taxol-resistant breast cancer cells by promoting apoptosis in these cells .\n\nCONCLUSION LDH-A plays an important role in Taxol resistance and inhibition of LDH-A re-sensitizes Taxol-resistant cells to Taxol .\n\nThis supports that Warburg effect is a property of Taxol resistant cancer cells and may play an important role in the development of Taxol resistance .\n\nTo our knowledge , this is the first report showing that the increased expression of LDH-A plays an important role in Taxol resistance of human breast cancer cells .\n\nThis study provides valuable information for the future development and use of targeted therapies , such as oxamate , for the treatment of patients with Taxol-resistant breast cancer .", "output": "Cellular energetics" }, { "input": "The relationship between DNA damage and repair of peripheral blood leukocytes , liver , kidney and brain cells was investigated in Swiss albino mice ( Mus musculus L. ) after exposure to sevoflurane ( 2.4 vol% for 2 h daily , for 3 days ) .\n\nGenetic damage of mouse cells was investigated by the comet assay and micronucleus test .\n\nTo perform the comet assay , mice were divided into a control group and 4 groups of exposed mice sacrificed on day 3 of the experiment , at 0 , 2 , 6 or 24 h after the last exposure to sevoflurane .\n\nMean tail length ( TL ) , tail moment ( TM ) , and tail intensity ( TI ) values were significantly higher in exposed mice ( all examined organs ) than in the control group .\n\nSignificant DNA damage immediately after exposure to sevoflurane was observed in leukocytes .\n\nDamage induction in the liver , kidney , and brain occurred 6 h later than in leukocytes , as expected according to the toxicokinetics of the drug , where blood is the first compartment to absorb sevoflurane .\n\nHowever , none of the tested tissues revealed signs of repair until 24 h after the exposure .\n\nTo distinguish the unrepaired genome damage in vivo , the micronucleus test was applied .\n\nNumber of micronuclei in reticulocytes showed a statistically significant increase , as compared with the control group at all observed times after the treatment .", "output": "Genomic instability and mutation" }, { "input": "Colorectal cancer ( CRC ) arises as the consequence of progressive changes from normal epithelial cells through polyp to tumor , and thus is an useful model for studying metabolic shift .\n\nIn the present study , we studied the metabolomic profiles using high analyte specific gas chromatography/mass spectrometry ( GC/MS ) and liquid chromatography tandem mass spectrometry ( LC/MS/MS ) to attain a systems-level view of the shift in metabolism in cells progressing along the path to CRC .\n\nColonic tissues including tumor , polyps and adjacent matched normal mucosa from 26 patients with sporadic CRC from freshly isolated resections were used for this study .\n\nThe metabolic profiles were obtained using GC/MS and LC/MS/MS .\n\nOur data suggest there was a distinct profile change of a wide range of metabolites from mucosa to tumor tissues .\n\nVarious amino acids and lipids in the polyps and tumors were elevated , suggesting higher energy needs for increased cellular proliferation .\n\nIn contrast , significant depletion of glucose and inositol in polyps revealed that glycolysis may be critical in early tumorigenesis .\n\nIn addition , the accumulation of hypoxanthine and xanthine , and the decrease of uric acid concentration , suggest that the purine biosynthesis pathway could have been substituted by the salvage pathway in CRC .\n\nFurther , there was a step-wise reduction of deoxycholic acid concentration from mucosa to tumors .\n\nIt appears that to gain a growth advantage , cancer cells may adopt alternate metabolic pathways in tumorigenesis and this flexibility allows them to adapt and thrive in harsh environment .", "output": "Cellular energetics" }, { "input": "Although the etiology of primary brain tumors is largely unknown , prior studies suggest that DNA repair polymorphisms may influence risk of glioma .\n\nAltered DNA repair is also likely to affect the risk of meningioma and acoustic neuroma , but these tumors have not been well studied .\n\nWe estimated the risk of glioma ( n = 362 ) , meningioma ( n = 134 ) , and acoustic neuroma ( n = 69 ) in non-Hispanic whites with respect to 36 single nucleotide polymorphisms from 26 genes involved in DNA repair in a hospital-based , case-control study conducted by the National Cancer Institute .\n\nWe observed significantly increased risk of meningioma with the T variant of GLTSCR1 rs1035938 ( OR(CT/TT) = 3.5 ; 95% confidence interval : 1.8-6.9 ; P(trend) .0006 ) , which persisted after controlling for multiple comparisons ( P = .019 ) .\n\nSignificantly increased meningioma risk was also observed for the minor allele variants of ERCC4 rs1800067 ( P(trend) .01 ) ; MUTYH rs3219466 ( P(trend) .02 ) , and PCNA rs25406 ( P(trend) .03 ) .\n\nThe NBN rs1805794 minor allele variant was associated with decreased meningioma risk ( P(trend) .006 ) .\n\nRisk of acoustic neuroma was increased for the ERCC2 rs1799793 ( P(trend) .03 ) and ERCC5 rs17655 ( P(trend) .05 ) variants and decreased for the PARP1 rs1136410 ( P(trend) .03 ) .\n\nDecreased glioma risk was observed with the XRCC1 rs1799782 variant ( P(trend) .04 ) .\n\nOur results suggest that common DNA repair variants may affect the risk of adult brain tumors , especially meningioma .", "output": "Genomic instability and mutation" }, { "input": "It is crucial for organ homeostasis that epithelia have effective mechanisms to restrict motility and cell proliferation in order to maintain tissue architecture .\n\nOn the other hand , epithelial cells need to rapidly and transiently acquire a more mesenchymal phenotype , with high levels of cell motility and proliferation , in order to repair epithelia upon injury .\n\nCross talk between cell-cell and cell-matrix signaling is crucial for regulating these transitions .\n\nThe Pak1-betaPIX-GIT complex is an effector complex downstream of the small GTPase Rac1 .\n\nWe previously showed that translocation of this complex from cell-matrix to cell-cell adhesion sites was required for the establishment of contact inhibition of proliferation .\n\nIn this study , we provide evidence that this translocation depends on cadherin function .\n\nCadherins do not recruit the complex by direct interaction .\n\nRather , we found that inhibition of the normal function of cadherin or Pak1 leads to defects in focal adhesion turnover and to increased signaling by phosphatidylinositol 3-kinase .\n\nWe propose that cadherins are involved in regulation of contact inhibition by controlling the function of the Pak1-betaPIX-GIT complex at focal contacts .", "output": "Evading growth suppressors" }, { "input": "The neurofibromatosis type 2 ( NF2 ) tumor suppressor gene encodes merlin , a membrane/cytoskeleton protein necessary for the maintenance of contact inhibition of growth in cells .\n\nBi-allelic inactivation of NF2 is known to cause multiple cancers in both humans and mice .\n\nHowever , the mechanism through which merlin exerts its tumor-suppressive function remains obscure .\n\nIn this report , we show that NF2 knockout mouse embryonic fibroblasts lost contact inhibition of cell proliferation and contained significantly increased canonical Wnt signaling .\n\nInhibition of Rac1 , the activity of which is inversely regulated by NF2 , through the use of a dominant-negative mutant , small hairpin RNA or a small molecule inhibitor in NF2-deficient cells , was able to suppress elevated Wnt signals as shown by reduced activity of the T-cell factor 4 ( TCF4 ) transcription factor .\n\nDominant-negative TCF4 or Rac1 mutant , as well as a small molecule inhibition of Wnt , were able to curb NF2 deficiency-elicited cell proliferation at the confluent state .\n\nThus , Rac1-mediated canonical Wnt signaling is essential for the loss of contact inhibition in NF2-deficient cells .", "output": "Evading growth suppressors" }, { "input": "The phosphatidylinositol-3-kinase ( PI3K)/Akt oncogenic pathway is critical in glioblastomas .\n\nLoss of PTEN , a negative regulator of the PI3K pathway or activated PI3K/Akt pathway that drive increased proliferation , survival , neovascularization , glycolysis , and invasion is found in 70%-80% of malignant gliomas .\n\nThus , PI3K is an attractive therapeutic target for malignant glioma .\n\nWe report that a new irreversible PI3K inhibitor , PX-866 , shows potent inhibitory effects on the PI3K/Akt signaling pathway in glioblastoma .\n\nPX-866 did not induce any apoptosis in glioma cells ; however , an increase in autophagy was observed .\n\nPX-866 inhibited the invasive and angiogenic capabilities of cultured glioblastoma cells .\n\nIn vivo , PX-866 inhibited subcutaneous tumor growth and increased the median survival time of animals with intracranial tumors .\n\nWe also assessed the potential of proton magnetic resonance spectroscopy ( MRS ) as a noninvasive method to monitor response to PX-866 .\n\nOur findings show that PX-866 treatment causes a drop in the MRS-detectable choline-to-NAA , ratio and identify this partial normalization of the tumor metabolic profile as a biomarker of molecular drug action .\n\nOur studies affirm that the PI3K pathway is a highly specific molecular target for therapies for glioblastoma and other cancers with aberrant PI3K/PTEN expression .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Overexpression of the adverse prognostic marker ERBB2 occurs in 30% of breast cancers ; however , therapies targeting this gene have not proved to be as effective as was initially hoped .\n\nTranscriptional profiling meta-analyses have shown that there are approximately 150 genes co-overexpressed with ERBB2 , suggesting that these genes may represent alternative factors influencing ERBB2-positive tumors .\n\nHere we describe an RNA interference-based analysis of these genes that identifies transcriptional regulators of fat synthesis and storage as being critical for the survival of these cells .\n\nThese transcription factors , nuclear receptor subfamily 1 , group D , member 1 ( NR1D1 ) and peroxisome proliferator activated receptor gamma binding protein ( PBP ) , both reside on ERBB2-containing 17q12-21 amplicons and are part of the ERBB2 expression signature .\n\nWe show that NR1D1 and PBP act through a common pathway in upregulating several genes in the de novo fatty acid synthesis network , which is highly active in ERBB2-positive breast cancer cells .\n\nMalate dehydrogenase 1 and malic enzyme 1 , enzymes that link glycolysis and fatty acid synthesis , are also regulated by NR1D1 .\n\nThe resulting high-level fat production from increased expression of these genes likely contributes to an abnormal cellular energy metabolism based on aerobic glycolysis .\n\nTogether , these results show that the cells of this aggressive form of breast cancer are genetically preprogrammed to depend on NR1D1 and PBP for the energy production necessary for survival .", "output": "Cellular energetics" }, { "input": "Ataxia-telangiectasia mutated ( ATM ) is a cellular damage sensor that coordinates the cell cycle with damage-response checkpoints and DNA repair to preserve genomic integrity .\n\nHowever , ATM also has been implicated in metabolic regulation , and ATM deficiency is associated with elevated reactive oxygen species ( ROS ) .\n\nROS has a central role in many physiological and pathophysiological processes including inflammation and chronic diseases such as atherosclerosis and cancer , underscoring the importance of cellular pathways involved in redox homeostasis .\n\nWe have identified a cytoplasmic function for ATM that participates in the cellular damage response to ROS .\n\nWe show that in response to elevated ROS , ATM activates the TSC2 tumor suppressor via the LKB1/AMPK metabolic pathway in the cytoplasm to repress mTORC1 and induce autophagy .\n\nImportantly , elevated ROS and dysregulation of mTORC1 in ATM-deficient cells is inhibited by rapamycin , which also rescues lymphomagenesis in Atm-deficient mice .\n\nOur results identify a cytoplasmic pathway for ROS-induced ATM activation of TSC2 to regulate mTORC1 signaling and autophagy , identifying an integration node for the cellular damage response with key pathways involved in metabolism , protein synthesis , and cell survival .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "BACKGROUND & OBJECTIVE Hereditary non-polyposis colorectal cancer ( HNPCC or Lynch syndrome ) , is a genetically heterogeneous disorder that is believed to account for 2-10 per cent of all the colorectal cancer cases .\n\nThe disease follows autosomal dominant inheritance pattern with high penetrance ( 85% ) and younger age of onset when compared to patients with sporadic tumours .\n\nHNPCC is associated with germ-line mutations in the DNA mismatch repair ( MMR ) genes namely MLH1 , MSH2 , MSH6 , and PMS2 .\n\nThe present study was aimed at analyzing mismatch repair gene(s) in an extended Indian family satisfying the Amsterdam criteria , and extending the analysis to general population to estimate frequency of the mutations/polymorphisms observed .\n\nMETHODS A total 12 members of the HNPCC family were studied for genetic investigation .\n\nEthnically matched 250 normal individuals were also included as controls to study the observed mutations/polymorphisms at population level .\n\nRESULTS The analysis resulted in identification of a 1975C>T mutation in exon 17 , resulting in substitution of arginine residue with stop codon at codon 659. 655A>G substitution was also observed , resulting in replacement of isoleucine with valine at codon 219 .\n\nSimilar analysis on 250 ethnically matched control subjects revealed complete absence of R659X mutation , while I219V variant was found in 9.8 per cent of the controls .\n\nINTERPRETATION & CONCLUSION R659X mutation correlates with disease phenotype , and 655A>G locus is highly polymorphic .\n\nOur study suggested that R659X substitution was prime cause for the disease phenotype in this family .\n\nI219V substitution is a polymorphism having no association with the disease onset or segregation .\n\nThe family members harbouring this mutation were advised to be under regular medical surveillance .", "output": "Genomic instability and mutation" }, { "input": "Previous studies with selenium and/or vitamin E in prostate carcinogenesis animal models have been negative , but these models may not involve oxidative stress mechanisms .\n\nIn this study , we examined the potential of selenomethionine and alpha-tocopherol to modulate prostate cancer development in the testosterone plus estradiol-treated NBL rat , a model that does involve sex hormone-induced oxidative stress mechanisms and prostatic inflammation .\n\nOne week following the implantation with hormone-filled Silastic implants , rats were fed diets containing l-selenomethionine ( 1.5 or 3.0 mg/kg ) , DL-alpha-tocopherol acetate ( 2,000 or 4,000 mg/kg ) , or a natural ingredient control diet ( NIH-07 ) .\n\nThe development of prostate carcinomas was not affected by dietary treatment with either agent .\n\nFood intake , body weight , and mortality were also not affected .\n\nThe high dose of selenomethionine reduced the severity of epithelial dysplasia in the lateral prostate that was not associated with inflammation , and alpha-tocopherol reduced in a dose-related fashion the incidence of marked inflammation and marked epithelial dysplasia in the lateral prostate , regardless of whether these lesions were associated with inflammation. alpha-Tocopherol significantly increased the incidence of adenocarcinomas of the mammary glands at both dietary concentrations .\n\nCollectively , our findings suggest that selenomethionine and alpha-tocopherol supplementation does not prevent prostate cancer in rats fed diets with nutritionally adequate levels of selenium and vitamin E. Importantly , the results of the current animal studies and those reported previously were fully predictive of the outcome of the Selenium and Vitamin E Cancer Prevention Trial .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND Lung cancer often develops in association with chronic pulmonary inflammatory diseases with an influx of neutrophils .\n\nMore detailed information on inflammatory pathways and the role of neutrophils herein is a prerequisite for understanding the mechanism of inflammation associated cancer .\n\nMETHODS In the present study , we used microarrays in order to obtain a global view of the transcriptional responses of the lung to LPS in mice , which mimics an acute lung inflammation .\n\nTo investigate the influence of neutrophils in this process , we depleted mice from circulating neutrophils by treatment with anti-PMN antibodies prior to LPS exposure .\n\nRESULTS A total of 514 genes was greater than 1.5-fold differentially expressed in the LPS induced lung inflammation model. 394 of the 514 were up regulated genes mostly involved in cell cycle and immune/inflammation related processes , such as cytokine/chemokine activity and signalling .\n\nDown regulated genes represented nonimmune processes , such as development , metabolism and transport .\n\nNotably , the number of genes and pathways that were differentially expressed , was reduced when animals were depleted from circulating neutrophils , confirming the central role of neutrophils in the inflammatory response .\n\nFurthermore , there was a significant correlation between the differentially expressed gene list and the promutagenic DNA lesion M1dG , suggesting that it is the extent of the immune response which drives genetic instability in the inflamed lung .\n\nSeveral genes that were specifically regulated by the presence of activated neutrophils could be identified and these were mostly involved in interferon signalling , oxidative stress response and cell cycle progression .\n\nThe latter possibly refers to a higher rate of cell turnover in the inflamed lung with neutrophils , suggesting that the neutrophil influx is associated with a higher risk for the accumulation and fixation of mutations .\n\nCONCLUSION Gene expression profiling identified specific genes and pathways that are related to neutrophilic inflammation and could be associated to cancer development and indicate an active role of neutrophils in mediating the LPS induced inflammatory response in the mouse lung .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "AIM Colorectal cancer is the third most common form of cancer in the industrial countries .\n\nDue to advances regarding the treatments , primarily development of improved surgical methods and the ability to make the earlier diagnosis , the mortality has remained constant during the past decades even though the incidence in fact has increased .\n\nTo improve chemotherapy and enable personalised treatment , the need of biomarkers is of great significance .\n\nIn this study , we evaluated the gene expression profiles of the colon cancer cell lines treated with SN-38 , the active metabolite of topoisomerase-1 inhibitor irinotecan which leads to cell cycle arrest and apoptosis .\n\nMATERIAL AND METHODS The study included 3 colon cancer cell lines : KM12C , KM12SM and KM12l4a .\n\nThe 3 cell lines were treated with SN-38 , and samples were obtained after 24 and 48 hour treatments .\n\nThe gene expression analyses were performed using oligonucleotide microarrays comprising of approximately 27,000 spots where the untreated controls were compared to the SN-38-treated samples .\n\nRESULTS Unsupervised clustering clearly distinguished the treated cell lines from the untreated .\n\nSupervised analysis identified 3,974 significant genes ( p = 0.05 ) differentiating the treated samples from the untreated , majority of which were down-regulated after treatment .\n\nThe top-ranked down-regulated genes in the treated cell lines included those related to receptor and kinase activity , signal transduction , apoptosis , RNA processing , protein metabolism and transport , cell cycle and transcription .\n\nA smaller number of genes were up-regulated in the cell lines after treatment and included genes involved in apoptosis , transcription , development and differentiation .\n\nCONCLUSIONS These results demonstrate that the expression of the genes involved in cell proliferation and apoptosis as well as RNA , DNA and protein metabolism were affected by SN-38 .\n\nThe impact of certain genes on colorectal cancer development needs to be further evaluated ; however , these results could serve as a basis for further studies in order to find targets for irinotecan treatment .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Hypoxia is widespread in solid tumors as a consequence of poorly structured tumor-derived neovasculature , which is recognized to play a role in the resistance of cancer cells to chemotherapy .\n\nEtoposide ( VP-16 ) , a drug commonly used in chemotherapy , leads to enhanced accumulation of cell populations in G2/M phase and increases levels of apoptosis as a topoisomerase II inhibitor .\n\nWe evaluated the effects of hypoxia on the response of the neuroblastoma cell line CHP126 to VP-16 , in order to delineate the mechanisms responsible for the hypoxia-induced chemoresistance of this clinically conventional anti-cancer agent , with an insight to determining potential indications in neuroblastoma therapy .\n\nIn this study , physiological hypoxia was shown to attenuate G2/M arrest and apoptosis induced in CHP126 cells by VP-16 .\n\nIt suppressed drug-related Cdk1 activity with a less elevation of regulator proteins such as cyclin B1 , Cdk7 and reduced caspase activation and PARP cleavage compared to the efficiency observed in normoxic condition , which were significantly relative with hypoxia-driven inhibition of p53 and p-ERK1/2 activation .\n\nThese results clearly demonstrated that hypoxia had a protective effect against VP-16-induced cytotoxicity , which is likely to provide a further therapeutic knowledge in neuroblastomas .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND Non-alcoholic fatty liver disease ( NAFLD ) is associated with obesity , insulin resistance and hepatic steatosis .\n\nNon-alcoholic steatohepatitis ( NASH ) is a serious consequence of NAFLD where chronic tissue damage and inflammation result in fibrosis which may progress to cirrhosis .\n\nTransforming growth factor beta1 ( TGFbeta1 ) , proinflammatory cytokines and oxidative stress are thought to play crucial roles in the pathogenesis of these conditions .\n\nThe contributions of individual liver cell types to fibrogenesis remain controversial and the influence of selenium status has not been investigated .\n\nMETHODS In this study we have used a cell culture model of fat-loading using oleate-treated human hepatoblastoma ( C3A ) cells to investigate how fat-loading and selenium status might influence the production of collagen in response to TGFbeta1 .\n\nThe secretion of inflammatory cytokines was also investigated , together with the epithelial character of the treated cells .\n\nRESULTS We found that in response to treatment with TGFbeta1 , C3A cells produced mRNA encoding the pro-alphaI chain of procollagen I , secreted procollagen I peptide , up-regulated production of the proinflammatory cytokine interleukin-8 ( IL-8 ) and the mesenchymal marker vimentin , and down-regulated albumin production .\n\nMost of these responses were considerably enhanced when cells were fat-loaded with oleate and were attenuated by selenium addition at a dose that optimised the expression of thioredoxin reductase and glutathione peroxidase .\n\nCONCLUSIONS Our data establish that both fat-loading and suboptimal selenium status enhance collagen and IL-8 production by C3A hepatocytes in response to TGFbeta1 , possibly as part of an epithelial to mesenchymal transition .\n\nGENERAL SIGNIFICANCE These findings suggest that the hepatocyte may be an important contributor to the pathogenesis of fibrosis associated with NAFLD .", "output": "Tumor promoting inflammation" }, { "input": "The aryl hydrocarbon receptor ( AhR ) is a ligand-activated transcription factor that mediates a spectrum of toxic and biological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds .\n\nSeveral reports have shown that the AhR plays an important role in the control of cell-cycle progression , and this function is thought to be partly associated with the tumor promotion activity of dioxin .\n\nHowever , the underling mechanisms are not fully understood .\n\nWe have previously shown that overexpression of AhR , as well as AhR ligand treatment , stimulates cell proliferation of human lung cancer A549 cells .\n\nIn AhR-activated cells , the expression levels of DNA synthesis-related genes such as proliferating cell nuclear antigen ( PCNA ) and RFC38 are notably increased .\n\nExpression of these genes is mainly regulated by E2F1 , a transcription factor that is crucial for transition of the cell cycle from G1 to S phase .\n\nWe show here that the transcriptional activity of E2F1 is increased by the AhR agonist treatment and that this effect depends on the presence of AhR .\n\nFunctional mapping of AhR showed that the Per-Arnt-Sim ( PAS ) B domain is required for promotion of E2F1 activity .\n\nThe mechanism involves formation of a complex of AhR and E2F1 on the regulatory region in the E2F1 target gene , followed by recruitment of coactivator activator for thyroid hormone and retinoid receptors ( ACTR ) .\n\nConsequently , the results in this study indicate the physiological function of AhR as a potent transcriptional coactivator of E2F1-dependent transcription and implicate the AhR-E2F1 interaction as a part of the mechanism by which AhR/Arnt promotes cell proliferation .", "output": "Sustaining proliferative signaling" }, { "input": "Although anticancer effect of gambogic acid ( GA ) and its potential mechanisms were well documented in past decades , limited information is available on the anticancer effect of gambogenic acid ( GNA ) , another major active component of Gamboge .\n\nHere we performed a study to determine whether GNA possesses anticancer effect and find its potential mechanisms .\n\nThe results suggested that GNA significantly inhibited the proliferation of several tumor cell lines in vitro and in vivo .\n\nTreatment with GNA dose and time dependently induced A549 cells apoptosis , arrested the cells to G0/G1 phase in vitro and down-regulated the expression of cyclin D1 and cyclooxygenase ( COX)-2 in mRNA level .\n\nIn addition , anticancer effect was further demonstrated by applying xenografts in nude mice coupled with the characteristic of apoptosis in the GNA treated group .\n\nTaken together , these observations might suggest that GNA inhibits tumor cell proliferation via apoptosis-induction and cell cycle arrest .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Cancer cells upregulate glycolysis , increasing glucose uptake to meet energy needs .\n\nA small fraction of a cell's glucose enters the hexosamine biosynthetic pathway ( HBP ) , which regulates levels of O-linked beta-N-acetylglucosamine ( O-GlcNAc ) , a carbohydrate posttranslational modification of diverse nuclear and cytosolic proteins .\n\nWe discovered that breast cancer cells upregulate the HBP , including increased O-GlcNAcation and elevated expression of O-GlcNAc transferase ( OGT ) , which is the enzyme catalyzing the addition of O-GlcNAc to proteins .\n\nReduction of O-GlcNAcation through RNA interference of OGT in breast cancer cells leads to inhibition of tumor growth both in vitro and in vivo and is associated with decreased cell-cycle progression and increased expression of the cell-cycle inhibitor p27(Kip1) .\n\nElevation of p27(Kip1) was associated with decreased expression and activity of the oncogenic transcription factor FoxM1 , a known regulator of p27(Kip1) stability through transcriptional control of Skp2 .\n\nReducing O-GlcNAc levels in breast cancer cells decreased levels of FoxM1 protein and caused a decrease in multiple FoxM1-specific targets , including Skp2 .\n\nMoreover , reducing O-GlcNAcation decreased cancer cell invasion and was associated with the downregulation of matrix metalloproteinase-2 , a known FoxM1 target .\n\nFinally , pharmacological inhibition of OGT in breast cancer cells had similar anti-growth and anti-invasion effects .\n\nThese findings identify O-GlcNAc as a novel mechanism through which alterations in glucose metabolism regulate cancer growth and invasion and suggest that OGT may represent novel therapeutic targets for breast cancer .", "output": "Cellular energetics, Evading growth suppressors" }, { "input": "Ataxia telangiectasia ( AT ) cells , with their defective double-strand break ( DSB ) repair processes , exhibit high sensitivity to low-LET radiation such as X-rays irradiation and gamma beams .\n\nSince heavy ion beam treatment for cancer is becoming increasingly common in Japan and elsewhere , it is important to also determine their sensitivity to high-LET radiation .\n\nFor this purpose we irradiated AT and normal human cells immortalized with the human telomerase gene using high- ( 24-60 keV/microm carbon and 200 keV/microm iron ions ) or low-LET ( X-rays ) radiation in non-proliferative conditions .\n\nIn normal cells the RBE ( relative biological effectiveness ) of carbon and iron ions increased from 1.19 to 1.81 in proportion to LET .\n\nIn contrast , their RBE in AT cells increased from 1.32 at 24 keV/microm to 1.59 at 40 keV/microm , and exhibited a plateau at over 40 keV/microm .\n\nIn normal cells most gamma-H2AX foci induced by both carbon- and iron-ion beams had disappeared at 40 h .\n\nIn AT cells , however , a significant number of gamma-H2AX foci were still observed at 40 h .\n\nThe RBEs found in the AT cells after heavy-ion irradiation were consistent with the effects predicted from the presence of non-homologous end joining defects .\n\nThe DSBs remaining after heavy-ion irradiation suggested defects in the AT cells ' DSB repair ability .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Inactivation of p53 is involved in arsenite-induced tumorigenesis ; however , the molecular mechanisms remain poorly understood .\n\nOBJECTIVE We investigated the molecular mechanisms underlying the inactivation of p53 and neoplastic transformation induced by arsenite in human embryo lung fibroblast ( HELF ) cells .\n\nMETHODS Anchorage-independent growth assays were performed , and tumorigenicity in intact animals was assessed to confirm arsenite-induced neoplastic transformation .\n\nWe determined the levels and functions of p53 , nuclear factor-kappa B ( NF-B ; a key transcriptional regulator ) , and mot-2 ( a p53 inhibitor ) and their relationships in arsenite-induced transformed HELF cells by two-dimensional electrophoresis , reverse-transcriptase polymerase chain reaction , Western blot , immunofluorescence , and co-immunoprecipitation assays .\n\nRESULTS Exposure of HELF cells to low levels of arsenite increased their proliferation rate and anchorage-independent growth and disrupted normal contact inhibition .\n\nWhen introduced into nude mice , transformed cells were tumorigenic .\n\nWe used proteomic analysis to identify proteins with altered expression between untreated and arsenite-exposed cells .\n\nWe found decreased expression of NF-B repressing factor ( NKRF ; an inhibitor of NF-B-mediated gene transcription ) , increased expression of mot-2 , and increased activation of NF-B .\n\nChanges in cells exposed to 1.0 microM arsenite were more marked than changes in cells exposed to 0.5 or 2.0 microM arsenite .\n\nInactivation of NF-B prevented malignant transformation induced by 1.0 microM arsenite .\n\nMoreover , we also identified a mechanism whereby NF-B regulated p53 .\n\nSpecifically , activation of NF-B up-regulated mot-2 expression , which prevented nuclear translocation of p53 and switched the binding preference of the p53 and NF-B coactivator CBP [ cyclic AMP-responsive element binding protein ( CREB ) binding protein ] from p53 to NF-B .\n\nCONCLUSIONS mot-2-mediated cross talk between NF-B and p53 appears to be involved in arsenite-induced tumorigenesis of HELF cells .", "output": "Evading growth suppressors" }, { "input": "Previous epidemiologic observational and experimental studies investigated the potential of antioxidant micronutrients to modulate cancer risk , but these studies produced inconsistent results .\n\nIn this pilot , randomized , double-blind , placebo-controlled clinical trial ( n = 47 ) , we assessed the effects of an antioxidant micronutrient combination ( 800 mg dl-alpha-tocopherol acetate , 24 mg beta-carotene , 1.0 g vitamin C , 200 microg l-selenomethionine , 7.2 mg riboflavin , 80 mg niacin , 60 mg zinc , 5 mg manganese ) given daily over 4 months on oxidative and inflammatory biomarkers in patients with a history of sporadic colorectal adenoma .\n\nPlasma tumor necrosis factor-alpha ( TNF-alpha ) , interleukin-6 , and F2-isoprostane concentrations were measured using ELISAs , and cystine ( CySS ) was measured using high-performance liquid chromatography .\n\nPlasma TNF-alpha concentration decreased in the active treatment group by 37% relative to the placebo group ( P = 0.002 ) , and CySS decreased by 19% ( P = 0.03 ) ; however , interleukin-6 and F2-isoprostane concentrations decreased in antioxidant-treated nonsmokers but increased in smokers , although these findings were not statistically significant .\n\nThe decreases of TNF-alpha and CySS were more pronounced in nonsmokers .\n\nThese data suggest that ( a ) an antioxidant micronutrient cocktail can modulate biomarkers of oxidative stress and inflammation in humans and ( b ) the effects of antioxidant micronutrient supplementation on biomarkers of inflammation and oxidative stress may differ according to smoking status .", "output": "Tumor promoting inflammation" }, { "input": "The tumor suppressor p27(Kip1) is an inhibitor of cyclin/cyclin-dependent kinase ( CDK ) complexes and plays a crucial role in cell cycle regulation .\n\nNevertheless , p27 function in the tumorigenesis of the uterine cervix has been poorly defined .\n\nSome phenomenon hints that HPV E7 protein can enhance p27 expression , which is contradictory to HPV E7's property of increasing cell proliferation rate .\n\nSo , in the present study , we have examined the effect of E7 on p27 expression .\n\nThough the levels of p27 are increased after HPV E7 expression , most of the p27 protein localized in the cytoplasm and have no function on cell cycle arrest and contact inhibition .\n\nThe cell migration rate is elevated when p27 is high expression and located in cytoplasm .\n\nThe results indicated that E7-p27 interaction not only abolished the p27's cell cycle inhibitory function by sequestering it to the cytoplasm , but also endow the cell with invasive property which is the feature of malignant cells .", "output": "Evading growth suppressors" }, { "input": "Bilateral spontaneous pneumothorax is a rare occurrence in patients with both primary and metastatic lung cancer .\n\nPneumothorax occurring as a complication of vascular endothelial growth factor receptor ( VEGFR ) inhibitor therapy has not been previously described in the medical literature .\n\nSunitinib malate is a VEGFR inhibitor approved for the treatment of advanced renal cell carcinoma .\n\nWe present a patient with metastatic renal cell carcinoma manifested as bilateral pulmonary nodules who developed a bilateral spontaneous pneumothorax 3 weeks after initiation of sunitinib therapy .\n\nWe believe that sunitinib therapy resulted in necrosis of multiple pleural-based pulmonary nodules with central cavernization and ultimately rupture with bronchopleural fistula formation .\n\nBased on this experience , we advise that practitioners exercise caution when prescribing anti-VEGFR therapy in patients with pleural-based pulmonary metastases and recognize that the efficacy and toxicity of these agents may be closely linked .", "output": "Resisting cell death" }, { "input": "DNA double strand breaks ( DSBs ) arise from spontaneous DNA damage due to metabolic activities or from direct and indirect damaging effects of stress .\n\nDSBs are also formed transiently during such processes as replication , transcription , and DNA repair .\n\nThe level of DSBs positively correlates with the activities of homologous and nonhomologous DNA repair pathways , which in turn inversely correlate with methylation levels and chromatin structure .\n\nThus , measurement of strand breaks can provide an informative picture of genome stability of a given cell .\n\nThe use of random oligonucleotide-primed synthesis for the analysis of DSB levels is described .\n\nApplications of the assay for quantitative detection of 3'OH , 3'P , or DNA strand breaks at a cleavage site of the deoxyribose residue are discussed .", "output": "Genomic instability and mutation" }, { "input": "INTRODUCTION Statins are cholesterol-lowering drugs with pleiotropic activities including inhibition of isoprenylation and reduction of signals driving cell proliferation and survival responses .\n\nMETHODS In this study we evaluated the effects of lovastatin acid and lactone on breast cancer MDAMB231 and MDAMB468 cells using a combination of proteomic and metabonomic profiling techniques .\n\nRESULTS Lovastatin inhibited proliferation of breast cancer cell lines .\n\nMDAMB231 cells were more sensitive to its effects , and in most cases lovastatin acid showed more potency towards the manipulation of protein expression than lovastatin lactone .\n\nIncreased expression of Rho inhibitor GDI-2 stabilized the non-active Ras homolog gene family member A ( RhoA ) leading to a decreased expression of its active , membrane-bound form .\n\nIts downstream targets cofilin , CDC42 and G3BP1 are members of the GTPase family affected by lovastatin .\n\nOur data indicated that lovastatin modulated the E2F1-pathway through the regulation of expression of prohibitin and retinoblastoma ( Rb ) .\n\nThis subsequently leads to changes of E2F-downstream targets minichromosome maintenance protein 7 ( MCM7 ) and MutS homolog 2 ( MSH2 ) .\n\nLovastatin also regulated the AKT-signaling pathway .\n\nIncreased phosphatase and tensin homolog ( PTEN ) and decreased DJ-1 expression lead to a down-regulation of the active pAkt .\n\nLovastatin's involvement in the AKT-signaling pathway was confirmed by an upregulation of its downstream target , tumor progressor NDRG1 .\n\nMetabolic consequences to lovastatin exposure included suppression of glycolytic and Krebs cycle activity , and lipid biosynthesis .\n\nCONCLUSIONS The combination of proteomics and metabonomics enabled us to identify several key targets essential to the antitumor activity of lovastatin .\n\nOur results imply that lovastatin has the potential to reduce the growth of breast cancer cells .", "output": "Sustaining proliferative signaling" }, { "input": "INTRODUCTION Various agents used in breast cancer chemotherapy provoke DNA double-strand breaks ( DSBs ) .\n\nDSB repair competence determines the sensitivity of cells to these agents whereby aberrations in the repair machinery leads to apoptosis .\n\nProteins required for this pathway can be detected as nuclear foci at sites of DNA damage when the pathway is intact .\n\nHere we investigate whether focus formation of repair proteins can predict chemosensitivity of breast cancer .\n\nMETHODS Core needle biopsy specimens were obtained from sixty cases of primary breast cancer before and 18-24 hours after the first cycle of neoadjuvant epirubicin plus cyclophosphamide ( EC ) treatment .\n\nNuclear focus formation of DNA damage repair proteins was immunohistochemically analyzed and compared with tumor response to chemotherapy .\n\nRESULTS EC treatment induced nuclear foci of gammaH2AX , conjugated ubiquitin , and Rad51 in a substantial amount of cases .\n\nIn contrast , BRCA1 foci were observed before treatment in the majority of the cases and only decreased after EC in thirteen cases .\n\nThe presence of BRCA1- , gammaH2AX- , or Rad51-foci before treatment or the presence of Rad51-foci after treatment was inversely correlated with tumor response to chemotherapy .\n\nDNA damage response ( DDR ) competence was further evaluated by considering all four repair indicators together .\n\nA high DDR score significantly correlated with low tumor response to EC and EC + docetaxel whereas other clinicopathological factors analyzed did not .\n\nCONCLUSIONS High performing DDR focus formation resulted in tumor resistance to DNA damage-inducing chemotherapy .\n\nOur results suggested an importance of evaluation of DDR competence to predict breast cancer chemosensitivity , and merits further studying into its usefulness in exclusion of non-responder patients .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Astaxanthin modulates immune response , inhibits cancer cell growth , reduces bacterial load and gastric inflammation , and protects against UVA-induced oxidative stress in in vitro and rodent models .\n\nSimilar clinical studies in humans are unavailable .\n\nOur objective is to study the action of dietary astaxanthin in modulating immune response , oxidative status and inflammation in young healthy adult female human subjects .\n\nMETHODS Participants ( averaged 21.5 yr ) received 0 , 2 , or 8 mg astaxanthin ( n = 14/diet ) daily for 8 wk in a randomized double-blind , placebo-controlled study .\n\nImmune response was assessed on wk 0 , 4 and 8 , and tuberculin test performed on wk 8 .\n\nRESULTS Plasma astaxanthin increased ( P < 0.01 ) dose-dependently after 4 or 8 wk of supplementation .\n\nAstaxanthin decreased a DNA damage biomarker after 4 wk but did not affect lipid peroxidation .\n\nPlasma C-reactive protein concentration was lower ( P < 0.05 ) on wk 8 in subjects given 2 mg astaxanthin .\n\nDietary astaxanthin stimulated mitogen-induced lymphoproliferation , increased natural killer cell cytotoxic activity , and increased total T and B cell subpopulations , but did not influence populations of Thelper , Tcytotoxic or natural killer cells .\n\nA higher percentage of leukocytes expressed the LFA-1 marker in subjects given 2 mg astaxanthin on wk 8 .\n\nSubjects fed 2 mg astaxanthin had a higher tuberculin response than unsupplemented subjects .\n\nThere was no difference in TNF and IL-2 concentrations , but plasma IFN-gamma and IL-6 increased on wk 8 in subjects given 8 mg astaxanthin .\n\nCONCLUSION Therefore , dietary astaxanthin decreases a DNA damage biomarker and acute phase protein , and enhances immune response in young healthy females .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "BACKGROUND The pTalpha/preTCR regulates the beta-selection , a crucial T-cell developmental checkpoint , providing a most potent survival advantage to thymocytes mediated by the src-kinase p56(Lck) .\n\nMETHODS To define the relevance of pTalpha in human T-cell lymphoblastic leukemia ( T-ALL ) , we analyzed in T-ALL cell lines ( n=14 ) pTalpha and p56(Lck) mRNA and protein expression as also the tyrosine-phosphorylation .\n\nThe p56(Lck) specific src-protein-tyrosine kinase inhibitor ( PTK-I ) PP1 was used in growth inhibition assays .\n\nIC(50) value determination , cell cycle- and apoptosis analyses were performed in T-ALL- , non-T-ALL- and murine transgenic cell lines .\n\nRESULTS pTalpha expression patterns were markedly different in T-ALL cell lines as compared to those reported for normal lymphoid counterparts .\n\nPP1 induced in 6/11 T-ALL cell lines a survival disadvantage resulting from a cell cycle arrest in the G(1/0) phase in thymic lymphoblastic cells and apoptosis induction in the immature cell line HSB-2 , respectively .\n\nPP1 sensitive cell lines expressed the target protein p56(Lck) and showed a corresponding P-Tyr signal .\n\nCONCLUSION Sensitivity of thymic T-ALLs to PP1 clearly underlines the impact of pTalpha mediated proliferation in this leukemic sub-type .\n\nIn addition , p56(Lck) represents also independently of pTalpha a promising therapeutical target for the src-kinase inhibitors in neoplastic lymphoid diseases .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Prostate cancer ( PCA ) is the most common invasive malignancy and the second leading cause of cancer-related death in males .\n\nThe present study investigated the effects of fangchinoline ( Fan ) , an important compound in Stephania Tetradra S. Moore ( Fenfangji ) with pain-relieving , blood pressure-depressing , and antibiotic activities , on human PCA .\n\nIt was found that Fan inhibited human prostate cancer cell lines ( PC3 ) cell proliferation in a dose- and time-dependent manner .\n\nStudies of cell-cycle progression showed that the anti-proliferative effect of Fan was associated with an increase in the G1/S phase of PC3 cells .\n\nWestern blot results indicated that Fan-induced G1/S phase arrest was mediated through inhibition of cyclin-regulated signaling pathways .\n\nFan induced p27 expression and inhibited cyclin D and proliferating cell nuclear antigen ( PCNA ) expression in PC3 cells .\n\nIncreased exposure time to Fan caused apoptosis of PC3 cells , which was associated with up-regulation of pro-apoptotic proteins Bax and caspase 3 , and down-regulation of anti-apoptotic protein Bcl-2 .\n\nFurthermore , Fan had anti-tumorigenic activity in vivo , including reduction of tumor volume and pro-apoptotic and anti-proliferative effects in a PC3 nude mouse xenograft .\n\nTaking all this together , it can be concluded that Fan is an effective anti-proliferative agent that modulates cell growth regulators in prostate cancer cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Although the immunomodulatory effects of many herbs have been extensively studied , research related to possible immunomodulatory effects of various spices is relatively scarce .\n\nHere , the potential immunomodulatory effects of black pepper and cardamom are investigated .\n\nOur data show that black pepper and cardamom aqueous extracts significantly enhance splenocyte proliferation in a dose-dependent , synergistic fashion .\n\nEnzyme-linked immunosorbent assay experiments reveal that black pepper and cardamom significantly enhance and suppress , respectively , T helper ( Th)1 cytokine release by splenocytes .\n\nConversely , Th2 cytokine release by splenocytes is significantly suppressed and enhanced by black pepper and cardamom , respectively .\n\nExperimental evidence suggests that black pepper and cardamom extracts exert pro-inflammatory and anti-inflammatory roles , respectively .\n\nConsistently , nitric oxide production by macrophages is significantly augmented and reduced by black pepper and cardamom , respectively .\n\nRemarkably , it is evident that black pepper and cardamom extracts significantly enhance the cytotoxic activity of natural killer cells , indicating their potential anti-cancer effects .\n\nOur findings strongly suggest that black pepper and cardamom exert immunomodulatory roles and antitumor activities , and hence they manifest themselves as natural agents that can promote the maintenance of a healthy immune system .\n\nWe anticipate that black pepper and cardamom constituents can be used as potential therapeutic tools to regulate inflammatory responses and prevent/attenuate carcinogenesis .", "output": "Tumor promoting inflammation" }, { "input": "OBJECTIVE To study the effects of genistein on the proliferation , apoptosis induction and expression of related gene proteins of human colon cancer cells in vitro and in vivo , and its mechanisms of action .\n\nMETHODS MTT colorimetric assay was used to detect the effects of genistein on the proliferation of human colon adenocarcinoma SW480 cells .\n\nLight and transmission electron microscopy were used to study the histological and ultrastructural changes .\n\nFlow cytometry was used to determine the effects of genistein on cell cycle and apoptosis .\n\nFlow cytometry and immunohistochemistry were used to determine the effects of genistein on apoptosis induction and expression of related gene proteins of colon cancer cells .\n\nRESULTS The MTT colorimetric assay showed that genistein inhibited the proliferation of SW480 cells in a dose-dependent and time-dependent manner , and the highest inhibition rate was 60.2% after 80 microg/ml genistein treatment for 72 h .\n\nThe light microscopy revealed that many genistein-treated cancer cells were shrunken , disrupted , or showing cytoplasmic vacuolization .\n\nThe electron microscopic examination showed cell shrinkage , nuclear fragmentation and pronounced chromatin condensation , sometimes formed crescent chromatin condensation attached to the nuclear membrane .\n\nThe results of flow cytometry showed that : after SW480 cells were treated with 0 , 20 , 40 , 80 microg/ml genistein for 48 h , the FI values of PCNA were 1.49 +/- 0.02 , 1.28 +/- 0.04 , 1.14 +/- 0.03 , and 0.93 +/- 0.08 ; the FI values of VEGF were 1.75 +/- 0.02 , 1.34 +/- 0.06 , 1.32 +/- 0.04 , and 1.23 +/- 0.04 ; the fluorescence index ( FI ) values of p21 were 1.26 +/- 0.05 , 1.36 +/- 0.06 , 1.61 +/- 0.03 , and 1.73 +/- 0.03 , respectively .\n\nThere were statistically significant differences between the control group and each treatment group ( P < 0.05 or P < 0.01 ) .\n\nThe scores of immunohistochemical staining of PCNA and VEGF proteins were decreased , while p21 increased .\n\nThere were statistically significant differences between the control group and each treatment group ( P < 0.05 or P < 0.01 ) .\n\nCONCLUSION Genistein can inhibit the growth of colon cancer cells via apoptosis induction and cell cycle arrest at G(2)/M phase .\n\nThe anti-tumor mechanisms of genistein may be related with the down-regulation of expression of VEGF and PCNA , and up-regulation of the expression of p21 .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND Dendritic cells ( DCs ) isolated from tumor bearing animals or from individuals with solid tumors display functional abnormalities and the DC impairment has emerged as one mechanism for tumor evasion from the control of the immune system .\n\nDuctal pancreatic adenocarcinoma ( PDAC ) , the most common pancreatic cancer , is recognized as a very aggressive cancer type with a mortality that almost matches the rate of incidence .\n\nMETHODS We examined the systemic influence ductal pancreatic adenocarcinoma ( PDAC ) exerted on levels of peripheral blood DCs and inflammatory mediators in comparison to the effects exerted by other pancreatic tumors , chronic pancreatitis , and age-matched controls .\n\nRESULTS All groups examined , including PDAC , had decreased levels of myeloid DCs ( MDC ) and plasmacytoid DCs ( PDC ) and enhanced apoptosis in these cells as compared to controls .\n\nWe found elevated levels of PGE2 and CXCL8 in subjects with PDAC , and chronic pancreatitis .\n\nLevels of these inflammatory factors were in part restored in PDAC after tumor resection , whereas the levels of DCs were impaired in the majority of these patients approximately 12 weeks after tumor removal .\n\nOur results prove that solid pancreatic tumors , including PDAC , systemically affect blood DCs .\n\nThe impairments do not seem to be tumor-specific , since similar results were obtained in subjects with chronic pancreatitis .\n\nFurthermore , we found that PDAC patients with a survival over 2 years had significant higher levels of blood DCs compared to patients with less than one year survival .\n\nCONCLUSIONS Our findings points to the involvement of inflammation in the destruction of the blood MDCs and PDCs .\n\nFurthermore , the preservation of the blood DCs compartment in PDAC patients seems to benefit their ability to control the disease and survival .", "output": "Tumor promoting inflammation" }, { "input": "Targeting cancer cell metabolism is a new promising strategy to fight cancer .\n\nMetformin , a widely used antidiabetic agent , exerts antitumoral and antiproliferative action .\n\nIn this study , the addition of metformin to 2-deoxyglucose ( 2DG ) inhibited mitochondrial respiration and glycolysis in prostate cancer cells leading to a severe depletion in ATP .\n\nThe combination of the two drugs was much more harmful for cancer cells than the treatment with metformin or 2DG alone , leading to 96% inhibition of cell viability in LNCaP prostate cancer cells .\n\nIn contrast , a moderate effect on cell viability was observed in normal prostate epithelial cells .\n\nAt the cellular level , the combination of metformin and 2DG induced p53-dependent apoptosis via the energy sensor pathway AMP kinase , and the reexpression of a functional p53 in p53-deficient prostate cancer cells restored caspase-3 activity .\n\nIn addition to apoptosis , the combination of metformin and 2DG arrested prostate cancer cells in G(2)-M .\n\nThis G(2)-M arrest was independent of p53 and correlated with a stronger decrease in cell viability than obtained with either drug .\n\nFinally , metformin inhibited 2DG-induced autophagy , decreased beclin 1 expression , and triggered a switch from a survival process to cell death .\n\nOur study reinforces the growing interest of metabolic perturbators in cancer therapy and highlights the potential use of the combination of metformin and 2DG as an anticancerous treatment .", "output": "Cellular energetics, Resisting cell death" }, { "input": "Tumor invasion and metastasis are the primary causes of cancer patient mortality , underscoring the need for identification of novel genes and signaling pathways that mediate these prognosis-determining phenomena .\n\nTo identify and characterize novel lung adenocarcinoma genes associated with lung cancer progression , we created a bioinformatics-based approach that focuses on human cell-cycle-regulated genes that have evolved only in higher organisms but not in lower eukaryotic cells .\n\nIn siRNA experiments in lung cancer cells , FLJ10540 was identified as one of several novel targets involved in cell migration and invasion .\n\nHere , we demonstrate that PI3K inhibition affects FLJ10540-mediated cell migration and invasion and further , that FLJ10540 knockdown ablates AKT-Ser(473) phosphorylation .\n\nTaken together , these findings indicate that the FLJ10540/PI3K/AKT pathway may harbor new therapeutic targets for treating invasive lung adenocarcinoma .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "1-(3-C-ethynyl-beta-d-ribo-pentofuranosyl)cytosine ( ECyd ) is a ribose-modified nucleoside analog of cytidine with potent anticancer activity in several cancers .\n\nThe main antitumor mechanism of this promising RNA-directed nucleoside anti-metabolite is efficient blockade of RNA synthesis in cancer cells .\n\nHere , we examined the therapeutic potential of this RNA-directed anti-metabolite in in vitro models of nasopharyngeal cancer ( NPC ) .\n\nIn a panel of 6 NPC cell lines , ECyd effectively inhibited cellular proliferation at nM concentrations ( IC(50) : approximately 13-44nM ) .\n\nMoreover , cisplatin-resistant NPC cells were highly sensitive to ECyd ( at nM concentration ) .\n\nThe ECyd-mediated growth inhibition was associated with G(2)/M cell cycle arrest , PARP cleavage ( a hallmark of apoptosis ) and Bcl-2 downregulation , indicating induction of apoptosis by ECyd in NPC cells .\n\nUnexpectedly , ECyd-induced significant downregulation of TIGAR , a newly described dual regulator of apoptosis and glycolysis .\n\nMore importantly , this novel action of ECyd on TIGAR was accompanied by marked depletion of NADPH , the major reducing power critically required for cell proliferation and survival .\n\nWe hypothesized that ECyd-induced TIGAR downregulation was crucially involved in the antitumor activity of ECyd .\n\nIndeed , overexpression of TIGAR was able to rescue NPC cells from ECyd-induced growth inhibition , demonstrating a novel mechanistic action of ECyd on TIGAR .\n\nWe demonstrated for the first time that an RNA-directed nucleoside analog , ECyd , exerts its antitumor activity via downregulation of a novel regulator of apoptosis , TIGAR .\n\nMoreover , ECyd may represent a novel therapy for NPC .", "output": "Cellular energetics, Resisting cell death" }, { "input": "OBJECTIVE To analyze histological factors not routinely assessed as potential prognostic factors in renal cell carcinoma , such as tumor necrosis , microscopic vascular invasion , and sinus fat invasion .\n\nMATERIALS AND METHODS A retrospective , analytical study was conducted of surgical specimens from 139 patients with localized renal cell carcinoma who underwent nephrectomy from 1993 to 2005 .\n\nTumor necrosis , microscopic vascular invasion , and sinus fat invasion were analyzed and compared to the classical factors : TNM classification , Fuhrman grade , and tumor size .\n\nFor statistical analysis , variables analyzed were categorized as pT1 , 2 vs pT3 , 4 ; Fuhrman grade 1 , 2 vs 3 , 4 ; tumor size < 7 cm vs >or= 7cm ; tumor necrosis vs no tumor necrosis ; microvascular invasion of sinus fat vs no invasion .\n\nCancer-specific survival probability and disease-free survival were calculated .\n\nA descriptive and analytical statistical analysis was performed using logistic regression for univariate and multivariate analyses .\n\nDependent variables were used to analyze cancer-specific survival rates .\n\nDisease-free survival was estimated using a Cox regression model and Kaplan-Meier curves .\n\nRESULTS In the univariate analysis , all variables analyzed had a significant influence on death for renal cell carcinoma .\n\nIn the multivariate analysis , the variable having the greatest influence was Fuhrman grade ( p = 0,032 ) .\n\nThe variables significantly influencing disease-free survival , estimated by the Cox method , were the pT stage ( p = 0.038 ) and Fuhrman grade ( p = 0.048 ) .\n\nCONCLUSIONS In patients with clinically localized renal cell carcinoma undergoing nephrectomy , pT stage and Fuhrman grade are the most important prognostic factors for survival and disease-free survival .\n\nTumor necrosis , microscopic vascular invasion , and sinus fat invasion are prognostic factors for death from renal carcinoma which are associated to TNM classification , Fuhrman grade , and tumor size .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "BACKGROUND Adenoid cystic carcinoma ( ACC ) of the Breast is a rare tumour ( less than 1 % of all breast carcinomas ) .\n\nThe aim of this study was to determine the clinical , histological and immunohistochemical characteristics of these tumours .\n\nMETHODS From the database of the Bergoni\u00e9 Institute of Bordeaux , 30 cases of ACC were identified .\n\nThe clinical and histological features of these carcinomas were characterized .\n\nAn immunohistochemical study was performed with the following antibodies : ER , PR , HER-2-neu , Vimentin , EGFR , P63 , SMA , CK5/6 , CK8/18 , CK14 , cKIT , MIB1 , CD44 and CD24 .\n\nRESULTS Thirty patients were included ( median age 60.7 years ) .\n\nThe 10 axillary lymph node dissections and two sentinel lymph procedures were negative .\n\nThe architecture was frequently of a mixed type ( 26/30 ) and less often solid ( 4/30 ) .\n\nAmong the 23 patients for whom follow up was available ( median follow-up : 84 months [ 2-288] ) , there were three local recurrences and three metastatic events .\n\nThe tumors with recurrence and metastasis showed more necrosis , a mitotic count greater than 4/10hpf , and in one case perineural infiltration .\n\nAll the tumours were ER , PR and Her-2-neu negative .\n\nMorphological and immunophenotypical analysis disclosed in each tumor , a basaloid and a luminal cell population with divergent immunophenotypical patterns .\n\nCONCLUSIONS The mammary ACC is made of two cell types and is of good prognosis despite its triple negative phenotype , similar to the basal-like infiltrating carcinoma NOS .\n\nAxillary lymph node dissection is not recommended .\n\nGood local control by at least large lumpectomy with long-term follow-up is necessary .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "OBJECTIVES To evaluate the impact of the category of unclassified renal cell carcinoma ( URCC ) on survival following nephrectomy .\n\nMETHODS Patients with clear cell RCC ( ccRCC , n = 3048 ) and URCC ( n = 38 ) were identified .\n\nPatients with URCC were matched 4:1 with ccRCC patients based on year of surgery , symptoms at presentation , tumor size , stage , regional lymph node involvement , metastases , grade , coagulative tumor necrosis , and sarcomatoid differentiation .\n\nSurvival was estimated using the Kaplan-Meier method and compared between ccRCC and URCC patients using log-rank tests .\n\nRESULTS Patients with URCC were more likely to have regional lymph node involvement ( P <.001 ) , higher grade ( P <.001 ) , tumor necrosis ( P <.001 ) , and sarcomatoid differentiation ( P <.001 ) as compared to patients with ccRCC .\n\nOverall survival was not significantly different between URCC and ccRCC patients in either the unmatched ( P = .337 ) or matched ( P = .345 ) cohorts .\n\nCancer-specific survival was significantly worse for URCC patients compared with unmatched ccRCC patients ( P = .020 ) .\n\nHowever , this difference was not statistically significant when the URCC patients were compared with the matched cohort ( P = .688 ) .\n\nDistant metastases-free survival was somewhat worse for M0 URCC patients compared with unmatched M0 ccRCC patients ( P = .063 ) , but not in the matched cohort ( P = .788 ) .\n\nCONCLUSIONS Although URCC is more likely to present with advanced clinicopathologic features compared with ccRCC , no statistically significant differences in outcome were noted after adjusting for these features in a matched analysis .", "output": "Resisting cell death" }, { "input": "Pilocytic astrocytoma is commonly viewed as a benign lesion .\n\nHowever , disease onset is most prevalent in the first two decades of life , and children are often left with residual or recurrent disease and significant morbidity .\n\nThe Hedgehog ( Hh ) pathway regulates the growth of higher WHO grade gliomas , and in this study , we have evaluated the activation and operational status of this regulatory pathway in pilocytic astrocytomas .\n\nExpression levels of the Hh pathway transcriptional target PTCH were elevated in 45% of tumor specimens analyzed ( ages 1-22 years ) and correlated inversely with patient age .\n\nEvaluation of a tissue array revealed oligodendroglioma-like features , pilomyxoid features , infiltration , and necrosis more commonly in specimens from younger patients ( below the median patient age of 10 years ) .\n\nImmunohistochemical staining for the Hh pathway components PTCH and GLI1 and the proliferation marker Ki67 demonstrated that patients diagnosed before the age of 10 had higher staining indices than those diagnosed after the age of 10 .\n\nA significant correlation between Ki67 and PTCH and GLI1 staining indices was measured , and 86% of Ki67-positive cells also expressed PTCH .\n\nThe operational status of the Hh pathway was confirmed in primary cell culture and could be modulated in a manner consistent with a ligand-dependent mechanism .\n\nTaken together , these findings suggest that Hh pathway activation is common in pediatric pilocytic astrocytomas and may be associated with younger age at diagnosis and tumor growth .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The BLM helicase is a member of the RecQ DNA helicase family and is mutated in the cancer-prone disorder Bloom syndrome .\n\nBLM plays a role in a number of cellular processes including DNA double-strand break repair , Holliday junction dissolution , and chromosome segregation .\n\nIn Drosophila melanogaster , the BLM ortholog ( DmBlm ) is encoded by the mus309 gene .\n\nTo study the role of DmBlm in double-strand break repair , we utilized a genetic assay in which a targeted DNA double-strand gap is created through excision of a P transposable element .\n\nBy recovering and molecularly analyzing individual repair products from wild-type and mus309 male pre-meiotic germline cells , we demonstrated that the DmBlm helicase is involved in homologous recombination downstream of strand invasion .\n\nThis assay can be adapted to test the roles of numerous DNA metabolic factors in DNA double-strand gap repair .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVES To investigate the effects of serotonin and melatonin ( MLT ) on the regulation of malignant growth and the activity of serotonin receptors ( 5HTR1a/-1b ) in prostate cancer ( PCa ) cell lines .\n\nMATERIALS AND METHODS In four PCa cell lines ( LNCaP , 22RV1 , PC3 , DU145 ) and two reference cell lines 5HTR1a and -1b , relative mRNA expression levels were assessed .\n\nDifferent serotonin and MLT receptor agonists and antagonists were used in stimulation and inhibition experiments .\n\nRESULTS mRNA expression of 5HTR1b was higher than that of 5HTR1a in all PCa cell lines .\n\nSerotonin showed a significant growth stimulatory effect in all PCa lines .\n\nThe 5HTR1a and -1b agonists/antagonists did not significantly affect viability .\n\nMLT inhibited viability only in PC3 cells .\n\nSimilarly , the 5HTR1a antagonist induced apoptotic changes in PC3 cells only at 10(-4)M , while the 5HTR1b antagonist induced necrosis at 10(-4)M in all cell lines .\n\nCell cycle alterations were seen in PC3 and DU145 cells under the influence of the 5HTR1a antagonist .\n\nCONCLUSIONS Serotonin receptor antagonists and agonists as well as MLT influence viability and apoptosis of PCa cell lines at supraphysiologic concentrations .\n\nIn contrast to other reports , our results do not support a regulatory role of serotonin or MLT receptor activation or inhibition in PCa growth .", "output": "Resisting cell death" }, { "input": "Paragangliomas are relatively uncommon neoplasms that arise in adrenal and extra-adrenal paraganglia of the autonomic nervous system .\n\nParasympathetic paraganglioma develop predominantly in the head and neck .\n\nIt is exceedingly uncommon to develop a primary intraparathyroid paraganglioma .\n\nThere is only a single case report in the English literature .\n\nThe information from the single previous case report ( Medline 1960-2009 ) was combined with this case report .\n\nOur patient was a 69 year old woman who presented with a thyroid gland mass , with extension into the substernal space .\n\nThe patient had a history of renal cell carcinoma removed 18 months before .\n\nAt surgery , a thyroid lobectomy and a parathyroidectomy were performed .\n\nThe parathyroid tissue showed a very well defined zellballen arrangement of paraganglion cells within the parenchyma of the parathyroid gland .\n\nThe cells had ample basophilic , granular cytoplasm .\n\nThe nuclei were generally round to oval with ' salt-and-pepper ' nuclear chromatin distribution .\n\nThere was a richly vascularized stroma .\n\nMitotic figures , necrosis , invasive growth , and profound nuclear pleomorphism were absent .\n\nThe neoplastic cells were strongly and diffusely immunoreactive with chromogranin , synaptophysin , CD56 , and focally with cyclin-D1 .\n\nThe paraganglioma showed a delicate S-100 protein positive supporting sustentacular framework .\n\nKeratin , CD10 , PTH , calcitonin and RCC markers were negative .\n\nThe patient showed no stigmata of Multiple Endocrine Neoplasia ( MEN ) and has no paraganglioma in any other anatomic site .\n\nShe is alive without any additional findings 12 months after surgery .\n\nIsolated paraganglioma within the parathyroid is rare , and should be separated from parathyroid adenoma , hyperplasia or metastatic disease to assure appropriate management .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "Ovarian smooth muscle tumors are a very rare type of ovarian tumor .\n\nIn this paper , we report the case of a 62-year-old woman who had a huge smooth muscle tumor of the right ovary .\n\nThe values of all the serum tumor markers were within normal limit .\n\nThe tumor measured 25 cm in diameter and weighed 6,200 g .\n\nHistological examination revealed that coagulative cellular atypia was moderate to severe , necrosis was not present and mitotic index was low .\n\nAccording to the criteria for the evaluation of the uterine smooth muscle tumors , this huge tumor was diagnosed as atypical leiomyoma .\n\nHowever , we finally made a diagnosis of this tumor as a smooth muscle tumor of uncertain malignant potential ( STUMP ) because of its huge size .\n\nFurther information is required regarding the characteristics of ovarian smooth muscle tumor and the propriety to introduce uterine tumor histological criteria to ovarian tumors .", "output": "Resisting cell death" }, { "input": "Stannous chloride ( SnCl(2) ) and UVA induce DNA lesions through ROS .\n\nThe aim of this work was to study the toxicity induced by UVA preillumination , followed by SnCl(2) treatment .\n\nE. coli BER mutants were used to identify genes which could play a role in DNA lesion repair generated by these agents .\n\nThe survival assays showed ( i ) The nfo mutant was the most sensitive to SnCl(2) ; ( ii ) lethal synergistic effect was observed after UVA pre-illumination , plus SnCl(2) incubation , the nfo mutant being the most sensitive ; ( iii ) wild type and nfo mutants , transformed with pBW21 plasmid ( nfo(+) ) had their survival increased following treatments .\n\nThe alkaline agarose gel electrophoresis assays pointed that ( i ) UVA induced DNA breaks and fpg mutant was the most sensitive ; ( ii ) SnCl(2)-induced DNA strand breaks were higher than those from UVA and nfo mutant had the slowest repair kinetics ; ( iii ) UVA + SnCl(2) promoted an increase in DNA breaks than SnCl(2) and , again , nfo mutant displayed the slowest repair kinetics .\n\nIn summary , Nfo protects E. coli cells against damage induced by SnCl(2) and UVA + SnCl(2) .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE Ovarian carcinomas mostly appear as large cystic masses .\n\nHowever , the exact prevalence of cysts in epithelial ovarian cancer ( EOC ) has never been documented as well as the tumor factors that are related to the presence of cysts .\n\nDemonstrating the prevalence of cysts in EOC is essential for research focused on predictive and prognostic biomarkers in ovarian cyst fluid .\n\nSTUDY DESIGN From 233 patients with primary EOC who underwent surgery , pathological data were collected from pathology reports .\n\nUnivariate and multivariate logistic regression were used to analyze the relationship between the presence of cysts and other tumor characteristics .\n\nRESULTS Cysts in EOC were present in 83.7% of the patients and were mostly ( 61% ) multilocular .\n\nThe most common histological subtypes ( serous , mucinous , endometrioid , clear cell ) contained cysts in more than 85% of the cases .\n\nIn univariate regression analysis , early FIGO stage , low tumor grade and a large tumor size were significantly associated with the presence of cysts ( OR ( 95% CI)=5.312 ( 1.81-15.57 ) , 6.906 ( 2.31-20.66 ) and 1.169 ( 1.08-1.27 ) , respectively ) .\n\nIn multivariate regression analysis , apart from tumor size , only tumor grade was independently associated with the presence of cysts ( adjusted OR ( 95% CI)=4.234 ( 1.36-13.22) ) .\n\nCONCLUSIONS The large majority of all EOCs contained cysts .\n\nHistological subtype , FIGO stage , tumor necrosis and age were not associated with the presence of cystic EOC .\n\nIn contrast , tumor grade and tumor size were independently related to the presence of cystic EOC .\n\nThis means that cystic EOCs represent a subgroup of larger and more well-differentiated tumors .\n\nThe evident relationship between the presence of cysts and differentiation grade is interesting from a clinical point of view as grading is especially important for the prognosis and treatment of patients with stage I EOC .", "output": "Resisting cell death" }, { "input": "PURPOSE Tamoxifen , a selective oestrogen receptor modulator ( SERM ) , and brivanib alaninate , a vascular endothelial growth factor receptor 2 ( VEGFR-2 ) inhibitor , are two target specific agents that result in a substantial decrease in tumour growth when given alone .\n\nTamoxifen activates SERM stimulated breast and endometrial tumour growth .\n\nTamoxifen and brivanib alaninate have side-effects that can affect therapeutic outcomes .\n\nThe primary goal of the current study was to evaluate the therapeutic effects of lower doses of both agents when given in combination to mice with SERM sensitive , oestrogen stimulated tumour xenografts ( MCF-7 E2 tumours ) .\n\nExperiments were conducted to evaluate the response of SERM stimulated breast ( MCF-7 Tam , MCF-7 Ral ) and endometrial tumours ( EnCa 101 ) to demonstrate the activity of brivanib alaninate in SERM resistant models .\n\nEXPERIMENTAL DESIGN In the current study , tumour xenografts were minced and bi-transplanted into the mammary fat pads of athymic , ovariectomised mice .\n\nPreliminary experiments were conducted to determine an effective oral dose of tamoxifen and brivanib alaninate that had minimal effect on tumour growth .\n\nDoses of 125 microg of tamoxifen and 0.05 mg/g of brivanib alaninate were evaluated .\n\nAn experiment was designed to evaluate the effect of the two agents together when started at the time of tumour implantation .\n\nAn additional experiment was done in which tumours were already established and then treated , to obtain enough tumour tissue for molecular analysis .\n\nRESULTS Brivanib alaninate was effective at inhibiting tumour growth in SERM sensitive ( MCF-7 E2 ) and SERM stimulated ( EnCa 101 , MCF-7 Ral , MCF-7 Tam ) models .\n\nThe effect of the low dose drug combination as an anti-tumour strategy for SERM sensitive ( MCF-7 E2 ) in early treatment was as effective as higher doses of either drug used alone .\n\nIn established tumours , the combination is successful at decreasing tumour growth , while neither agent alone is effective .\n\nMolecular analysis revealed a decreased phosphorylation of VEGFR-2 in tumours that were treated with brivanib alaninate and an increase in VEGFA transcription to compensate for the blockade of VEGFR-2 by increasing the transcription of VEGFA .\n\nTamoxifen increases the phosphorylation of VEGFR-2 and this effect is abrogated by brivanib alaninate .\n\nThere was also increased necrosis in tumours treated with brivanib alaninate .\n\nCONCLUSION Historically , tamoxifen has a role in blocking angiogenesis as well as the blockade of the ER .\n\nTamoxifen and a low dose of an angiogenesis inhibitor , brivanib alaninate , can potentially be combined not only to maximise therapeutic efficacy but also to retard SERM resistant tumour growth .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "Certain steroidal compounds have demonstrated an antiproliferative effect against several tumor cell lines ; however , their complete role on cancer cells is not currently established .\n\nHerein , we report the synthesis and evaluation of two new 26-hydroxy-22-oxocholestanic steroids on cervical cancer CaSki cells .\n\nThe title compounds were prepared from diosgenin and hecogenin in excellent yields .\n\nWe determined their effect on cell proliferation , cell cycle , and cell death .\n\nThe cytotoxic effect of the title compounds on CaSki and human lymphocytes was also evaluated , indicating that the main cell death process is not necrosis ; the null effect on lymphocytes implies that they are not cytotoxic .\n\nThe observation of apoptotic bodies as well as the increase in the expression of active caspase-3 along with the fragmentation of DNA confirmed that such new cholestanic frameworks induced apoptosis in tumor cells .\n\nSignificantly , their antiproliferative activity on tumor cells did not affect the proliferative potential of normal fibroblasts from cervix and peripheral blood lymphocytes .\n\nThe title compounds show selective antitumor activity and therefore serve as promising lead candidates for further optimization .", "output": "Resisting cell death" }, { "input": "Exposure to benzo[a]pyrene ( BaP ) can induce inflammatory skin diseases and skin cancer , which are both associated to oxidative stress .\n\nBaP is known to bind with high specificity to the aryl hydrocarbon receptor ( AhR ) , modifying the expression of CYP1A1 , involved both in cancer and inflammation .\n\nWhile the current knowledge is based on murine skin and cell culture data , in this study human healthy skin has been treated with 5muM BaP in conditions simulating occupational and environmental exposure .\n\nAhR and CYP1A1 expression was evaluated by Western blotting , which revealed their presence even in control untreated skin ; both enzyme and receptor increased more than twofold after exposure to BaP .\n\nAhR expression level was lower than CYP1A1 in basal conditions and following induction .\n\nOxidative stress was evaluated in terms of MTT reduction , protein peroxidation and reactive oxygen species ( ROS ) formation .\n\nA significant increase in ROS and carbonyl compound production , as well as reduced tissue viability have been determined by BaP .\n\nThe results of this experiment indicate that BaP , an AhR agonist , can significantly increase receptor and CYP1A1 expression and induce oxidative stress in human skin , confirming the involvement of this pathway in the pathogenesis of tissue damage due to polycyclic aromatic hydrocarbons .", "output": "Tumor promoting inflammation" }, { "input": "During the course of inflammation and its resolution , macrophages are exposed to various cytotoxic materials , including reactive oxygen species .\n\nThus , macrophages require a protective machinery against oxidative stress to survive at the inflammatory site .\n\nHere , we showed that xCT , a component of transport system x(c)(-) , was significantly up-regulated in activated infiltrating cells , including macrophages and neutrophils at the inflammatory site .\n\nSystem x(c)(-) mediates the uptake of extracellular L-cystine and is consequently responsible for maintenance of intracellular glutathione levels .\n\nWe established a loss-of-function mouse mutant line of xCT by N-ethyl-N-nitrosourea mutagenesis .\n\nMacrophages from xCT(mu/mu) mice showed cell death in association with the excessive release of high mobility group box chromosomal protein 1 upon stimulation with LPS , suggesting that xCT deficiency causes unremitting inflammation because of the impaired survival of activated macrophages at the inflammatory site .\n\nSubcutaneous injection of 3-methylcholanthrene ( 3-MCA ) induced the generation of fibrosarcoma in association with inflammation .\n\nWhen 3-MCA was injected s.c. into mice , xCT mRNA was up-regulated in situ .\n\nIn xCT(mu/mu) mice , inflammatory cytokines ( such as IL-1beta and TNFalpha ) were overexpressed , and the generation of 3-MCA-induced fibrosarcoma was accelerated .\n\nThese results clearly indicate that the defect of the protective system against oxidative stress impaired survival of activated macrophages and subsequently enhanced tumorigenecity .", "output": "Tumor promoting inflammation" }, { "input": "Colon cancer is a leading cause of morbidity and mortality in Western countries .\n\nBasic fibroblast growth factor ( bFGF ) was up-regulated in patients with colon cancer and was considered as a potential therapeutic target .\n\nIn this study , we first demonstrated that a novel bFGF-binding peptide ( named P7 ) inhibited proliferation of several colon cancer cell lines including HT-29 , LoVo , and Caco2 cells stimulated by bFGF .\n\nFurther investigations with HT-29 cells indicated that P7 arrested the cell cycle at the G0/G1 phase of bFGF-stimulated cells , reduced the levels of phospho-Erk1/Erk2 induced by bFGF , and caused significant changes in the expression of proteins related to proliferation , cell cycle , and cancer .\n\nOur results suggested that the bFGF-binding peptide has a potential antitumor effect on colon cancer .", "output": "Sustaining proliferative signaling" }, { "input": "Cantharidin is an active constituent of mylabris , a traditional Chinese medicine .\n\nIt is a potent and selective inhibitor of protein phosphatase 2A ( PP2A ) that plays an important role in control of cell cycle , apoptosis , and cell-fate determination .\n\nOwing to its antitumor activity , cantharidin has been frequently used in clinical practice .\n\nIn the present study , we investigated the therapeutic potential of cantharidin in pancreatic cancer .\n\nCantharidin efficiently inhibited the growth of pancreatic cancer cells , but presented a much lighter toxicity effect against normal pancreatic duct cells .\n\nIt caused G2/M cell-cycle arrest that was accompanied by the down-regulation of cyclin-dependent kinase 1 ( CDK1 ) and up-regulation of p21 expression .\n\nIt induced apoptosis and elevated the expressions of pro-apoptotic factors tumor necrosis factor-alpha ( TNF-alpha ) , TNF-related apoptosis inducing receptor 1 ( TRAILR1 ) , TRAILR2 , Bad , Bak , and Bid , and decreased the expression of anti-apoptotic Bcl-2 .\n\nActivation of caspase-8 and caspase-9 suggested that both extrinsic and intrinsic pathways are involved in the induction of apoptosis .\n\nInterestingly , unlike previous studies on other cancer cells , we found that the inhibitory role of cantharidin is independent of oxidative stress in pancreatic cancer cells .\n\nMitogen-activated protein kinases ( MAPKs ) , including ERK , JNK , and p38 , were activated after treatment with cantharidin .\n\nInhibition of JNK , but not ERK or p38 , alleviated the cytotoxity effect of cantharidin , suggesting cantharidin exerted its anticancer effect through the JNK-dependent way .\n\nHence , in addition to being an attractive candidate compound with therapeutic potential , cantharidin also highlighted the possibility of using PP2A as a therapeutic target for pancreatic cancer treatment .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "\" Reactive \" or activated stroma characterizes many malignancies including breast cancers .\n\nRecently , we isolated a reactive mouse mammary gland stromal cell line called BJ3Z .\n\nThese cells express alpha-smooth muscle actin and stromal cell-derived factor 1 ( SDF-1 ) and are tumorigenic when injected into mice .\n\nHere we show that , in vivo , BJ3Z cells influence the angiogenesis and proliferation of xenografted estrogen receptor ( ER)-positive MCF-7 human breast cancer cell-derived solid tumors .\n\nThe growth-promoting effects of BJ3Z cells are equivalent to those of estradiol ( E(2) ) .\n\nBJ3Z cells also increase the proliferation of normal mouse mammary luminal cells adjacent to tumors .\n\nIn vitro , BJ3Z cells reorganize and increase the proliferation of cocultured malignant MCF-7 and normal human breast MCF10A cells grown as organoids in three-dimensional culture .\n\nThe effects of BJ3Z cells on MCF-7 cells are equivalent to those of E(2) .\n\nIn contrast , BJ3Z cells do not alter the growth of highly aggressive ER-negative MDA-MB-231 human breast cancer cells .\n\nWe show that BJ3Z cells secrete vascular endothelial growth factor ( VEGF ) .\n\nThe growth of MCF-7 organoids induced by BJ3Z can be inhibited by antagonists of VEGF and SDF-1 .\n\nConversely , recombinant VEGF stimulates the proliferation of MCF-7 , but not MDA-MB-231 , organoids .\n\nWe conclude that , in addition to angiogenesis , VEGF released by activated stroma increases the growth of ER-positive malignant epithelial cells and of adjacent normal epithelium .\n\nBecause activated stroma can substitute for E(2) and fosters hormone-independent growth of ER-positive tumors , we suggest that breast cancers exhibiting intrinsic hormone resistance may respond to antiangiogenic therapies .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Resveratrol is a naturally occurring polyphenol that exhibits pleiotropic health beneficial effects , including anti-inflammatory , cardio-protective , and cancer-protective activities .\n\nIt is recognized as one of the more promising natural molecules in the prevention and treatment of chronic inflammatory and autoimmune disorders .\n\nUlcerative colitis is an idiopathic , chronic inflammatory disease of the colon associated with a high colon cancer risk .\n\nHere , we used a dextran sulfate sodium ( DSS ) mouse model of colitis , which resembles human ulcerative colitis pathology .\n\nResveratrol mixed in food ameliorates DSS-induced colitis in mice in a dose-dependent manner .\n\nResveratrol significantly improves inflammation score , downregulates the percentage of neutrophils in the mesenteric lymph nodes and lamina propria , and modulates CD3(+) T cells that express tumor necrosis factor-alpha and IFN-gamma .\n\nMarkers of inflammation and inflammatory stress ( p53 and p53-phospho-Ser(15) ) are also downregulated by resveratrol .\n\nBecause chronic colitis drives colon cancer risk , we carried out experiments to determine the chemopreventive properties of resveratrol .\n\nTumor incidence is reduced from 80% in mice treated with azoxymethane ( AOM ) + DSS to 20% in mice treated with AOM + DSS + resveratrol ( 300 ppm ) .\n\nTumor multiplicity also decreased with resveratrol treatment .\n\nAOM + DSS-treated mice had 2.4 +/- 0.7 tumors per animal compared with AOM + DSS + 300 ppm resveratrol , which had 0.2 +/- 0.13 tumors per animal .\n\nThe current study indicates that resveratrol is a useful , nontoxic complementary and alternative strategy to abate colitis and potentially colon cancer associated with colitis .", "output": "Tumor promoting inflammation" }, { "input": "In the present study , we investigated if the intracellular Cl(-) affects cell growth and cell cycle progression of androgen-independent prostate cancer PC3 cells .\n\nPC3 cells cultured in a medium containing 113 mM Cl(-) for 96 h grew up 9-fold in cell number , while PC3 cells cultured in an 8 mM-Cl(-)-containing culture medium showed complete arrest of cell growth even after culture for 96 h .\n\nExposure of cells to the 8 mM-Cl(-) culture medium diminished phosphorylation levels of Rb and cdc2 , which are respectively key accelerators of transition from G(1) to S phase and G(2) to M phase in cell cycle progression .\n\nCulturing cells in the 8 mM-Cl(-)-containing culture medium upregulated the protein expression level of p21 ( a CDK inhibitor ) inhibiting transition of G(1) to S phase , and diminished the incorporation of 5-ethynyl-2'-deoxyuridine ( EdU ; a thymidine analogue ) into DNA .\n\nThese results suggest that cells cultured in the low Cl(-) medium prolonged the duration of all phases of the cell cycle ( G(1) , S , and G(2)/M ) , thereby abolishing overall cell cycle progression .\n\nEffects of culturing cells in the low Cl(-) culture medium on cell cycle progression would be mediated via a change in the intracellular Cl(-) concentration ( [ Cl(-)](i) ) , since [ Cl(-)](i) was decreased under a low Cl(-) culture medium .\n\nTo clarify this possibility , we studied effects of furosemide and bumetanide , Na+/K+/2Cl(-) cotransporter ( NKCC ) inhibitors , on proliferation of PC3 cells .\n\nFurosemide and bumetanide decreased [ Cl(-)](i) and cell growth of PC3 cells .\n\nThese results suggest that a change in [ Cl(-)](i) would play a critical role in this growth mechanism .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "BACKGROUND The current staging system provides an anatomical classification of lung tumors ; its secondary purpose is to allow the prognostic stratification of patients into homogeneous groups after surgery .\n\nIn this work , intratumoral perineural invasion , lymphatic and blood vessel invasion together with the necrosis content of the tumor exclusive of the non-small cell cancer staging system were studied .\n\nMETHODS During a 4-year period , 152 patients operated for non-small cell lung cancer ( NSCLC ) at our hospital were analyzed .\n\nMean age of patients was 55.7 +/- 10.1 years .\n\nRESULTS Overall 5-year survival was 42.2 % .\n\nMediastinal lymph node involvement , tumor size , incomplete resection , pneumonectomy , presence of necrosis and perineural invasion were significant prognosticators ( P = 0.03 , 0.04 , 0.0001 , 0.046 , 0.0246 , < 0.0001 , respectively ) .\n\nMultivariate analysis revealed that N status , perineural invasion , and the presence of necrosis were independent prognostic factors ( P = 0.006 , P = 0.001 , P = 0.001 , respectively ) .\n\nPatients who had stage I tumor with necrosis and perineural invasion had a lower survival rate than those with stage IIIA tumor without these histopathological features ( P = 0.04 ) .\n\nThe presence of these histopathological characteristics in stage IIIA patients was a sign of a poorer prognosis ( P = 0.0001 ) .\n\nCONCLUSIONS Perineural invasion and the presence of necrosis independently indicated a dismal prognosis and their prognostic power is comparable to those of the TNM classification .\n\nThese factors could be candidates for better survival stratification and the indicators of the need for adjuvant therapy in early stage lung cancer patients .", "output": "Resisting cell death, Activating invasion and metastasis" }, { "input": "It has been shown that injecting a suspension of IFN-\u03b3-secreting tumor cells results in their rejection .\n\nThis effect has been attributed to IFN-\u03b3 preventing tumor stroma formation but not to a direct effect on the cancer cells .\n\nHowever , it is not known , which influence IFN-\u03b3 has on tumors with an established stroma .\n\nTo address this question , the plasmacytoma cell line J558L was transduced with a vector allowing doxycycline-inducible IFN-\u03b3 gene expression .\n\nAfter the injection of the tumor cells into mice , IFN-\u03b3 was induced at different time points .\n\nTumors did not grow when inducing IFN-\u03b3 immediately after tumor cell inoculation , while approximately half of the tumors were rejected when IFN-\u03b3 was induced in early established tumors within 2 weeks .\n\nInduction of IFN-\u03b3 2-3 weeks after tumor cell inoculation was less efficient ( 0-17% rejection ) .\n\nIFN-\u03b3 induction in established tumors led to a reduction of CD146(+) endothelial cells and massive necrosis .\n\nTogether , we show that vascularized tumors can be rejected by local IFN-\u03b3 expression , but that rejection of established tumors was less efficient over time .\n\nThis suggests that transplanted tumors became less susceptible to local IFN-\u03b3 treatment the better they are established .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "Palmitoylation is required for the activities of several cancer-associated proteins , making the palmitoyl acyltransferase ( PAT ) enzymes that catalyze these reactions potential targets for anticancer therapeutics .\n\nIn this study , we sought to identify and characterize a human PAT with activity toward N-terminally myristoylated and palmitoylated proteins .\n\nNIH/3t3 cells were stably transfected with vectors containing no insert , wild type human DHHC20 , or a serine-substituted DHHS20 mutant .\n\nCompared with control cells , cells overexpressing wild-type DHHC20 displayed an increase in palmitoylation activity toward a peptide that mimics the N-terminus of myristoylated and palmitoylated proteins , but had no change in activity toward a peptide that mimics the C-terminus of farnesylated and palmitoylated proteins .\n\nCells expressing DHHS20 had no significant change in activity toward either peptide .\n\nOverexpression of DHHC20 also caused phenotypic changes consistent with cellular transformation , including colony formation in soft agar , decreased contact inhibition of growth , and increased proliferation under low-serum conditions .\n\nQuantitative polymerase chain reaction analyses of human tissues demonstrated that DHHC20 is expressed in a tissue-specific manner , and is overexpressed in several types of human tumors , including ovarian , breast and prostate .\n\nOverall , these results demonstrate that DHHC20 is a human N-terminal-myristoyl-directed PAT involved in cellular transformation , that may play a role in cancer .", "output": "Evading growth suppressors" }, { "input": "PURPOSE Pyruvate kinase isoenzyme M2 ( PKM2 ) is a key enzyme in aerobic glycolysis ; inhibition of PKM2 leads to the tumor growth inhibition .\n\nIn this study , the effects of combined treatment with cisplatin ( DDP ) and a plasmid that expresses a short hairpin RNA ( shRNA ) targeting PKM2 on the growth of human A549 xenograft lung cancer model were investigated .\n\nMETHODS The expression of PKM2 in A549 cells was determined by immunofluorescence .\n\nPKM2 expression levels were evaluated by Western blot analysis .\n\nIn a human A549 lung cancer xenograft model , the effects of treatment with shRNA , with or without cisplatin , on tumor volume were determined .\n\nApoptosis and cell proliferation status were examined to determine the mechanisms of tumor growth inhibition .\n\nRESULTS Expression of shRNA targeting PKM2 resulted in inhibition of PKM2 expression in A549 cells .\n\nIn the lung cancer xenograft model , average tumor volume in the group treated with both cisplatin and shRNA was statistically lower than those of other groups ( P < 0.05 ) .\n\nThe levels of apoptotic cells were significantly higher in samples from animals in the combined treatment group than those from untreated animals ( P < 0.05 ) .\n\nThe cell proliferation rate , as determined by counting cells labeled with an anti-phospho-histone H3 , a marker for mitosis , was lower in samples from animals treated with both cisplatin and shRNA than in samples from other groups ( P < 0.05 ) .\n\nCONCLUSIONS Use of RNA interfering ( RNAi ) targeting PKM2 significantly inhibited tumor growth when combined with cisplatin in a human A549 lung cancer xenograft model .\n\nThe enhanced antitumor activity of the combined treatment compared to treatment with shRNA alone may result in part from increased induction of apoptosis and augmented inhibition of cancer cell proliferation .", "output": "Resisting cell death" }, { "input": "Certain hexavalent chromium ( Cr(VI) ) compounds are well established occupational respiratory tract carcinogens .\n\nHowever , despite extensive studies , the cellular and molecular mechanisms underlying Cr(VI)-induced lung cancer remain poorly understood .\n\nIn fact , the models used were often suboptimal and yielded conflicting results that were heavily dependent upon the system and experimental conditions employed .\n\nHere , we investigated the effects of chronic subcytotoxic and mildly cytotoxic ( 0.1-2 microM ) Cr(VI) exposures on cultures of human bronchial epithelial cells , the main targets of Cr(VI) carcinogenicity .\n\nOur studies with the nontumorigenic BEAS-2B cell line suggest that relatively short exposures ( h ) to sublethal Cr(VI) doses ( 0.1-1 microM ) may render these cells less sensitive to contact inhibition .\n\nWe have also observed a reduced sensitivity to Cr(VI)-induced apoptosis shortly after the beginning of exposure to a mildly cytotoxic Cr(VI) dose ( 2 microM ) .\n\nFurther studies are needed to determine whether these two phenotypes are involved in the Cr(VI)-induced carcinogenic process .\n\nAdditionally , evidence gathered in this study strongly points to a Cr(VI) interference with cell adhesion to the substratum and with cell-cell interactions .\n\nFinally , by chronically exposing BEAS-2B cells to mildly cytotoxic Cr(VI) doses ( 1 and 2 microM ) , we were able to induce changes in cell morphology and pattern of growth characteristic of an early phase of pre-malignant progression .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "To investigate the molecular mechanisms underlying altered T cell response in renal cell carcinoma ( RCC ) patients , we compared autologous and allogeneic CD8(+) T cell responses against RCC line from RCC patients and their HLA-matched donors , using mixed lymphocyte/tumor cell cultures ( MLTCs ) .\n\nIn addition , we analyzed the expression of molecules associated with cell cycle regulation .\n\nAutologous MLTC responder CD8(+) T cells showed cytotoxic activity against RCC cell lines ; however the analysis of the distribution of CD8(+) T-cell subsets revealed that allogenic counterparts mediate superior antitumor efficacy .\n\nIn RCC patients , a decreased proliferative response to tumor , associated with defects in JAK3/STAT5/6 expression that led to increased p27KIP1 expression and alterations in the cell cycle , was observed .\n\nThese data define a molecular pathway involved in cell cycle regulation that is associated with the dysfunction of tumor-specific CD8(+) effector cells .\n\nIf validated , this may define a therapeutic target in the setting of patients with RCC .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Currently , there are no satisfactory biomarkers available to screen for nasopharyngeal carcinoma ( NPC ) .\n\nNitric oxide ( NO ) , produced by inducible nitric oxide synthase ( iNOS ) , has been suggested to cause nitrative and oxidative stress , leading to the accumulation of 8-nitroguanine ( 8-NitroG ) and 8-hydroxy-2'-deoxyguanosine ( 8-OHdG ) and the subsequent transversion mutation of DNA .\n\nThe aim of this study was to evaluate iNOS expression and the status of nitrative and oxidative stress in NPC .\n\nFifty-nine cases of NPC and 39 cases of chronic nasopharyngitis were investigated to examine the expression of iNOS and the formation of 8-NitroG and 8-OHdG , using double-immunofluorescent staining .\n\nThe statistical differences in immunoreactivities were analyzed using the Mann-Whitney test .\n\nThirty-six patients from the 57 cases of NPC and 36 healthy controls were investigated to examine the level of serum 8-OHdG , using enzyme-linked immunosorbent assay ( ELISA ) .\n\nThe statistical differences were analyzed using a t test .\n\nStrong DNA lesions were observed in the cancer cells of NPC patients .\n\nAll cases of NPC were positive for 8-NitroG and 8-OHdG , and 54 ( 94.7% ) were positive for iNOS .\n\nNPC samples exhibited significantly more intense staining for 8-NitroG , 8-OHdG and iNOS than those of chronic nasopharyngitis ( P < 0.05 , respectively ) .\n\nThe mean value of serum 8-OHdG in the 36 NPC patients was 0.538 \u00b1 0.336 ng/ml compared to 0.069 \u00b1 0.059 ng/ml for the healthy controls .\n\nThe difference in the serum levels of 8-OHdG between the NPC patients and controls was statistically significant ( P < 0.05 ) .\n\nOur present findings suggest that pathological stimulation of nasopharyngeal tissue , caused by bacterial , viral or parasitic inflammation , may lead to nitrative and oxidative DNA lesions , caused by NO .\n\nThis may contribute to the cause and development of NPC .\n\nThus , 8-NitroG and 8-OHdG could be potential biomarkers for evaluating the risk of NPC .\n\nBetter understanding of the molecular mechanisms underlying nitrative and oxidative DNA damage may provide clues to molecular targets for new approaches of NPC prevention .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "This research aims to give a new insight to the relationship between host local immune response and the biological behaviour of the tumor by evaluating of intratumoral natural killer ( NK ) cells and tumor necrosis factor-alpha ( TNFalpha ) expressions in oral squamous cell carcinomas .\n\nNew paraffin sections of the deepest parts of the 46 cases of oral squamous cell carcinomas were immunohistochemically treated by CD57 , selected as NK cell indicator , and TNFalpha monoclonal antibodies .\n\nThe tumors were graded according to histopathologic grading scores of invasive margins and categorized into 2 groups as \" good \" and \" poor \" prognostic groups .\n\nFifteen cases , from which could be obtained full clinical data , were clinically staged and because of the scarcity of the cases in each group were divided , again , two groups as group 1 : stage I+stage II and group 2 : stage III+stage IV .\n\nThe expression levels of CD57 and TNFalpha were evaluated according to histopathologic grading groups and clinical staging groups .\n\nThe results showed that the density of CD57+cells ( NK cells ) was statistically lower in tumors graded as poor prognostic group compared to the cases in good prognostic group .\n\nOn the contrary , expression level of TNFalpha was statistically higher in poor prognostic group .\n\nThese findings suggested that increased secretion of TNFalpha in the tumors , which show high invasive potential , may be one of the facilitating factors for tumor invasion and be responsible from suppression of NK cells .\n\nWithdrawal of NK cells in the high invasive tumor areas also reminds the necessity of certain shared genetic rearrangements in tumor cells for getting protected from NK cell attacks and moving ahead within the extracellular matrix .", "output": "Activating invasion and metastasis" }, { "input": "We report that all-trans retinoic acid ( ATRA ) in combination with zoledronic acid has strong synergistic cytotoxic and apoptotic effects against human hormone- and drug-refractory prostate cancer cells , PC-3 and DU-145 , in a time- and dose-dependent manner .\n\nWe further investigated the effect of the combination treatment on the apoptotic process by both oligoarray and protein array analysis in DU-145 cells , in which the drug combination shows much more strong synergistic effects , as compared to PC-3 cells .\n\nMoreover , we have also performed real time-PCR array analysis to validate oligoarray results .\n\nWe demonstrated that the combination of ATRA and zoledronic acid is a strong inducer of apoptotic related cell death in human androgen-and drug refractory prostate cancer cells DU-145 , at either transcriptional or translational levels .\n\nWhile expression of proapoptotic genes such as tumor necrosis factor receptor superfamily ( TNFRSF ) , Bad , Bax , Fas , FADD are induced with the exposure of the combination , expression of antiapoptotic genes or proteins such as members of inhibitor apoptosis family ( IAPs ) , MCL-1 , LTBR , p53 and bcl-2 are reduced .\n\nBecause this novel combination treatment has fewer side effects than is generally the case with conventional cytotoxic agents , this regimen may be a good option for treatment of elderly prostate cancer patients .", "output": "Resisting cell death" }, { "input": "The transcription factor NF-kappaB is constitutively active in pancreatic adenocarcinoma .\n\nHere we explore the contribution of NF-kappaB to the malignant phenotype of pancreatic cancer cells in addition to its anti-apoptotic role .\n\nBlock of NF-kappaB signalling by non-destructible IkappaBalpha rendered cells resistant to TGF-beta-induced epithelial-mesenchymal transition ( EMT ) .\n\nIn contrast , NF-kappaB activation by TNF-alpha or expression of constitutively active IKK2 induced an EMT-phenotype with up-regulation of vimentin and ZEB1 , and down-regulation of E-cadherin .\n\nEMT could also be induced in cells with defective TGF-beta signalling .\n\nFunctional assays demonstrated reduced or strongly enhanced migration and invasion upon NF-kappaB inhibition or activation , respectively .", "output": "Activating invasion and metastasis" }, { "input": "Dendritic cells ( DCs ) are key regulators of innate and acquired immunity .\n\nThe maturation of DCs is directed by signal transduction events downstream of toll-like receptors ( TLRs ) and other pattern recognition receptors .\n\nHere , we demonstrate that , in mouse DCs , TLR agonists stimulate a profound metabolic transition to aerobic glycolysis , similar to the Warburg metabolism displayed by cancer cells .\n\nThis metabolic switch depends on the phosphatidyl inositol 3'-kinase/Akt pathway , is antagonized by the adenosine monophosphate ( AMP)-activated protein kinase ( AMPK ) , and is required for DC maturation .\n\nThe metabolic switch induced by DC activation is antagonized by the antiinflammatory cytokine interleukin-10 .\n\nOur data pinpoint TLR-mediated metabolic conversion as essential for DC maturation and function and reveal it as a potential target for intervention in the control of excessive inflammation and inappropriately regulated immune responses .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "We have investigated the possibility that photoexcited titanium dioxide ( TiO2 ) could inhibit the growth of malignant cells .\n\nWe studied the anti-glioma effects of nano-TiO2 excited with ultraviolet A ( UVA ) irradiation both in vitro and in vivo .\n\nTransmission electron microscopy demonstrated that glioma cells take up TiO2 by phagocytosis , and vital staining revealed that TiO2 alone has no effect on glioma cell proliferation .\n\nHowever , if TiO2 was combined with UVA irradiation the proliferation rate was decreased significantly compared to controls ( P<0.05 ) .\n\nRT-PCR suggested that TiO2 induction of glioma cell apoptosis is associated with changes in the expression of genes encoding Bcl-2 family members .\n\nWe then investigated the in vivo antitumor effects of combined TiO2 plus UVA treatment of established glioma tumors .\n\nTiO2 plus UVA led to pronounced areas of necrosis , elevated indices of apoptosis , delayed tumor growth , and increased survival compared with the TiO2-alone control group ( P<0.001 ) .\n\nLog-rank survival analysis showed that median survival duration was prolonged ( P<0.001 ) .\n\nThese findings suggest that nano-TiO2 based photodynamic therapy has potential in the treatment of glioma .", "output": "Resisting cell death" }, { "input": "OBJECTIVE To study the effect of Zilongjin ( ZLJ , a composite Chinese drug ) on proliferation and apoptosis of human breast carcinoma cell line MCF-7 .\n\nMETHODS MCF-7 cells were randomly divided into four groups depending on the culture solution used , the control group , cultured with 1640 medium not contained ZLJ ; and the three ZJL groups cultured with medium contained low ( 1.5 mg/mL ) , moderate ( 3 mg/mL ) and high ( 6 mg/mL ) dosage of ZLJ crude drug respectively .\n\nThe changes of cell proliferation were assessed by cell growth curve assay and methyl thiazolyl tetrazolium ( MTT ) assay .\n\nAnd the cell apoptosis was analyzed by flow cytometry , Hoechst 33342 staining and DNA ladder assay .\n\nRESULTS Compared with that in the normal control , the counts of cells in the three ZLJ groups were decreased significantly ( P<0.05 ) at such time point as 24 , 48 , 72 , 96 , 120 , and 144 h .\n\nFurthermore , apart from the comparison of the growth inhibition rate between the low and moderate dosage group at 24 and 72 h which were found to be no significant difference ( P>0.05 ) , the comparison f that among the three ZLJ groups appeared to be significant difference ( P<0.05 ) .\n\nThe inhibitory effect of ZLJ on cell proliferation of MCF-7 was time- and dose-dependent ; it could retard cells in G0/G1 cell phase ; apoptosis of MCF-7 cell was induced by moderate and high dosage of ZLJ with revealing of apoptotic body and DNA ladder formation .\n\nCONCLUSION ZLJ shows cell proliferation inhibitory and apoptosis inducing effects on human breast cancer cell line MCF-7 , and thus to realize its anti-tumor action .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Mouse models can be useful for increasing the understanding of lung tumorigenesis and assessing the potential of chemopreventive agents .\n\nWe explored the role of inflammation in lung tumor development in mice with knockout of the tumor suppressor Gprc5a .\n\nExamination of normal lung tissue and tumors from 51 Gprc5a(+/+) ( adenoma incidence , 9.8% ; adenocarcinoma , 0% ) and 38 Gprc5a(-/-) mice ( adenoma , 63% ; adenocarcinoma , 21% ) revealed macrophage infiltration into lungs of 45% of the Gprc5a(-/-) mice and 8% of Gprc5a(+/+) mice and the direct association of macrophages with 42% of adenomas and 88% of adenocarcinomas in the knockout mice .\n\nGprc5a(-/-) mouse lungs contained higher constitutive levels of proinflammatory cytokines and chemokines and were more sensitive than lungs of Gprc5a(+/+) mice to stimulation of NF-kappaB activation by lipopolysaccharide in vivo .\n\nStudies with epithelial cells cultured from tracheas of Gprc5a(-/-) and Gprc5a(+/+) mice revealed that Gprc5a loss is associated with increased cell proliferation , resistance to cell death in suspension , and increased basal , tumor necrosis factor alpha-induced , and lipopolysaccharide-induced NF-kappaB activation , which were reversed partially in Gprc5a(-/-) adenocarcinoma cells by reexpression of Gprc5a .\n\nCompared with Gprc5a(+/+) cells , the Gprc5a(-/-) cells produced higher levels of chemokines and cytokines and their conditioned medium induced more extensive macrophage migration .\n\nSilencing Gprc5a and the p65 subunit of NF-kappaB in Gprc5a(+/+) and Gprc5a(-/-) cells , respectively , reversed these effects .\n\nThus , Gprc5a loss enhances NF-kappaB activation in lung epithelial cells , leading to increased autocrine and paracrine interactions , cell autonomy , and enhanced inflammation , which may synergize in the creation of a tumor-promoting microenvironment .", "output": "Tumor promoting inflammation" }, { "input": "The c-Jun NH(2)-terminal kinase ( JNK ) signaling cascade has been implicated in a wide range of diseases , including cancer .\n\nIt is unclear how different JNK proteins contribute to human cancer .\n\nHere , we report that JNK2 is activated in more than 70% of human squamous cell carcinoma ( SCC ) samples and that inhibition of JNK2 pharmacologically or genetically impairs tumorigenesis of human SCC cells .\n\nMost importantly , JNK2 , but not JNK1 , is sufficient to couple with oncogenic Ras to transform primary human epidermal cells into malignancy with features of SCC .\n\nJNK2 prevents Ras-induced cell senescence and growth arrest by reducing the expression levels of the cell cycle inhibitor p16 and the activation of NF-kappaB .\n\nOn the other hand , JNK , along with phosphoinositide 3-kinase , is essential for Ras-induced glycolysis , an energy-producing process known to benefit cancer growth .\n\nThese data indicate that JNK2 collaborates with other oncogenes , such as Ras , at multiple molecular levels to promote tumorigenesis and hence represents a promising therapeutic target for cancer .", "output": "Enabling replicative immortality, Cellular energetics" }, { "input": "Chronic inflammation , increased reactivity to self-antigens and incidences of cancer are hallmarks of aging .\n\nHowever , the underlying mechanisms are not well understood .\n\nAge-associated alterations in the DNA either due to oxidative damage , defects in DNA repair or epigenetic modifications such as methylation that lead to mutations and changes in the expression of genes are thought to be partially responsible .\n\nHere we report that epigenetic modifications in aged DNA also increase its immunogenicity rendering it more reactive to innate immune system cells such as the dendritic cells .\n\nWe observed increased upregulation of costimulatory molecules as well as enhanced secretion of IFN-alpha from dendritic cells in response to DNA from aged donors as compared to DNA from young donors when it was delivered intracellularly via Lipofectamine .\n\nInvestigations into the mechanisms revealed that DNA from aged subjects is not degraded , neither is it more damaged compared to DNA from young subjects .\n\nHowever , there is significantly decreased global level of methylation suggesting that age-associated hypomethylation of the DNA may be the cause of its increased immunogenicity .\n\nIncreased immunogenicity of self DNA may thus be another mechanism that may contribute to the increase in age-associated chronic inflammation , autoimmunity and cancer .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "Tumor necrosis factor related apoptosis-inducing ligand ( TRAIL ) and agonistic anti-DR4/TRAIL-R1 and anti-DR5/TRAIL-R2 antibodies are currently under clinical investigation for treatment of different malignancies .\n\nTRAIL activates DR4 and DR5 and thereby triggers apoptotic and non-apoptotic signaling pathways , but possible different roles of DR4 or DR5 in these responses has poorly been addressed so far .\n\nIn the present work , we analyzed cell viability , DISC formation as well as IL-8 and NF-kappaB activation side by side in responses to TRAIL and agonistic antibodies against DR4 ( mapatumumab ) and against DR5 ( lexatumumab ) in pancreatic ductal adenocarcinoma cells .\n\nWe found that all three reagents are able to activate cell death and pro-inflammatory signaling .\n\nDeath-inducing signaling complex ( DISC ) analysis revealed that mapatumumab and lexatumumab induce formation of homocomplexes of either DR4 or DR5 , whereas TRAIL additionally stimulated the formation of heterocomplexes of both receptors .\n\nNotably , blocking of receptors using DR4- and DR5-specific Fab fragments indicated that TRAIL exerted its function predominantly via DR4 .\n\nInterestingly , inhibition of PKC by Goe6983 enabled DR5 to trigger apoptotic signaling in response to TRAIL and also strongly enhanced lexatumumab-mediated cell death .\n\nOur results suggest the existence of mechanisms that silence DR5 for TRAIL- but not for agonistic-antibody treatment .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Ataxia telangiectasia mutated ( ATM ) kinase is a central player in cellular response to DNA damage .\n\nPhosphorylation of the histone H2AX by ATM is required for the accumulation of repair proteins at the sites of double-strand breaks .\n\nRecently , it was reported that the histone acetyltransferase Tat interactive protein-60 ( IPP60 ) is required to acetylate ATM prior to its activation .\n\nThe RuvB-like proteins TIP48 and TIP49 are known to be necessary for the assembly and functional activity of the TIP60 acetyltransferase complex .\n\nIn the present communication , we investigated the requirements of IIP48 and IIP49 for ATM activation by monitoring the cell cycle distribution and H2AX phosphorylation after irradiation of IIP48- and IIP49-depleted cells .\n\nWe found that neither the cell cycle norgammay-H2AX were affected in IIP48- and IIP49-silenced cells , suggesting that the IIP60 chromatin modification complex is not engaged in DNA damage signaling upstream of ATM .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Autotaxin ( ATX ) is an extracellular lysophospholipase D that generates lysophosphatidic acid ( LPA ) from lysophosphatidylcholine ( LPC ) .\n\nBoth ATX and LPA have been shown to be involved in many cancers .\n\nHowever , the functional role of ATX and the regulation of ATX expression in human hepatocellular carcinoma ( HCC ) remain elusive .\n\nRESULTS In this study , ATX expression was evaluated in tissues from 38 human HCC and 10 normal control subjects .\n\nATX was detected mainly in tumor cells within tissue sections and its over-expression in HCC was specifically correlated with inflammation and liver cirrhosis .\n\nIn addition , ATX expression was examined in normal human hepatocytes and liver cancer cell lines .\n\nHepatoma Hep3B and Huh7 cells displayed stronger ATX expression than hepatoblastoma HepG2 cells and normal hepatocytes did .\n\nProinflammtory cytokine tumor necrosis factor alpha ( TNF-alpha ) promoted ATX expression and secretion selectively in Hep3B and Huh7 cells , which led to a corresponding increase in lysophospholipase-D activity .\n\nMoreover , we explored the mechanism governing the expression of ATX in hepatoma cells and established a critical role of nuclear factor-kappa B ( NF-kappaB ) in basal and TNF-alpha induced ATX expression .\n\nFurther study showed that secreted enzymatically active ATX stimulated Hep3B cell invasion .\n\nCONCLUSIONS This report highlights for the first time the clinical and biological evidence for the involvement of ATX in human HCC .\n\nOur observation that links the TNF-alpha/NF-kappaB axis and the ATX-LPA signaling pathway suggests that ATX is likely playing an important role in inflammation related liver tumorigenesis .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "BACKGROUND Paclitaxel and pirarubicin exhibit cytotoxic and antitumor activities .\n\nHowever , little is known about the apoptosis-inducing effects of paclitaxel and pirarubicin on human osteosarcoma MG-63 cells .\n\nMETHODS The effects of paclitaxel and pirarubicin on cell cycle arrest and apoptosis were studied in MG-63 cells using flow cytometry .\n\nPCNA , Bcl-2 , Bax , cyclin D1 and cyclin E expression was assessed by Western blotting .\n\nRESULTS Paclitaxel and pirarubicin caused G2/M and G0/G1 cell cycle arrest in MG-63 cells , respectively .\n\nApoptosis of MG-63 cells mediated by paclitaxel was dependent on treatment duration .\n\nInterestingly , in cells treated with pirarubicin , apoptosis was related to treatment duration at concentrations of 10(2)-10(3) nM , whereas the effect of treatment duration was less marked at concentrations >10(4)-10(5) nM .\n\nFurthermore , paclitaxel and pirarubicin suppressed the expression of PCNA , cyclin D1 , cyclin E and Bcl-2 , and increased Bax expression .\n\nCONCLUSION These results suggest that the G2/M or G0/G1 cell cycle arrest and apoptosis induced by paclitaxel and pirarubicin are Bcl-2/Bax dependent , suggesting favorable effects of combination therapy with paclitaxel and pirarubicin in the treatment of osteosarcoma .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Treatment for glioblastoma multiforme includes the alkylating agent temozolomide combined with ionizing radiation .\n\nPersistent O6-guanine methylation by temozolomide in O6-methylguanine methyl transferase negative tumors causes cytotoxic lesions recognized by DNA mismatch repair , triggering apoptosis .\n\nResistance ( intrinsic or acquired ) presents obstacles to successful temozolomide treatment , limiting drug efficacy and life expectancy .\n\nTwo glioma cell lines , SNB19 and U373 , sensitive to temozolomide ( GI(50) values 36 and 68 microM , respectively ) were exposed to increasing temozolomide concentrations ( 1-100 microM ) .\n\nVariant cell lines ( SNB19VR , U373VR ) were generated that displayed acquired temozolomide resistance ( GI(50) values 280 and 289 microM , respectively ) .\n\nCross-resistance to mitozolomide was observed in U373VR cells only .\n\nIn clonogenic and MTT assays , methylguanine methyltransferase ( MGMT ) depletion using O6-benzylguanine sensitized U373VR cells to temozolomide , indicating the resistance mechanism involves MGMT re-expression .\n\nIndeed , Western blot analyses revealed MGMT protein in cell lysates .\n\nIn SNB19VR cells , down-regulation of MSH6 message and protein expression may confer temozolomide tolerance .\n\nInhibition of poly(ADP-ribose) polymerase-1 ( a key base excision repair ( BER ) enzyme ) partially restored sensitivity , and DNA repair gene arrays demonstrated up-regulation ( >5-fold ) of BER gene NTL1 in SNB19VR cells .\n\nIn conclusion , we have developed two glioma cell lines whose distinct mechanisms of acquired resistance to temozolomide , involving expression of MGMT , or inactivation of DNA mismatch repair and recruitment of BER enzymes , are consistent with clinical observations .\n\nThese cell lines provide valuable models for the development of strategies to combat temozolomide resistance .", "output": "Genomic instability and mutation" }, { "input": "Epidemiologic studies have examined the association between fruit and vegetable ( F&V ) consumption and the risk of cancer .\n\nSeveral cancer-preventive mechanisms have been proposed , such as antioxidant properties and modulation of biotransformation enzyme activities ; both may be associated with reducing DNA damage and hence the mutation rate .\n\nWe investigated , in a randomized , controlled , crossover feeding trial , the effect of 10 servings/day of botanically defined F&V for 2 wk on endogenous DNA damage ; resistance to gamma -irradiation damage ; and DNA repair capacity in lymphocytes , measured by the Comet assay .\n\nWe also explored the association between the UGT1A1*28 polymorphism and serum bilirubin concentrations and DNA damage and repair measures .\n\nHealthy men ( n = 11 ) and women ( n = 17 ) , age 20 to 40 yr , provided blood samples at the end of each feeding period .\n\nOverall , F&V did not affect DNA damage and repair measures in lymphocytes .\n\nThe number of UGT1A1*28 alleles was inversely associated with sensitivity to gamma -irradiation exposure and DNA repair capacity , but a biological mechanism to explain this association is unclear .\n\nA larger sample size is needed to investigate the association between bilirubin concentrations and endogenous DNA damage .\n\nWith inconsistent findings in the literature , additional dietary intervention studies on the effect of F&V on DNA damage and repair are needed .", "output": "Genomic instability and mutation" }, { "input": "INTRODUCTION Astrocytic gliomas are the most common intracranial central nervous system neoplasias , accounting for about 60% of all primary central nervous system tumors .\n\nDespite advances in the treatment of gliomas , no effective therapeutic approach is yet available ; hence , the search for a more realistic model to generate more effective therapies is essential .\n\nOBJECTIVE To develop an experimental malignant astrocytoma model with the characteristics of the human tumor .\n\nMETHOD Primary cells from subcutaneous xenograft tumors produced with malignant astrocytoma U87MG cells were inoculated intracerebrally by stereotaxis into immunosuppressed ( athymic ) Rowett rats .\n\nRESULTS All four injected animals developed non-infiltrative tumors , although other glioblastoma characteristics , such as necrosis , pseudopalisading cells and intense mitotic activity , were observed .\n\nCONCLUSION A malignant astrocytoma intracerebral xenograft model with poorly invasive behavior was achieved in athymic Rowett rats .\n\nTumor invasiveness in an experimental animal model may depend on a combination of several factors , including the cell line used to induce tumor formation , the rat strains and the status of the animal's immune system .", "output": "Resisting cell death" }, { "input": "OBJECTIVE As we previously demonstrated , the inhibitory effect of iodine on thyroid cell growth is mediated by iodolactones , especially 6-iodo-5-hydroxy-eicosatrienoic acid ( delta-iodolactone ) .\n\nIn this communication we compare the effect of iodide , molecular iodine and delta-iodolactone on growth inhibition and apoptosis on three human thyroid carcinoma cell lines ( B-CPAP cells , FTC-133 cells and 8505C cells ) as well as on human breast cancer cells ( MCF 7 ) .\n\nMETHODS Thyroid carcinoma cells were cultured in Dulbecco's modified Eagle's medium ( DMEM ) and MCF 7 cells in Rowswell Park Memorial Institute ( RPMI ) culture medium , both containing 10% ( v/v ) Fetal Calf Serum ( FCS ) , until they were confluent .\n\nAround 2000 cells were then distributed in 12-well plates and grown for 48 h in either DMEM ( thyroid cancer cells ) or in RPMI medium ( MCF 7 cells ) both containing 5% FCS .\n\nThereafter , different concentrations of iodide , iodine or delta-iodolactone were added for 24 h .\n\nGrowth rate was estimated by cell counting in a Coulter Counter adapted for epithelial cells .\n\nApoptosis was determined by a mitochondrial potential assay .\n\nRESULTS The growth rate of B-CPAP cells was unaffected by iodide , but was reduced by high concentreations of molecular iodine ( 100 and 500 microM ) .\n\nHowever , delta-iodolactone significantly reduced cell proliferation already with low concentrations ( 5 microM and 10 microM ) and further in a dose-dependent manner up to 82% .\n\nFTC-133 and 8505C cells were unaffected by iodide , iodine or delta-iodolactone .\n\nIn contrast , in MCF 7 cells , molecular iodine ( 100 microM ) inhibited growth from 100% to 83% but delta-iodolactone ( 1 , 5 and 10 microM ) dose-dependently decreased growth rate from 100% to 82% and 62% , respectively .\n\nThe inhibition of growth was through apoptosis , and not necrosis , as the amount of apoptotic cells corresponded to the growth inhibition .\n\nCONCLUSION delta-Iotaodolactone seems to be the main iodocompound which can inhibit growth and induce apoptosis in B-CPAP cells as well as in MCF 7 breast cancer cells .", "output": "Resisting cell death" }, { "input": "NKX3.1 is a homeodomain protein that functions as a dosage sensitive prostate-specific transcription factor .\n\nDiminished NKX3.1 expression is associated with prostate epithelial cell proliferation in vitro and with increasing Gleason grade in patient samples .\n\nMouse Nkx3.1 also functions as a negative regulator of prostate cell growth in prostate cancer models .\n\nIdentifying biological and environmental factors that modulate NKX3.1 accumulation is therefore central to efforts aimed at elucidating prostate growth control mechanisms .\n\nTo determine the effect of inflammation on Nxk3.1 accumulation , bacterial prostatitis was induced by intraurethral inoculation of a uropathogenic E. coli strain in mice .\n\nNkx3.1 expression was profoundly reduced in infected prostate lobes and correlated with increased expression of a proliferation marker .\n\nAndrogen receptor levels were also reduced in concert with Nkx3.1 , and a marked increase in the basal cell marker p63 was observed .\n\nAnalyses of the inflammatory infiltrate revealed a classic acute inflammatory response that attained characteristics of a chronic state within fourteen days postinoculation .\n\nComparison of the four prostate lobes revealed clear differences in the extent of inflammation .\n\nThese data demonstrate that acute inflammation in response to a bacterial agent in the prostate is associated with a significant diminution in the level of a key regulator of prostate cell proliferation .\n\nThese observations provide a plausible mechanism whereby prostate inflammation may establish a local environment conducive to epithelial cell growth .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "OBJECTIVE To clarify the relationship between the expression of pol beta and DNA damage /repair induced by Benzo(a)pyrene ( BaP ) .\n\nMETHODS pol beta wild-type cells ( pol beta +/+ ) , pol beta null cells ( pol beta -/- ) and wild-type pol beta overexpressed cells ( pol beta oe ) which had the same genetic background were studied .\n\nFirstly , RT-PCR and Western blot targeting to pol beta were carried out to measure the expression of pol beta mRNA and protein in above three kinds of cells , then MTT test and single cell gel electrophoresis ( comet assay ) were used to compare cell viability and DNA damage/repair of the three kinds of cells when exposed to BaP .\n\nRESULTS There was pol beta deletion in pol beta -/- cells and the level of pol beta mRNA and protein in pol beta oe cells was twice higher than that in pol beta +/+ cells .\n\nBaP could induce DNA damage and reduce cell viability , when compared with pol beta +/+ cells , IC50 of pol beta -/- cells was remarkably lower , DNA was prone to damage and more difficult to be repaired , on the other hand , IC50 of pol beta oe cells was obviously higher and the damage effect on DNA was weaker and prone to be repaired .\n\nCONCLUSION Pol beta played an important role in the repair of DNA damage induced by BaP , deficiency of pol beta could decrease the DNA repair capability of cells , and overexpression of pol beta could help cells response to DNA damage and protect cells from death in a certain degree .", "output": "Genomic instability and mutation" }, { "input": "The tumor suppressor LKB1 is inactivated in 90% of Peutz-Jeghers cancer syndrome , 30-40% of non-small cell lung carcinoma , and a variety of other cancers , indicating the loss of LKB1 activity is a critical step in oncogenesis .\n\nHowever , current understanding of LKB1 function is largely limited to the results from cancer cells , and how LKB1 inactivation initiates malignant transformation in normal cells remains unclear .\n\nHere we ablated endogenous expression of LKB1 in two normal cell lines : human embryonic kidney 293T cells ( HEK-293T cells ) and human umbilical vein endothelial cells ( HUVECs ) by LKB1-specific short hairpin RNAs .\n\nDownregulation of endogenous LKB1 lead to a facilitated G(1)/S transition , accompanied by a concomitant increase in Rb phosphorylation ( Ser(807/811) ) .\n\nFurthermore , reduced expression of p53 and p16 was observed in LKB1 ablated cells , while no differences were detected for cyclin D1 and cyclin E. These results jointly suggest that endogenous LKB1 knockdown accelerates cell cycle progression through G(1)/S checkpoint in HEK-293T cells and HUVECs , which is at least in part , mediated by decline of p53 and p16 pathways .\n\nOur findings provided a plausible mechanism by which loss of LKB1 expression in normal cells contributes to the formation of malignancies .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Although contact inhibition is a fundamental process for multicellular organisms , how proliferation is inhibited at high cellular densities remains poorly characterized .\n\nHere we show that 4E-BP1 , one major repressor of cap-dependent translation , plays a critical role in density-mediated cell cycle arrest. 4E-BP1 promoter is activated and 4E-BP1 protein amount increases as cells reach confluence .\n\nConversely , a much less marked density-dependent inhibition of cell proliferation is observed upon 4E-BP1 silencing .\n\nWe further show that at high density , progression through the G\u2081 phase of the cell cycle is faster and Cyclin D1 protein is induced in different cell types where 4E-BP1 has been either downregulated ( stable shRNA expression or transient siRNA transfection ) or removed ( knock-out ) .\n\nThus 4E-BP1 appears as an important mediator of contact inhibition .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "MG132 , as a proteasome inhibitor , can induce apoptotic cell death through formation of reactive oxygen species ( ROS ) .\n\nIn this study , we investigated the effects of MAPK ( MEK , JNK , and p38 ) inhibitors on MG132-treated A549 lung cancer cells in relation to cell growth , cell death , ROS , and glutathione ( GSH ) levels .\n\nTreatment with 10 microM MG132 inhibited the growth of A549 cells at 24 h .\n\nMG132 also induced apoptosis , which was accompanied by the loss of mitochondrial membrane potential ( MMP ; deltapsi(m) ) .\n\nROS were not increased in MG132-treated A549 cells .\n\nMG132 increased GSH-depleted cell numbers and decreased GSH levels .\n\nMEK and JNK inhibitors did not strongly affect cell growth , cell death , ROS , and GSH levels in MG132-treated A549 cells .\n\nIn contrast , p38 inhibitor reduced cell growth inhibition , apoptosis , and MMP ( deltapsi(m) ) loss by MG132 .\n\nHowever , p38 inhibitor did not change ROS level and GSH content .\n\nIn conclusion , MG132 inhibited the growth of A549 cells via apoptosis without formation of ROS .\n\nTreatment with p38 inhibitor rescued some cells from MG132-induced apotposis , which was not affected by ROS and GSH level changes .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The development and progression of esophageal cancer is associated with multiple alterations in the genome , including loss of the tumor suppressor phosphatase and tensin homolog deleted from the chromosome 10 ( PTEN ) gene .\n\nThe purpose of this study was to determine the effects of adenovirus-mediated MMAC/PTEN expression on the growth and survival of human esophageal cancer cells in vitro and in vivo .\n\nWe found that compared to control cells , overexpression of PTEN significantly suppressed growth and induced apoptosis in esophageal cancer cell lines Eca-109 and TE-1 via downregulation of Bcl-2 expression and changes in cell-cycle progression .\n\nAdenovirus PTEN also inhibited the growth of subcutaneous tumor xenografts by significantly reducing tumor size in vivo .\n\nThus our results confirm the proposed functional role of MMAC/PTEN as a regulator of esophageal cancer progression in vivo and in vitro .\n\nPTEN might be an important biological marker and potential therapeutic target in the treatment of human esophageal cancer .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "OBJECTIVE To study the effect of anandamide ( AEA ) on necrosis in HepG2 cells and to explore the role of AEA in progression of liver cancer .\n\nMETHODS Localization of the fatty acid hydrolytic enzyme ( FAAH ) , cannabinoid receptors 1(CB1) and cannabinoid receptors 2 ( CB2 ) proteins was detected in L02 and HepG2 cells using immunofluorescence .\n\nL02 and HepG2 cells were treated with different concentrations of AEA and methyl-beta-cyclodextrin , and the rates of cells necrosis were examined by PI stain .\n\nMeanwhile , the expression levels of FAAH , CB1 and CB2 receptor proteins , as well as P38 mitogen-activated protein kinase ( p-P38 MAPK ) and c-Jun-NH2-terminal kinase ( p-JNK ) proteins , were analyzed by Western blot .\n\nRESULTS The FAAH , CB1 and CB2 receptor proteins were observed both in cytoplasm and on membrane in L02 and HepG2 cells .\n\nThe expression level of FAAH protein was higher in HepG2 than in L02 cells .\n\nThe expression level of CB1 receptor protein was very low in both L02 and HepG2 cells .\n\nThe expression level of CB2 receptor protein was high in both L02 and HepG2 cells .\n\nAEA treatment induced necrosis in HepG2 cells but not in L02 cells .\n\nMethyl-beta-cyclodextrin treatment prevented necrosis in HepG2 cells ( t = 3.702 ; 5.274 ; 3.503 , P less than 0.05 ) .\n\nThe expression patterns of FAAH , CB1 and CB2 receptor protein in L02 and HepG2 cells were confirmed by western blot , which were consistent with the immunofluorescence results .\n\nAEA treatment increased the levels of p-P38MAPK and p-JNK proteins in a dose-dependent manner in HepG2 cells ( F = 11.908 ; 26.054 , P less than 0.05 ) and the increase can be partially by prevented by MCD ( t = 2.801 ; t = 12.829 , P less than 0.05 ) .\n\nCONCLUSION AEA treatment induces necrosis in HepG2 cells via CB1 and CB2 receptors and lipid rafts .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Elevated levels of air pollution are associated with increased risk of lung cancer .\n\nParticulate matter ( PM ) contains transition metals that may potentiate neoplastic development through the induction of oxidative stress and inflammation , a lung cancer risk factor .\n\nVanadium pentoxide ( V2O5 ) is a component of PM derived from fuel combustion as well as a source of occupational exposure in humans .\n\nIn the current investigation we examined the influence of genetic background on susceptibility to V2O5-induced inflammation and evaluated whether V2O5 functions as a tumor promoter using a 2-stage ( initiation-promotion ) model of pulmonary neoplasia in mice .\n\nRESULTS A/J , BALB/cJ ( BALB ) , and C57BL/6J ( B6 ) mice were treated either with the initiator 3-methylcholanthrene ( MCA ; 10 microg/g ; i.p. ) or corn oil followed by 5 weekly aspirations of V2O5 or PBS and pulmonary tumors were enumerated 20 weeks following MCA treatment .\n\nSusceptibility to V2O5-induced pulmonary inflammation was assessed in bronchoalveolar lavage fluid ( BALF ) , and chemokines , transcription factor activity , and MAPK signaling were quantified in lung homogenates .\n\nWe found that treatment of animals with MCA followed by V2O5 promoted lung tumors in both A/J ( 10.3 +/- 0.9 tumors/mouse ) and BALB ( 2.2 +/- 0.36 ) mice significantly above that observed with MCA/PBS or V2O5 alone ( P < 0.05 ) .\n\nNo tumors were observed in the B6 mice in any of the experimental groups .\n\nMice sensitive to tumor promotion by V2O5 were also found to be more susceptible to V2O5-induced pulmonary inflammation and hyperpermeability ( A/J>BALB>B6 ) .\n\nDifferential strain responses in inflammation were positively associated with elevated levels of the chemokines KC and MCP-1 , higher NFkappaB and c-Fos binding activity , as well as sustained ERK1/2 activation in lung tissue .\n\nCONCLUSIONS In this study we demonstrate that V2O5 , an occupational and environmentally relevant metal oxide , functions as an in vivo lung tumor promoter among different inbred strains of mice .\n\nFurther , we identified a positive relationship between tumor promotion and susceptibility to V2O5-induced pulmonary inflammation .\n\nThese findings suggest that repeated exposures to V2O5 containing particles may augment lung carcinogenesis in susceptible individuals through oxidative stress mediated pathways .", "output": "Tumor promoting inflammation" }, { "input": "The mechanisms of formation of sister chromatid exchanges ( SCEs ) and chromosome aberrations following inhibition of poly(ADP-ribose) polymerase by 3-aminobenzamide were studied in Chinese hamster ovary cell lines deficient in different repair pathways .\n\nThe results confirm earlier findings that ( a ) the ' spontaneous ' SCEs are formed due to the incorporated BrdU in the DNA , ( b ) ' spontaneous ' and induced SCEs originate from different mechanisms , and ( c ) SCEs and chromatid exchanges are formed by different pathways .", "output": "Genomic instability and mutation" }, { "input": "Polycyclic aromatic hydrocarbons ( PAH ) such as dibenzo[a,l]pyrene ( DBP ) are wide-spread environmental pollutants most probably mutagenic and carcinogenic to humans .\n\nDetailed data on the cytogenetic effects of anti-11,12-dihydroxy-13,14-epoxy-11,12,13,14-tetrahydrodibenzo[a,l]pyrene ( DBPDE ) in mammalian cells are not available in the literature .\n\nThe aim of this study is to elucidate the mechanisms involved in the induction of chromosomal aberrations and sister chromatid exchanges ( SCEs ) by DBPDE in mammalian cells .\n\nIn order to achieve this a parental ( AA8 ) and different DNA repair-deficient Chinese hamster ovary cell lines such as UV4 , UV5 , UV61 ( nucleotide excision repair , NER ) , EM9 ( base excision repair , BER ) , irs1SF ( homologous recombination repair , HRR ) and V3-3 ( non-homologous end joining , NHEJ ) were used .\n\nThe most sensitive cell lines for DBPDE-induced chromosome aberrations were EM9 and irs1SF , while EM9 and V3-3 cell lines were the most sensitive in terms of SCEs induction .\n\nIt can be suggested that the BER pathway plays an important role in the repair of lesions induced by DBPDE , affecting both chromosomal aberrations and SCEs induction .\n\nMoreover , the HRR pathway seems to play a role in cellular resistance to DBPDE mainly in terms of chromosomal aberration induction while the NHEJ pathway takes part affecting only the induction of SCEs .", "output": "Genomic instability and mutation" }, { "input": "Bulky DNA adducts are considered a potential biomarker of cancer risk .\n\nIn this study , the association between various lifestyle , environmental , and genetic factors and the levels of bulky DNA adducts in peripheral leukocytes was examined in a study group nested within a population-based prospective Danish cohort .\n\nAt enrollment , blood samples were collected and information on lifestyle , including dietary and smoking habits , obtained .\n\nPreviously , bulky DNA adducts were measured in 245 individuals who developed lung cancer and 255 control members of the cohort .\n\nOf these 500 individuals , data on 375 individuals were included in this study , excluding 125 cases , which developed lung cancer within the first 3 yr after blood sampling .\n\nBulky DNA adduct levels were measured by 32P-postlabeling technique and polymorphisms in carcinogen metabolism and DNA repair genes were determined .\n\nPotential predictors of bulky DNA adduct levels were analyzed by univariate and multivariate regression analyses .\n\nWomen tended to have higher adduct levels than men .\n\nLiving in central Copenhagen and surface darkness of fried meat and fish were associated with quantitative higher adduct levels .\n\nNo significant associations were found between dietary factors or smoking and DNA adduct levels .\n\nFurther , the results showed no prominent associations between any of 12 genetic polymorphisms and adduct levels .\n\nOverall , our study showed only few associations between dietary , environmental , and genetic factors and levels of bulky DNA adducts measured in peripheral leukocytes in a general Danish population .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND AND AIM The importance of glycolysis in cancer cells is well documented .\n\nThe effects of inhibiting glycolysis using metabolic inhibitors iodoacetate ( IAA ) , an inhibitor of GAPDHase , and 3-bromopyruvate ( 3BP ) , an inhibitor of hexokinase-II , on survival and signaling of pancreatic cancer cells ( Panc-1 ) were investigated .\n\nMATERIALS AND METHODS Cellular survival was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide ( MTT ) assay .\n\nLactate dehydrogenase ( LDH ) assay was used to analyze the induced necrosis and protein levels were evaluated using Western blot analysis .\n\nRESULTS The results show that the inhibitors lowered cellular survival and increased cellular necrosis .\n\nMitogenic signaling pathways were affected by 3BP but not by IAA .\n\nCONCLUSION We conclude that there may be a cross-talk between signaling pathways and glycolysis in regulating pancreatic cancer cell survival and signaling .\n\nThus , a combination of agents that inhibit both energy production and cell signaling may provide a novel and effective approach to target pancreatic cancer effectively .", "output": "Cellular energetics, Resisting cell death" }, { "input": "A novel camptothecin derivative ( TLC388 ) with higher efficacy and reduced toxicity has been synthesized and tested as a novel chemoradiosensitizing agent .\n\nThis study investigated the mechanisms of the chemoradiosensitizing effects of TLC388 on H23 human non-small cell lung cancer ( NSCLC ) cells .\n\nUsing the TUNEL assay , a significantly higher percentage of apoptotic cells was observed in the group treated with TLC388 plus X-ray radiation than those in groups treated with drug or radiation alone .\n\nThe sensitizer enhancement ratio ( SER ) was 1.91 .\n\nApoptosis increased with drug concentration and radiation dose , exhibiting dose-dependent pattern .\n\nThe results suggested that apoptosis could be a main mode of cell death that might underlie the increased chemoradio-sensitization of TLC388 .\n\nTreatment with 30 nM of TLC388 plus 4 Gy X-ray also produced up to 42% of necrotic cells that were measured by trypan blue exclusion assay , but with TLC388 alone or 4 Gy radiation alone 9.8% or 11.1% necrotic cells were detected , respectively .\n\nAn immunofluorescent staining method was employed to determine the levels of gamma-H2AX ( phosphorylated H2AX , a variant of the H2A protein family , which is a component of the histone octomer in nucleosomes and is phosphorylated by kinases like ATM and ATR in the PI3K pathway , as the first step in recruiting and localizing DNA repair proteins ) as a molecular biomarker of DNA double strand breaks ( DSBs ) in cells treated with TLC388 +/-radiation , or radiation alone .\n\nThe formation of gamma-H2AX foci was observed after TLC388 or radiation exposure and when the cells were treated with 30 nM TLC388 plus radiation at a dose of 2 Gy , the percentage of cells containing gamma-H2AX foci increased significantly .\n\nEven more interesting , a markedly higher percentage ( 65.4% ) of mitotic cells displayed gamma-H2AX foci after treatment with 30 nM TLC388 plus 0.5 Gy radiation , compared to only 5.9% or 26.1% of the M-phase cells treated with 30 nM TLC388 alone or 0.5 Gy radiation alone , respectively .\n\nIt is suggested that mitotic cells become very sensitive to the production of DSBs after TLC388-radiation combined treatment and the formation of DSBs is strongly suggested to lead to the induction of apoptosis at doses lower than 4 Gy and to some necrosis at doses of 4 Gy or above .\n\nTLC388 enhances the production of DSBs and inhibits their repair , which contributes to the elucidation of the mechanisms of chemoradiosensitization of TLC388 and its development as a novel chemoradiosensitizing drug for improved radiotherapy .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "The hypoxic tumor microenvironment is associated with malignant progression and poor treatment response .\n\nThe glucose transporter Glut-1 is a prognostic factor and putative hypoxia marker .\n\nSo far , studies of Glut-1 in cancer have utilized conventional immunohistochemical analysis in a series of individual biopsy or surgical specimens .\n\nTissue microarrays , however , provide a rapid , inexpensive means of profiling biomarker expression .\n\nTo evaluate hypoxia markers , tissue cores must show the architectural features of hypoxia ; i.e. viable tissue surrounding necrotic regions .\n\nGlut-1 may be a useful biomarker to validate tissue microarrays for use in studies of hypoxia-regulated genes in cancer .\n\nIn this study , we carried out immunohistochemical detection of Glut-1 protein in many tumor and normal tissue types in a range of tissue microarrays .\n\nGlut-1 was frequently found in peri-necrotic regions , occurring in 9/34 lymphomas , 6/12 melanomas , and 5/16 glioblastomas ; and in 43/54 lung , 22/84 colon , and 23/60 ovarian tumors .\n\nExpression was rare in breast ( 6/40 ) and prostate ( 1/57 ) tumors , and in normal tissue , was restricted to spleen , tongue , and CNS endothelium .\n\nIn conclusion , tissue microarrays enable the observation of Glut-1 expression in peri-necrotic regions , which may be linked to hypoxia , and reflect previous studies showing differential Glut-1 expression across tumor types and non-malignant tissue .", "output": "Resisting cell death" }, { "input": "Obesity and related metabolic abnormalities are risk factors for colorectal cancer .\n\nA state of chronic inflammation and adipocytokine imbalance may play a role in colorectal carcinogenesis .\n\nStatins , which are commonly used for the treatment of hyperlipidemia , are known to possess anti-inflammatory effects .\n\nStatins also exert chemopreventive properties against various cancers .\n\nThe present study examined the effects of pitavastatin , a recently developed lipophilic statin , on the development of azoxymethane ( AOM)-initiated colonic premalignant lesions in C57BL/KsJ-db/db ( db/db ) obese mice .\n\nMale db/db mice were administrated weekly subcutaneous injections of AOM ( 15 mg/kg body weight ) for 4 weeks and then were subsequently fed a diet containing 1 ppm or 10 ppm pitavastatin for 8 weeks .\n\nFeeding with either dose of pitavastatin significantly reduced the number of colonic premalignant lesions , beta-catenin accumulated crypts , by inhibiting proliferation and the surrounding inflammation .\n\nPitavastatin increased the serum levels of adiponectin while conversely decreasing the serum levels of total cholesterol , tumor necrosis factor-alpha ( TNF-alpha ) , interleukin ( IL)-6 , IL-18 , and leptin .\n\nPitavastatin also caused a significant increase in the expression of phosphorylated form of the AMP-activated kinase ( AMPK ) protein on the colonic mucosa of AOM-treated mice .\n\nIn addition , the expression levels of TNF-alpha , IL-6 , IL-18 , and COX-2 mRNAs on the colonic mucosa of AOM-treated mice were decreased by treatment with this agent .\n\nThese findings suggest that pitavastatin attenuates chronic inflammation and improves the imbalance of adipocytokines , both of which are caused by the presence of excess adipose tissues , thereby preventing the development of colonic premalignancies in an obesity-related colon cancer model .\n\nTherefore , some types of statins , including pitavastatin , may be a useful chemoprevention modality for colon cancer in obese individuals .", "output": "Tumor promoting inflammation" }, { "input": "Thyroid-like low-grade nasopharyngeal papillary adenocarcinoma ( LGNPPA ) is extremely rare ; only four cases have been reported .\n\nHerein are presented the case reports of two Japanese male patients with thyroid-like LGNPPA .\n\nMacroscopically , these tumors were pedunculated polypoid masses on the roof of the nasopharynx .\n\nMicroscopically , they were characterized by papillary and glandular epithelial proliferation .\n\nThe papillae were complex and tightly packed with hyalinized fibrovascular cores and lined by columnar and pseudostratified cells with intervening spindle-shaped cells .\n\nBoth cell types had round to oval vesicular nuclei with tiny nucleoli and mildly eosinophilic cytoplasm .\n\nMitotic figures were not evident and necrosis was not observed .\n\nPsammoma bodies were seen focally in one of the patients .\n\nTransition from normal surface epithelium to tumor cells was identified in both cases .\n\nOn immunohistochemistry the tumor cells were positive for cytokeratin ( CK)7 , CK19 , thyroid transcription factor-1 ( TTF-1 ) and vimentin .\n\nThey were negative for CK5/6 , CK20 , thyroglobulin , S-100 protein and CD15 .\n\nIn situ hybridization for EBV was negative .\n\nNasopharyngeal tumors with similar morphological appearance should be examined for TTF-1 immunoreactivity , and patients should be clinically followed to determine the course of this unusual disease and the significance of TTF-1 expression .", "output": "Resisting cell death" }, { "input": "We studied the effect of the immune system on two differentially aggressive melanomas developing in mice on conditional deletion of the INK4A/ARF tumor suppressor gene , with concomitant expression of oncogene H-Ras(G12V) and a natural cancer-germline tumor antigen ( TA ) .\n\n\" Slow progressor \" melanomas contained no activated T lymphocytes ( TL ) .\n\nIn contrast , \" aggressive \" melanomas were infiltrated by activated TLs lacking effector molecules and expressing high levels of PD-1 , indicating an exhausted phenotype .\n\nAggressive melanomas were also infiltrated by immature myeloid cells ( IMC ) .\n\nInfiltration was associated with local inflammation and systemic Th2/Th17-oriented chronic inflammation that seemed to impair further activation of TLs , as tumor-specific T cells adoptively transferred into mice bearing aggressive melanomas were poorly activated and failed to infiltrate the melanoma .\n\nThis immunosuppression also led to the incapacity of these mice to reject inoculated TA-positive tumors , in contrast to slow-progressing melanoma-bearing mice , which were responsive .\n\nTo test the role of adaptive immunity in tumor progression , we induced melanomas in immunodeficient RagKO compound mice .\n\nThese mice developed aggressive but not slow-progressing melanomas at a higher frequency and with a shorter latency than immunocompetent mice .\n\nImmunodeficient mice also developed abnormal inflammation and infiltration of IMCs in a manner similar to immunocompetent mice , indicating that this phenotype was not dependent on adaptive immunity .\n\nTherefore , tumor-intrinsic factors distinguishing the two melanoma types control the initiation of inflammation , which was independent of adaptive immunity .\n\nThe latter delayed development of aggressive melanomas but was overridden by inflammation .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "OBJECTIVE Although downregulation of neural cell adhesion molecule ( NCAM ) has been correlated with poor prognosis in colorectal cancer ( CRC ) , it is also possible that colon cancer spreading comes from reducing tumor cell adhesion through NCAM polysialylation , as occurs in lung carcinoma or Wilms ' tumor .\n\nMETHODS To prove this hypothesis , we have performed a prospective study on tumor and control specimens from 39 CRC patients , which were immunostained for NCAM and PSA ( polysialic acid ) expression .\n\nRESULTS Tumor versus control expression of NCAM and PSA epitopes in tissue specimens , as well as correlation between tumor expression and clinicopathological features , were statistically analyzed .\n\nResults showed a low constitutive expression of NCAM and PSA ( PSA-NCAM ) in control tissue , which reached a statistically significant increase in the tumor tissue .\n\nLikewise , the presence and number of lymph node metastases at surgery were correlated with NCAM expression and PSA/NCAM coexpression .\n\nCONCLUSIONS These data highlight the importance of taking into account PSA-associated epitopes when dealing with NCAM cell expression studies in tumor development and progression .\n\nThe analysis of PSA and NCAM expression in CRC suggests a new way , other than downregulation of NCAM , in order to escape contact inhibition and promote cell tumor spreading in colorectal cancer .", "output": "Evading growth suppressors" }, { "input": "Polycylic aromatic hydrocarbons ( PAHs ) are ubiquitous contaminants in the marine environment .\n\nTheir toxicity is mainly linked to the ability of marine species to biotransform them into reactive metabolites .\n\nPAHs are thus often detected at trace levels in animal tissues .\n\nFor biomonitoring purposes , this findings have two main consequences , ( i ) the determination of the PAH tissue concentration is not suitable for the evaluation of individual exposure to PAHs ( ii ) it can explain sometimes the lack of correlations obtained with relevant markers of toxicity such as genotoxicity biomarkers .\n\nThe aim of the present study was to better investigate the link between PAH exposure and genotoxicity in marine flatfish .\n\nDuring a laboratory experiment , juvenile soles were exposed for four weeks to a mixture of three PAHs , namely benzo[a]pyrene , fluoranthene and pyrene , followed by one week of depuration .\n\nFish were exposed via the trophic route to a daily PAH concentration of 120 \u03bcg/g food .\n\nFish were sampled at different time points .\n\nThe bioavailability and the biotransformation of PAHs were assessed by the measurement of biliary metabolites using a sensitive UPLC MS/MS method .\n\nThe 7-ethoxyresorufine-O-deethylase was also measured in liver subcellular fractions as a biomarker of phase I biotransformation activities .\n\nGenotoxicity was assessed in parallel by the measurement of DNA strand breaks in fish erythrocytes by the alkaline comet assay .\n\nDuring this study , the high amount of PAH metabolites produced in sole demonstrated the bioavailability of PAHs and their biotransformation by fish enzymes .\n\nA positive correlation was observed between the level of hydroxylated PAH metabolites and genotoxicity as measured by the alkaline comet assay .", "output": "Genomic instability and mutation" }, { "input": "Principal components analysis ( PCA ) combined to Fourier transform infrared ( FTIR ) microspectroscopic imaging was used to screen biochemical changes associated to C6 glioma tumor from 7 to 19 days growth .\n\nNormal brain and tumor obtained at 7 , 12 , 19 days after C6 cell injection were used to develop a diagnostic model of brain glioma based on PCA analysis .\n\nThis classification model was validated using extra-measurements on normal and tumor at 9 and 15 days post-implantation .\n\nThe spatial and biochemical information obtained from FTIR/PCA maps can be used to improve the discrimination between normal and grading human glioma .\n\nThe first 4 PCs which account for more than 93.6% of total spectral variance were used to construct pseudo-color scores maps and compared each map to the corresponding hematoxylin and eosin ( H&E ) staining .\n\nOur results reported that by correlating pseudocolor map scores with H&E staining it was possible to screen histological changes associated with tissue transformation .\n\nIn fact , PC1 and PC4 were associated to the tumor , surrounding tumor and necrosis .\n\nIndeed , at day 7 after tumor implantation , FTIR investigations displayed a very small abnormal zone associated with the proliferation of C6 cells in the caudate putamen ( CP ) .\n\nPC2 and PC3 described normal brain structures such as white matter ( corpus callosum ( CC ) and commissura anterior ( CA) ) and some cortex layers ( grey matter ) .\n\nAfter delipidation of the tissues , we were still able to differentiate between different tissue features based on nucleic acid and protein content .\n\nBy comparing the patterns of the PC loads with the spectra of lipids extracted from white and gray matters , and DNA , we have identified some biochemical changes associated with tissue transformation .\n\nThis work demonstrated that our classification model provides a successful histological classification of different brain structures .", "output": "Resisting cell death" }, { "input": "Oxazaphosphorines are a class of DNA alkylating agents .\n\nThe aim of the present study was to compare the possible influence of three new generation oxazaphosphorines , D-17272 ( mafosfamide cyclohexylamine salt ) , D-18864 ( 4-hydro-peroxy-cyclophosphamide ) , and D-19575 ( glufosfamide , beta-D-glucose-isophosphoramide mustard ) on DNA damage induction in the human histiocytic lymphoma U937 cells .\n\nThe flow cytometry APO-BRDU assay , based on the TUNEL method , was used for the in situ detection of DNA strand breaks .\n\nAfter exposure of U937 cells to the oxazaphosphorines , the patterns of temporary changes in the frequency of TUNEL positive U937 cells , expressing DNA breakage , were determined .\n\nThe effects of the oxazaphosphorines on U937 cells were dependent on the agent tested and its dose , and the time intervals after the drug application .\n\nThe different potential of D-17272 , D-18864 and D-19575 to induce DNA strand breakage in the human histiocytic lymphoma U937 cells was shown .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND/AIMS Adenocarcinoma of the gallbladder is a highly malignant neoplasm. p16 is a tumor suppressor gene protein , which is a cyclin-dependent kinase inhibitor that regulates the G1-S phase of the cell cycle .\n\nThe purpose of the present study was to investigate the expression of p16 in gallbladder carcinoma and its precancerous conditions and to examine the relationship between p16 expression and clinicopathological parameters .\n\nMETHODOLOGY Formalin-fixed , paraffin-embedded tissue sections from 20 cases of normal gallbladder , 20 cases of chronic cholecystitis , 20 cases of gallbladder adenoma , 20 cases of dysplasia , and 58 cases of adenocarcinoma were examined .\n\nThe expression of p16 was evaluated by immunohistochemical analysis .\n\nRESULTS In normal gallbladder , no expression of p16 was found .\n\nIn chronic cholecystitis , expression of p16 was not found .\n\nIn gallbladder adenomas , expression of p16 was found in 20% ( 4/20 ) .\n\nIn low grade dyspalsias , expression of p16 was not found .\n\nIn high grade dysplasias , p16 expression was present in 45.0% ( 9/20 ) .\n\nIn gallbladder adenocarcinomas , p16 expression was found in 27.6% ( 16/58 ) .\n\nExpression of p16 correlated significantly with histologic grade ( p < 0.05 ) .\n\nNo correlation was found between p16 expression and age , gender , tumor size , gross type , location , vascular invasion , lymph node metastasis , and TNM stage , respectively .\n\nCONCLUSIONS P16 protein overexpression is an early and relatively common event in carcinogenesis of gallbladder carcinoma .\n\nExpression of p16 protein is absent in normal or chronic cholecystitis .\n\nExpression of p16 may be an ancillary diagnostic marker of gallbladder carcinoma and its precancerous conditions .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND/AIMS The use of chemotherapy in hepatocellular carcinoma is still controversial .\n\nThe aim of this study was to investigate whether the combined use of epirubicin and progesterone has a synergistic effect on cell proliferation and apoptosis in HepG2 cells .\n\nMETHODOLOGY Different concentrations of epirubicin ( 0.1 microg/ml , 0.25 microg/ml and 0.5microg/ml ) or progesterone ( 12.5 microM , 25 microM and 50 microM ) were added to HepG2 cells either alone or in combinations consisting of different concentrations of the two .\n\nTheir effects on HepG2 cells were studied by ( 1 ) XTT assay for analysis of cell proliferation , ( 2 ) 3H-Thymidine incorporation for DNA synthesis , ( 3 ) annexin V-FITC/ propidium iodide ( PI ) flowcytometery for cell apoptosis , ( 4 ) flowcytometry for cell cycle distributions , and ( 5 ) reverse transcription-polymerase chain reaction for expression of cell cycle modulator , cyclin D1 .\n\nRESULTS 50 microM progesterone increased both the cytotoxic and apoptotic effects of 0.1 microg/ml epirubicin on HepG2 cells at 48 hr culture due to 50 microM progesterone accumulated cells in S phase of the cell cycle and subsequently reduced cyclin D1 expression .\n\nThese effects on HepG2 cells induced by this combination were comparable to those induced by 0.5 microg/ml epirubicin alone .\n\nCONCLUSIONS In vitro , progesterone can increase the cytotoxicity and apoptosis induced by epirubicin on HepG2 cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Among several types of DNA lesions , the DNA double strand breaks ( DSBs ) are one of the most deleterious and harmful .\n\nMammalian cells mount a coordinated response to DSBs with the aim of appropriately repair the DNA damage .\n\nIndeed , failure of the DNA damage response ( DDR ) can lead to the development of cancer-prone genetic diseases .\n\nThe identification and development of drugs targeting proteins involved in the DDR is even more investigated , as it gives the possibility to specifically target cancer cells .\n\nIndeed , the administration of DNA repair inhibitors could be combined with chemo- and radiotherapy , thus improving the eradication of tumor cells .\n\nHere , we provide an overview about DSBs damage response , focusing on the role of the DSBs repair mechanisms , of chromatin modifications , and of the cancer susceptibility gene BRCA1 which plays a multifunctional role in controlling genome integrity .\n\nMoreover , the most investigated DSBs enzyme inhibitors tested as potential therapeutic agents for anti-cancer therapy are reported .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE To explore the association between DNA damage induced by vinyl chloride monomer ( VCM ) and polymorphisms of DNA repair genes and xenobiotic metabolism genes of VCM .\n\nMETHODS Comet assay was employed to detect DNA damage .\n\nBased on the status of DNA damage , the VCM exposure workers were divided into two groups : DNA damage group ( 75 ) and control group ( 75 ) .\n\nCase-control design was used to investigate the association between the genetic polymorphisms and DNA damage induced by VCM .\n\nGenotypes of XRCC1 ( Arg194Trp , Arg280His and Arg399Gln ) , XPD ( Ile199Met , Asp312Asn and Lys751Gln ) and CYP2E1 were identified by the PCR-RFLP .\n\nPCR assay was used to detect positive and null genotype of GSTT1 and GSTM1 .\n\nRESULTS Univariate analysis showed that the CYP2E1 c1c2/c2c2 and XPD751 Lys/Gln and Gln/Gln genotypes were significantly associated with the increased levels of DNA damage , XRCCI 339 Arg/Gln and Gln/Gln genotypes were significantly associated with the decreased levels of DNA damage ( P < 0.01 , P < 0.05 , respectively ) .\n\nLogistic regression analysis showed that there was significant association between the genotypes of XRCC1 194 , XRCC1 399 , XPD 751 , CYP2E1 and DNA damages .\n\nA prominent risk decreasing of DNA damage was observed for those individuals possessing XRCC1 399Arg/Gln + Gln/Gln genotypes ( OR : 0.35 , 95%CI : 0.12 approximately 1.01 , respectively ) ; The results also showed that there were significant associations between CYP2E1 c1c2/c2c2 and DNA damage both in high and low VCM-exposed groups ( OR : 2.57 , 95%CI : 1.01 approximately 6.59 and OR : 2.57 , 95%CI : 0.99 approximately 6.87 ) .\n\nCONCLUSION Cumulative exposure dose and genotypes of XRCC1 194 , XRCC1 399 , XPD 751 and CYP2E1 may modulate the DNA damage induced by VCM exposure .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVES The aim of this study is to examine the expression of MCM-2 and conventional proliferation marker Ki-67 in breast carcinoma by stereologic technique and to compare it with various clinicopathologic parameters .\n\nMETHODS The expression of MCM-2 and Ki-67 on paraffin-embedded tumor tissue sections of patients with invasive breast carcinoma was analyzed immunohistochemically .\n\nStereologic method was used for evaluation of the percentage of positively stained tumor cells .\n\nRESULTS Significant positive correlation was found between the expression of MCM-2 and that of Ki-67 ( r = 0.74 , p < 0.001 ) .\n\nMCM-2 and Ki-67 expression was significantly associated with histologic grade ( p < 0.05 ) , and negative correlation was observed between MCM-2 or Ki-67 expression and estrogen status ( p < 0.05 ) .\n\nNo significant association was observed between MCM-2 or Ki-67 expression and patient age , tumor size , lymph node status , clinical stage and menopausal status .\n\nCONCLUSION Our results suggest that MCM-2 expression is significantly associated with histologic grade of breast carcinoma and with cell proliferation capacity ( Ki-67 labelling index ) .\n\nAdditional studies are required using the stereologic method to compare and understand the utility of Ki-67 and MCM-2 expression in invasive breast carcinoma ( Tab. 1 , Fig. 4 , Ref. 34 ) .\n\nFull Text ( Free , PDF ) www.bmj.sk .", "output": "Sustaining proliferative signaling" }, { "input": "Recent surveys indicate that Pi intake has increased steadily as Pi-containing foods have increased .\n\nOur previous study demonstrated that high dietary Pi strongly stimulated lung tumorigeneis .\n\nIn order to answer the issue whether low Pi may be chemopreventive , we examined the effects of low Pi on lung cancer .\n\nEighteen 5-wk-old male K-ras(LA1) lung cancer model mice were randomly allocated to 2 groups .\n\nOne group was fed a normal diet ( 0.5% Pi ) and other group was fed low Pi ( 0.1% Pi ) diet for 4 wk .\n\nLung cancer development was evaluated by histopathological examination , Western blot , kinase assay , and immunohistochemistry .\n\nLow Pi increased the expression of sodium-dependent phosphate co-transporter 2b , and activated Akt signal with decreased PTEN expression in the lungs of K-ras(LA1) mice .\n\nLow Pi increased the Akt/mTOR-mediated protein translation through upregulating the phosphorylation of p70S6K and 4E-BP1 .\n\nIn addition , low Pi stimulated cell cycling as evidenced by altered cell cycle regulators such as cyclin D1 and D3 .\n\nFinally , low Pi increased lung tumorigenesis in K-ras(LA1) mice compared to the normal diet group .\n\nOur results clearly demonstrated that low Pi also promoted lung tumorigenesis , thus suggesting that an appropriate intake of dietary Pi may be critical for lung cancer prevention as well as treatment .", "output": "Sustaining proliferative signaling" }, { "input": "Caveolin-1 ( -/- ) null stromal cells are a novel genetic model for cancer-associated fibroblasts and myofibroblasts .\n\nHere , we used an unbiased informatics analysis of transcriptional gene profiling to show that Cav-1 ( -/- ) bone-marrow derived stromal cells bear a striking resemblance to the activated tumor stroma of human breast cancers .\n\nMore specifically , the transcriptional profiles of Cav-1 ( -/- ) stromal cells were most closely related to the primary tumor stroma of breast cancer patients that had undergone lymph-node ( LN ) metastasis .\n\nThis is consistent with previous morphological data demonstrating that a loss of stromal Cav-1 protein ( by immuno-histochemical staining in the fibroblast compartment ) is significantly associated with increased LN-metastasis .\n\nWe also provide evidence that the tumor stroma of human breast cancers shows a transcriptional shift towards oxidative stress , DNA damage/repair , inflammation , hypoxia , and aerobic glycolysis , consistent with the \" Reverse Warburg Effect \" .\n\nFinally , the tumor stroma of \" metastasis-prone \" breast cancer patients was most closely related to the transcriptional profiles derived from the brains of patients with Alzheimer's disease .\n\nThis suggests that certain fundamental biological processes are common to both an activated tumor stroma and neuro-degenerative stress .\n\nThese processes may include oxidative stress , NO over-production ( peroxynitrite formation ) , inflammation , hypoxia , and mitochondrial dysfunction , which are thought to occur in Alzheimer?s disease pathology .\n\nThus , a loss of Cav-1 expression in cancer-associated myofibroblasts may be a protein biomarker for oxidative stress , aerobic glycolysis , and inflammation , driving the \" Reverse Warburg Effect \" in the tumor micro-environment and cancer cell metastasis .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Cellular energetics" }, { "input": "BACKGROUND CDC4/FBXW7 , encoding a ubiquitin ligase , maps to 4q32 and has been implicated as a tumor suppressor gene and therapeutic target in many tumor types .\n\nMutations in colonic adenomas , and the frequent losses on 4q described in gastric cancer prompt speculation about the role of CDC4/FBXW7 in gastric carcinogenesis .\n\nMETHODS We assessed the role of CDC4/FBXW7 in gastric cancer , through loss of heterozygosity ( LOH ) and multiplex ligation-dependent probe amplification ( MLPA ) on 47 flow-sorted gastric carcinomas including early-onset gastric cancers ( EOGC ) and xenografted conventional gastric carcinomas .\n\nPloidy analysis was carried out on 39 EOGCs and immunohistochemistry of CDC4/FBXW7 and its substrates c-myc , c-jun , Notch and cyclin E was performed on 204 gastric carcinomas using tissue microarrays ( TMAs ) .\n\nSequence analysis of CDC4/FBXW7 was carried out on gastric carcinoma cell lines and xenografts .\n\nRESULTS Loss of heterozygosity of CDC4/FBXW7 occurred in 32% of EOGCs , and correlated with loss of expression in 26% .\n\nLoss of expression was frequent in both EOGC and conventional gastric cancers .\n\nNo CDC4/FBXW7 mutations were found and loss of CDC4/FBXW7 did not correlate with ploidy status .\n\nThere was a significant correlation between loss of CDC4/FBXW7 expression and upregulation of c-myc .\n\nCONCLUSION Loss of CDC4/FBXW7 appears to play a role in both EOGC and conventional gastric carcinogenesis , and c-myc overexpression is likely to be an important oncogenic consequence of CDC4/FBXW7 loss .", "output": "Genomic instability and mutation" }, { "input": "XRCC4 , in association with DNA ligase IV , is thought to play a critical role in the ligation of two DNA ends in DNA double-strand break ( DSB ) repair through non-homologous end-joining ( NHEJ ) pathway .\n\nIn the present study , we captured radiation-induced chromatin-recruitment of XRCC4 by biochemical fractionation using detergent Nonidet P-40 .\n\nA subpopulation of XRCC4 changed into a form that is resistant to the extraction with 0.5% Nonidet P-40-containing buffer after irradiation .\n\nThis form of XRCC4 was liberated by micrococcal nuclease treatment , indicating that it had been tethered to chromatin DNA .\n\nThis chromatin-recruitment of XRCC4 could be seen immediately ( < 0.1 hr ) after irradiation and remained up to 4 hr after 20 Gy irradiation .\n\nIt was seen even after irradiation of small doses , i.e. , 2 Gy , but the residence of XRCC4 on chromatin was very transient after 2 Gy irradiation , returning to near normal level in 0.2-0.5 hr after irradiation .\n\nThe chromatin-bound XRCC4 represented only approximately 1% of total XRCC4 molecules even after 20 Gy irradiation and the quantitative analysis using purified protein as the reference suggested that only a few XRCC4-DNA ligase IV complexes were recruited to each DNA end .\n\nWe further show that the chromatin-recruitment of XRCC4 was not attenuated by wortmannin , an inhibitor of DNA-PK , or siRNA-mediated knockdown of the DNA-PK catalytic subunit ( DNA-PKcs ) , indicating that this process does not require DNA-PKcs .\n\nThese results would provide us with useful experimental tools and important insights to understand the DNA repair process through NHEJ pathway .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE To study the clinicopathologic features , immunophenotype and ultrastructural features of sinonasal inflammatory myofibroblastic tumors ( IMT ) .\n\nMETHODS The clinical and histologic features of 5 cases of sinonasal IMT were reviewed .\n\nImmunohistochemical study for vimentin , MSA , SMA , calponin , h-caldesmon , desmin , ALK , fibronectin , CK , S-100 and Ki-67 was carried out .\n\nUltrastructural examination was also performed in two of the cases .\n\nRESULTS The patients age ranged from 28 to 62 years ( mean = 43 years ) .\n\nThe male-to-female ratio was 2:3 .\n\nThe clinical presentation included nasal obstruction , nasal discharge , nasal bleeding , facial pain , facial swelling , toothache and tear overflow .\n\nAll of the 5 patients suffered from disease relapses ; and 4 of them had recurrences for more than 5 times .\n\nOne patient had lymph node metastasis and 3 patients died of the disease .\n\nHistologically , the tumor cells were arranged in interlacing fascicles and sometimes haphazard in fashion .\n\nThey were spindly in shape , cytoplasm eosinophilic with mild nuclear atypia and a low mitotic activity .\n\nThe intervening stroma was myxoid in appearance accompanied by lymphocyte and plasma cell infiltration , abundant blood vessels and focal collagenized areas .\n\nIn 3 of the recurrent cases , the tumor cells displayed increased nuclear atypia and mitotic activity ( average about 5 to 6 per 10 high-power fields ) , accompanied by patchy necrosis , less inflammatory cell infiltration and focal sarcomatous changes .\n\nImmunohistochemical study showed that the tumor cells were diffusely positive for vimentin .\n\nSMA , MSA , calponin and fibronectin were variably expressed .\n\nDesmin was weakly positive in 1 case .\n\nThe staining for h-caldesmon , ALK , S-100 and CK was negative .\n\nThe Ki-67 proliferation index increased with tumor recurrences .\n\nElectron microscopy revealed abundant rough endoplasmic reticulum and dense body formation in the cytoplasm .\n\nThere were an increased amount of collagen fibers in the stroma .\n\nCONCLUSIONS IMT rarely occurs in nasal cavity and paranasal sinuses .\n\nThe tumor is prone to local invasion and recurrences , with subsequent progression to frank malignancy and distant metastasis , resulting in high mortality and poor prognosis .\n\nComplete surgical resection remains the main modality of treatment .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Recent research has shown that the Na(+)/K(+)-ATPase alpha1 subunit is a novel anti-cancer target , which plays pivotal roles in malignant cell ion transport , metabolism , migration and signal transduction .\n\nThe purpose of the present study was to investigate the anti-cancer effects of ouabain and Na(+)/K(+)-ATPase alpha1 small interfering ribonucleic acid ( siRNA ) on HepG2 cell proliferation , apoptosis and cell cycle , and to explore the molecular mechanisms .\n\nThe expression of Na(+)/K(+)-ATPase alpha1 subunit in human hepatocellular carcinoma ( HCC ) , normal liver tissues and human HCC line ( HepG2 , SMMC-7721 and Bel-7402 ) has been investigated .\n\nUsing the ouabain and Na(+)/K(+)-ATPase alpha1 subunit siRNA , which target the Na(+)/K(+)-ATPase , we have evaluated the effects of inhibiting Na(+)/K(+)-ATPase alpha1 in human HepG2 cells with respect to cell proliferation , morphology , cell cycle , impact on intracellular Ca2++ , reactive oxygen species ( ROS ) concentration , and correlated gene expression level on messenger ribonucleic acid ( mRNA ) and protein .\n\nOur data showed that the expression Na(+)/K(+)-ATPase alpha1 subunit in HCC tissues is higher than that in normal liver tissues .\n\nOuabain and Na(+)/K(+)-ATPase alpha1 siRNA could inhibit HepG2 cell proliferation .\n\nOuabain could induce HepG2 cell apoptosis and generate S phase arrest , and siRNA could enhance the anti-cancer effect of ouabain that induced HepG2 cells apoptosis via an intracellular Ca(2+) and ROS increase-mediated , and generated cell cycle S phase arresting by decreasing the CyclinA1/cyclin-dependent kinase 2 ( CDK2)/proliferating cell nuclear antigen ( PCNA ) complex product and increasing the expression of cyclin-dependent kinase inhibitor 1A ( P21(CIP1) ) .\n\nWe believe that targeting of the Na(+)/K(+)-ATPase alpha1 subunit in human HCC cells could provide new sight into the treatment of HCC .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Squamous cell carcinoma ( SCC ) is the most frequent cancer in organ transplant recipients ( OTRs ) .\n\nThe immune system plays a major role in the fight against SCC , however , little is known about the local inflammatory response in SCC at all .\n\nWe analyzed quantity and quality of the perineoplastic inflammatory SCC microenvironment in immunocompetent patients and immmunosuppressed OTRs .\n\nRNA expression profile of SCC patients was analyzed for 8 different sets of genes relating to Th1 versus Th2 response using Gene Set Enrichment Analysis .\n\nSCC from immunocompetent patients and OTRs were analyzed by real-time polymerase chain reactions for CD4 , CD8 , TBET , GATA-3 , FOXP3 , RORC , IFN-gamma , IL-4 , TGF-beta , IL-10 , and IL-17A mRNA expression .\n\nImmunohistochemistry was carried out in SCC for CD3 , CD4 , CD8 , and FOXP3 expression .\n\nConsiderable inflammation was seen in both patient groups .\n\nSCC in immunocompetent patients and OTRs was associated with a mixed Th1 and Th2 gene expression signature .\n\nCD4(+) mRNA was diminished in immunosuppression .\n\nSkin adjacent to SCC in OTRs showed Th2 expression pattern as compared with immunocompetent patients .\n\nT-BET and IFN-gamma mRNA expression were decreased in the OTR group .\n\nAlthough Th17-weighted inflammation was unchanged , IL-17A mRNA level was markedly decreased with immunosuppression .\n\nRegulatory T cells , characterized by FOX-P3 and TGF-beta mRNA level , were decreased in OTRs .\n\nOur findings support the hypothesis that nontumor-bearing skin adjacent to SCC in OTRs is not necessarily normal and that the local microenvironment may contribute to a field effect contributing to higher recurrence rates and more aggressive behavior observed in these patients .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "Oral squamous cell carcinoma ( OSCC ) is the most common malignant tumor in the oral and maxillofacial region .\n\nThe mechanism of carcinogenesis of OSCC is still unclear .\n\nBased on the previous cell line , HIOEC-B(a)P-96 ( HB96 ) , which we obtained by HPV16 E6/E7-immortalized human oral epithelial cells ( HIOEC ) and benzo(a)pyrene inducement , we prepared a new HB-second generation cancer cell line ( HB-2 ) by continuous passage .\n\nIts characteristics such as morphology , proliferation activity , karyotype , and tumorigenesis were studied .\n\nThe HB-2 cells displayed uncontrolled cell division and lost contact inhibition leading to cell overlap .\n\nCells were polygonal and unevenly shaped , with an increased nucleus versus plasma ratio .\n\nIncreased proliferative activity was confirmed by MTT assays .\n\nThe tumorigenicity was confirmed by tumor growth experiments in nude mice .\n\nTherefore , the HB-2 and HB96 cell lines are useful tools to study the mechanism of carcinogenesis of OSCC in vitro for future genomic and proteomic analyses .", "output": "Evading growth suppressors" }, { "input": "Regulation of adaptive immunity by innate immune cells is widely accepted .\n\nConversely , adaptive immune cells can also regulate cells of the innate immune system .\n\nHere , we report for the first time the essential role of B cells in regulating macrophage ( M\u03c6 ) phenotype .\n\nIn vitro B cell/M\u03c6 co-culture experiments together with experiments in transgenic mice models for B-cell deficiency or overexpression showed B1 cells to polarize M\u03c6 to a distinct phenotype .\n\nThis was characterized by downregulated TNF-\u03b1 , IL-1\u03b2 and CCL3 , but upregulated IL-10 upon LPS stimulation ; constitutive expression of M2 M\u03c6 markers ( e.g .\n\nYm1 , Fizz1 ) and overexpression of TRIF-dependent cytokines ( IFN-\u03b2 , CCL5 ) .\n\nMechanistically , this phenotype was linked to a defective NF-\u03baB activation , but a functional TRIF/STAT1 pathway .\n\nB1-cell-derived IL-10 was found to be instrumental in the polarization of these M\u03c6 .\n\nFinally , in vivo relevance of B1-cell-induced M\u03c6 polarization was confirmed using the B16 melanoma tumor model where adoptive transfer of B1 cells induced an M2 polarization of tumor-associated M\u03c6 .\n\nCollectively , our results define a new mechanism of M\u03c6 polarization wherein B1 cells play a key role in driving M\u03c6 to a unique , but M2-biased phenotype .\n\nFuture studies along these lines may lead to targeting of B1 cells to regulate M\u03c6 response in inflammation and cancer .", "output": "Tumor promoting inflammation" }, { "input": "The effect of polychlorinated biphenyls ( PCBs ) on Vero cell proliferation was investigated , with the attempts to assess the possible hormetic dose-response and to compare their structure-dependent toxicity .\n\nBoth PCB congeners revealed low doses stimulation in our experiment .\n\nHowever , significant cytotoxicity was only observed in PCB 52 concentrations larger than 0.1 microg ml(-1) , while there was no significant inhibition in PCB 77-treated cells at concentrations selected .\n\nFurthermore , the time-dependent cytotoxic trends were different .\n\nThe comparison between PCB 52 and PCB 77 indicated that the cytotoxic mechanisms involved in coplanar or non-coplanar PCB congener exposure were different , and this difference might be associated with individual genotoxicity and the release of contact inhibition , respectively .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "Abstract The use of immunotherapeutics in melanoma has received much attention , and recent advances to further characterize the regulatory components of the immune system and the importance of co-stimulatory molecules have opened a new area for clinical investigation .\n\nCytotoxic T lymphocyte-associated antigen 4 ( CTLA-4 ) serves as a negative regulator of immunity .\n\nRecent trials administering fully human anti-CTLA-4 monoclonal antibodies to melanoma patients have demonstrated clinically meaningful responses .\n\nTreatment with CTLA-4 blocking antibodies , however , is not without potential toxicities .\n\nAutoimmune side-effects , the most common being colitis-associated diarrhea , are frequently associated with clinical responses .\n\nIn efforts to build upon prior vaccination efforts as well as attempt to offer patients clinically meaningful immune responses with a CTLA-4 blockade but without significant toxicities , we conducted a clinical trial in patients who previously received autologous tumor cells engineered to secrete granulocyte-macrophage colony stimulating factor ( GVAX ; Cell Genesys , South San Francisco , CA , USA ) with periodic infusions of CTLA-4 blocking antibodies .\n\nThis sequential treatment resulted in clinically significant anti-tumor immunity without grade 3 or 4 toxicity in most patients .\n\nPathological analyses following treatment of pre-existing tumors revealed a linear correlation between tumor necrosis and the ratio of intra-tumoral CD8+ effector cells to FoxP3+ regulatory cells ( T(regs) ) .\n\nEffective anti-tumor immunity and serious autoimmunity can be disassociated .\n\nFurther targeting of anti-tumor T(regs)in combinatorial therapy approaches may be a rich avenue of future investigation .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "Pam3CSK4 , a synthetic TLR2 ligand , has been shown to expand CD4+ regulatory T cells ( Treg cells ) .\n\nLess is known about the function of CD8+ Treg cells than about the function of CD4+ Treg cells generated during allergen-specific immunotherapy ( IT ) .\n\nThis study investigated whether Dermatophagoides pteronyssinus-specific IT could expand the CD8+CD25+Foxp3+ Treg population and whether Pam3CSK4 could enhance the Treg population .\n\nPBMCs were isolated from healthy control subjects and from mite-sensitive asthmatic patients during IT at three specific times : before IT and 6 mo and 1 y after the maximum-tolerated dose .\n\nThis study was performed without a placebo-controlled group .\n\nD. pteronyssinus-specific IT induced a significant increase in CD8+Foxp3+ Treg cells expressing intracellular IL-10 and granzyme B. Costimulation of PBMCs with Pam3CSK4 and D. pteronyssinus 2 expanded the CD8+CD25+Foxp3+ Treg population and inhibited D. pteronyssinus 2-induced IL-4 production .\n\nPam3CSK4-treated CD8+CD25+ Treg cells directly suppressed CD4+ T cell proliferation by cell-contact inhibition .\n\nTUNEL revealed that CD8+CD25+ Treg cells , but not CD4+CD25+ Treg cells , directly induced CD4+CD45ROhi+ apoptosis .\n\nOur results provide direct evidence that Pam3CSK4 induces an immunomodulatory effect by inducing CD8+ Treg cells ; therefore , it may be a good adjuvant for the treatment of mite allergies .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "OBJECTIVES Toxicity caused by pharmacological and chemical substances , including carbon tetrachloride ( CCl(4) ) , is a major pathological factor for liver injury .\n\nTherefore , strategies to prevent toxicity are needed for maintaining a healthy liver .\n\nThis study was designed to determine whether recombinant bovine pancreatic trypsin inhibitor ( rBPTI ) , a non-specific serine protease inhibitor , prevents CCl(4)-induced liver injury in mice .\n\nMETHODS Mice were treated with CCl(4) in the presence or absence of co-treatment with rBPTI .\n\nLiver sections were prepared for histopathological assessment .\n\nLiver function was evaluated by detecting serum levels of alanine aminotransferase ( ALT ) and aspartate aminotransferase ( AST ) and liver index .\n\nLiver oxidative stress and inflammation were examined by detecting the liver malondialdehyde level and glutathione and superoxide dismutase activity , and serum tumour necrosis factor-alpha level , respectively .\n\nKEY FINDINGS CCl(4) induced hepatocyte necrosis , inflammatory cell infiltration and fatty degeneration , which were ameliorated by co-treatment with rBPTI in a concentration-dependent manner .\n\nFurthermore , rBPTI prevented CCl(4)-induced disruption of liver function .\n\nImportantly , rBPTI reduced CCl(4)-induced liver oxidative stress response and pro-inflammatory cytokine production .\n\nCONCLUSIONS These results indicated that rBPTI exerted a protective effect on CCl(4)-induced liver injury in mice .\n\nThus , rBPTI may have potential application for prevention of liver injury induced by metabolism of drugs and toxic substances .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "In order to increase the permeability of cell membranes for low doses of cytostatic drugs , two bioelectrochemical methods have been compared : ( a ) electric pore formation in the plasma membranes by single electric impulses ( electroporation ) , and ( b ) reordering of membrane structure by alternating currents ( capacitively coupled ) .\n\nThese treatments were applied to human leukemic K-562 cells and human lymphoma U-937 cells , yielding apoptotic and necrotic effects , determined by flow cytometry .\n\nAdditional cell death occurs after exposure to light irradiation at wavelengths lambda > 600 nm , of cells which were electroporated and had incorporated actinomycin-C or daunomycin ( daunorubicin ) .\n\nIt is observed that drug uptake after an exponentially decaying electroporation pulse of the initial field strength Eo=1.4 kV/cm and pulse time constants in the time range 0.5-3 ms is faster than during PEMF-treatment , i.e. , application of an alternating current of 16 kHz , voltage U<100 V , I=55 mA , and exposure time 20 min .\n\nHowever , at the low a.c. voltage of this treatment , more apoptotic and necrotic cells are produced as compared to the electroporation treatment with one exponentially decaying voltage pulse .\n\nThus , additional photodynamic action appears to be more effective than solely drugs and electroporation as applied in clinical electrochemotherapy , and more effective than the noninvasive pulsed electromagnetic fields ( PEMFs ) , for cancer cells in general and animals bearing tumors in particular .", "output": "Resisting cell death" }, { "input": "INTRODUCTION Necrosis at the tumor center is a common feature of aggressive breast cancers and has been associated with poor prognosis .\n\nIt is commonly identified by means of invasive histopathology , which often correlates with morbidity and potential tumor cell dissemination , and limits the reconstruction of the whole necrotic domain .\n\nIn this study we hypothesized that non covalent association to serum albumin ( SA ) and covalent binding to ligands for tumor-abundant cell receptors should synergistically drive selective accumulation and prolonged retention of imaging and therapeutic agents in breast tumor necrotic domains enabling in vivo identification , imaging and possibly treatment of such tumors .\n\nMETHODS Cyclo-Arg-Gly-Asp-D-Phe-Lys ( c(RGDfK) ) were conjugated to bacteriochlorophyll-derivatives ( Bchl-Ds ) , previously developed as photodynamic agents , fluorescent probes and metal chelators in our lab .\n\nThe c(RGDfK) component drives ligation to alphaVbeta3 integrin receptors over-expressed by tumor cells and neo-vessels , and the Bchl-D component associates to SA in a non-covalent manner .\n\nSTL-6014 , a c(RGDfK)-Bchl-D representative , was i.v. injected to CD-1 , nude female mice bearing necrotic and non-necrotic human MDA-MB-231-RFP breast cancer tumors .\n\nThe fluorescence signals of the Bchl-Ds and RFP were monitored over days after treatment , by quantitative whole body imaging and excised tumor/tissue samples derived thereof .\n\nComplementary experiments included competitive inhibition of STL-6014 uptake by free c(RGDfK) , comparative pharmacokinetics of nonconjugated c(RGDfK) Bchl-D ( STL-7012 ) and of two human serum albumin ( HSA ) conjugates : HSA-STL-7012 and HSA-STL-6014 .\n\nRESULTS STL-6014 and STL-7012 formed complexes with HSA ( HSA/STL-6014 , HSA/STL-7012 ) .\n\nSTL-6014 , HSA-STL-7012 and HSA-STL-6014 , selectively accumulated at similar rates , in tumor viable regions over the first 8 h post administration .\n\nThey then migrated into the necrotic tumor domain and presented tumor half lifetimes ( T1/2 ) in the range of days where T1/2 for HSA-STL-6014 > STL-6014 > HSA-STL-7012 .\n\nNo accumulation of STL-7012 was observed .\n\nPre-injection of c(RGDfK) excess , prevented the uptake of STL-6014 in the small , but not in the large tumors .\n\nCONCLUSIONS Non-covalent association to SA and covalent binding to c(RGDfK) , synergistically enable the accumulation and prolonged retention of Bchl-Ds in the necrotic regions of tumors .\n\nThese findings provide novel guidelines and strategy for imaging and treatment of necrotic tumors .", "output": "Resisting cell death" }, { "input": "Hepatocellular carcinoma ( HCC ) , one of the most frequent and deadliest cancers , has been increasing considerably in the United States .\n\nIn the absence of a proven effective therapy for HCC , novel chemopreventive strategies are urgently needed to lower the current morbidity and mortality of HCC .\n\nRecently , we have reported that resveratrol , a compound present in grapes and red wine , significantly prevents diethylnitrosamine ( DENA)-induced liver tumorigenesis in rats , although the mechanism of action is not completely understood .\n\nIn the present study , we have examined the underlying mechanisms of resveratrol chemoprevention of hepatocarcinogenesis by investigating the effects of resveratrol on oxidative damage and inflammatory markers during DENA-initiated rat liver carcinogenesis .\n\nThere was a significant increase in hepatic lipid peroxidation and protein oxidation in carcinogen control animals compared with their normal counterparts at the end of the study ( 20 weeks ) .\n\nElevated expressions of inducible nitric oxide synthase and 3-nitrotyrosine were noticed in the livers of the same animals .\n\nDietary resveratrol ( 50-300 mg/kg ) administered throughout the study reversed all the aforementioned markers in a dose-responsive fashion in rats challenged with DENA .\n\nResveratrol also elevated the protein and mRNA expression of hepatic nuclear factor E2-related factor 2 ( Nrf2 ) .\n\nResults of the present investigation provide evidence that attenuation of oxidative stress and suppression of inflammatory response mediated by Nrf2 could be implicated , at least in part , in the chemopreventive effects of this dietary agent against chemically induced hepatic tumorigenesis in rats .\n\nThe outcome of this study may benefit the development of resveratrol in the prevention and intervention of human HCC .", "output": "Tumor promoting inflammation" }, { "input": "Tumor aerobic glycolysis , or the Warburg effect , plays important roles in tumor survival , growth , and metastasis .\n\nPyruvate kinase isoenzyme M2 ( PKM2 ) is a key enzyme that regulates aerobic glycolysis in tumor cells .\n\nRecent research has shown that PKM2 can be used as a tumor marker for diagnosis and , in particular , as a potential target for cancer therapy .\n\nWe investigated the effects of combining shRNA targeting PKM2 and docetaxel on human A549 lung carcinoma cells both in vivo and in vitro .\n\nWe observed that the shRNA can significantly downregulate the expression level of PKM2 .\n\nThe decrease of PKM2 resulted in a decrease in ATP synthesis , which caused intracellular accumulation of docetaxel .\n\nFurthermore , the combination of pshRNA-pkm2 and docetaxel inhibited tumor growth and promoted more cancer cell apoptosis both in vivo and in vitro .\n\nOur findings suggest that targeting tumor glycolysis can increase the efficacy of chemotherapy .\n\nIn particular , the targeting of PKM2 could , to some extent , be a new way of reversing chemotherapy resistance to cancer therapy .", "output": "Cellular energetics" }, { "input": "By intravenous ( but not oral ) application of ascorbate , millimolar serum concentrations can be reached , which are preferentially cytotoxic to cancer cells .\n\nCytotoxicity is mediated by transition metal-dependent generation of H(2)O(2) in the interstitial space .\n\nIn this study , the sensitivity of neuroblastoma cells ( Kelly , SK-N-SH ) to ascorbate and H(2)O(2) and their defense mechanisms against H(2)O(2) were investigated .\n\nSince aerobic glycolysis ( the Warburg effect ) is a feature of many tumour cells , their glucose consumption and lactate production were monitored .\n\nFurthermore , synthesis and release of ferritin by neuroblastoma cells were analysed in order to examine whether ferritin is possibly an iron source for H(2)O(2) generation .\n\nAscorbate ( 0.6-5.0 mM ) and H(2)O(2) ( 25-100 muM ) were found to be similarly cytotoxic to Kelly and SK-N-SH cells .\n\nIn each case , cytotoxicity increased if cell concentrations decreased , in accordance with low cell concentrations having lower capacities to detoxify H(2)O(2) .\n\nKelly and SK-N-SH cells produced and released remarkable amounts of lactate and ferritin .\n\nWe propose the selective cytotoxicity of high dose ascorbate to tumour cells to be due to the preferential generation of H(2)O(2) in the acidic and ferritin-rich tumour microenvironment , combined with reduced defense systems against H(2)O(2) as a consequence of aerobic glycolysis .", "output": "Cellular energetics" }, { "input": "The Ras-assocation domain family ( RASSF ) of tumor suppressor proteins until recently contained six proteins named RASSF1-6 .\n\nRecently , four novel family members , RASSF7-10 , have been identified by homology searches for RA-domain-containing proteins .\n\nThese additional RASSF members are divergent and structurally distinct from RASSF1-6 , containing an N-terminal RA domain and lacking the Sav/RASSF/Hpo ( SARAH ) domain .\n\nHere , we show that RASSF8 is ubiquitously expressed throughout the murine embryo and in normal human adult tissues .\n\nFunctionally , RNAi-mediated knockdown of RASSF8 in non-small-cell lung cancer ( NSCLC ) cell lines , increased anchorage-independent growth in soft agar and enhanced tumor growth in severe combined immunodeficiency ( SCID ) mice .\n\nFurthermore , EdU staining of RASSF8-depleted cells showed growth suppression in a manner dependent on contact inhibition .\n\nWe show that endogenous RASSF8 is not only found in the nucleus , but is also membrane associated at sites of cell-cell adhesion , co-localizing with the adherens junction ( AJ ) component beta-catenin and binding to E-cadherin .\n\nFollowing RASSF8 depletion in two different lung cancer cell lines using alternative small interfering RNA ( siRNA ) sequences , we show that AJs are destabilized and E-cadherin is lost from the cell membrane .\n\nThe AJ components beta-catenin and p65 are also lost from sites of cell-cell contact and are relocalized to the nucleus with a concomitant increase in beta-catenin-dependent and nuclear factor-kappaB ( NF-kappaB)-dependent signaling following RASSF8 depletion .\n\nRASSF8 may also be required to maintain actin -cytoskeletal organization since immunofluorescence analysis shows a striking disorganization of the actin- cytoskeleton following RASSF8 depletion .\n\nAccordingly , scratch wound healing studies show increased cellular migration in RASSF8-deficient cells .\n\nThese results implicate RASSF8 as a tumor suppressor gene that is essential for maintaining AJs function in epithelial cells and have a role in epithelial cell migration .", "output": "Evading growth suppressors" }, { "input": "A 33-year-old man presented with pain and palsy of the leg in 2008 for treatment of hepatocellular carcinoma with huge distant metastases .\n\nThe patient's tumors had slowly enlarged despite several treatments .\n\nOral administration of sorafenib at 800 mg/day with careful observation was commenced in 2009 .\n\nLaboratory investigations on day 7 showed massive tumor lysis .\n\nAn abdominal CT showed multiple low density areas and tumor markers decreased , indicating extended tumor necrosis .\n\nIn conclusion , clinicians should bear in mind not only the published adverse effects , but also massive tumor lysis , when treating patients with large tumor burden by sorafenib .", "output": "Resisting cell death" }, { "input": "Cav-1 ( -/- ) deficient stromal cells are a new genetic model for myofibroblasts and cancer-associated fibroblasts .\n\nUsing an unbiased informatics analysis of the transcriptional profile of Cav-1 ( -/- ) deficient mesenchymal stromal cells , we have now identified many of the major signaling pathways that are activated by a loss of Cav-1 , under conditions of metabolic restriction ( with low glucose media ) .\n\nOur informatics analysis suggests that a loss of Cav-1 induces oxidative stress , which mimics a constitutive pseudo-hypoxic state , leading to ( 1 ) aerobic glycolysis and ( 2 ) inflammation in the tumor stromal microenvironment .\n\nThis occurs via the activation of two major transcription factors , namely HIF ( aerobic glycolysis ) and NF\u03baB ( inflammation ) in Cav-1 ( -/- ) stromal fibroblastic cells .\n\nExperimentally , we show that Cav-1 deficient stromal cells may possess defective mitochondria , due to the over-production of nitric oxide ( NO ) , resulting in the tyrosine nitration of the mitochondrial respiratory chain components ( such as complex I ) .\n\nElevated levels of nitro-tyrosine were observed both in Cav-1 ( -/- ) stromal cells , and via acute knock-down with siRNA targeting Cav-1 .\n\nFinally , metabolic restriction with mitochondrial ( complex I ) and glycolysis inhibitors was synthetically lethal with a Cav-1 ( -/- ) deficiency in mice .\n\nAs such , Cav-1 deficient mice show a dramatically reduced mitochondrial reserve capacity .\n\nThus , a mitochondrial defect in Cav-1 deficient stromal cells could drive oxidative stress , leading to aerobic glycolysis , and inflammation , in the tumor microenvironment .\n\nThese stromal alterations may underlie the molecular basis of the \" reverse Warburg effect \" , and could provide the key to targeted anti-cancer therapies using metabolic inhibitors .\n\nIn direct support of these findings , the transcriptional profile of Cav-1 ( -/- ) stromal cells overlaps significantly with Alzheimer disease , which is characterized by oxidative stress , NO over-production ( peroxynitrite formation ) , inflammation , hypoxia and mitochondrial dysfunction .\n\nWe conclude that Cav-1 ( -/- ) deficient mice are a new whole-body animal model for an activated lethal tumor microenvironment , i.e. , \" tumor stroma \" without the tumor .\n\nSince Cav-1 ( -/- ) mice are also an established animal model for profibrotic disease , our current results may have implications for understanding the pathogenesis of scleroderma ( systemic sclerosis ) and pulmonary fibrosis , which are also related to abnormal mesenchymal stem cell function .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "Overexpression/amplification of human epidermal growth factor receptor ( HER)2/neu ( erbB-2 ) oncogene plays a causal role in carcinogenesis and correlates with a poor clinical prognosis .\n\nHowever , little is known about HER2 in gastric cancer .\n\nIn this study , we explored the pharmacological activities of natural triterpenoid corosolic acid ( CRA ) in HER2 signaling and its role in gastric cancer development and progression .\n\nIn this study , CRA dramatically inhibited HER2 expression in a dose- and time-dependent manner , effectively inhibited cell proliferation , and induced G(0)/G(1) arrest through the induction of p27(kip1) and cyclin D(1) down-regulation .\n\nCRA exposure enhanced apoptotic cell death , as confirmed by caspase-3 and poly ( ADP-ribose ) polymerase cleavage activities .\n\nCRA inhibited signaling pathways downstream of HER2 , including phospho-proteins such as Akt and Erk .\n\nIn addition , CRA combined with adriamycin and 5-fluorouracil enhanced this growth inhibition , but not with docetaxel and paclitaxel .\n\nThese findings demonstrate that CRA suppresses HER2 expression , which in turn promotes cell cycle arrest and apoptotic cell death of gastric cancer cells , providing a rationale for future clinical trials of CRA in the treatment of HER2-positive gastric cancers .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "INTRODUCTION A subpopulation of cancer cells , tumor-initiating cells , is believed to be the driving force behind tumorigenesis and resistance to radiation and chemotherapy .\n\nThe persistence of tumor-initiating cells may depend on altered regulation of DNA damage and checkpoint proteins , as well as a reduced propensity to undergo apoptosis or senescence .\n\nMETHODS To test this hypothesis , we isolated CD24-/low/CD44+ tumor-initiating cells ( as mammospheres ) from MCF-7 breast cancer cells grown in adherent monolayer culture , and carried out a comprehensive comparison of cell death and DNA damage response pathways prior to and after exposure to ionizing radiation in mammospheres and monolayer MCF-7 cells .\n\nSingle and double-strand break repair was measured by single-cell gel electrophoresis .\n\nThe latter was also examined by phosphorylation of histone H2AX and formation of 53BP1 and Rad51 foci .\n\nApoptosis was quantified by flow-cytometric analysis of annexin V-binding and senescence was analyzed on the basis of cellular beta-galactosidase activity .\n\nWe employed the telomeric repeat amplification protocol to quantify telomerase activity .\n\nExpression of key DNA repair and cell cycle regulatory proteins was detected and quantified by western blot analysis .\n\nRESULTS Our data demonstrate that in comparison to the bulk population of MCF-7 cells ( predominantly CD24+/CD44+ ) , the MCF-7 mammosphere cells benefit from a multifaceted approach to cellular protection relative to that seen in monolayer cells , including a reduced level of reactive oxygen species , a more active DNA single-strand break repair ( SSBR ) pathway , possibly due to a higher level of expression of the key SSBR protein , human AP endonuclease 1 ( Ape1 ) , and a significantly reduced propensity to undergo senescence as a result of increased telomerase activity and a low level of p21 protein expression .\n\nNo significant difference was seen in the rates of double-strand break repair ( DSBR ) between the two cell types , but DSBR in mammospheres appears to by-pass the need for H2AX phosphorylation .\n\nCONCLUSIONS Enhanced survival of MCF-7 tumor-initiating cells in response to ionizing radiation is primarily dependent on an inherent down-regulation of the senescence pathway .\n\nSince MCF-7 cells are representative of cancer cells that do not readily undergo apoptosis , consideration of senescence pathways may play a role in targeting stem cells from such tumors .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "BACKGROUND The immune system plays an important role in the multifactorial biologic system during the development of neoplasias .\n\nHowever , the involvement of the inflammatory response in the promotion/control of malignant cells is still controversial , and the cell subsets and the mechanisms involved are poorly investigated .\n\nThe goal of this study was to characterize the clinical-pathological status and the immunophenotyping profile of tumor infiltrating lymphocytes and their association with the animal survival rates in canine mammary carcinomas .\n\nMETHODS Fifty-one animals with mammary carcinomas , classified as carcinomas in mixed tumors-MC-BMT = 31 and carcinomas-MC = 20 were submitted to systematic clinical-pathological analysis ( tumor size ; presence of lymph node and pulmonary metastasis ; clinical stage ; histological grade ; inflammatory distribution and intensity as well as the lymphocytic infiltrate intensity ) and survival rates .\n\nTwenty-four animals ( MC-BMT = 16 and MC = 8 ) were elected to the immunophenotypic study performed by flow cytometry .\n\nRESULTS Data analysis demonstrated that clinical stage II-IV and histological grade was I more frequent in MC-BMT as compared to MC .\n\nUnivariate analysis demonstrated that the intensity of inflammation ( moderate/intense ) and the proportion of CD4+ ( > or = 66.7% ) or CD8+ T-cells ( <33.3% ) were not associated with worse survival rate .\n\nMultivariate analysis demonstrated that only lymphocytic infiltrate intensity > or = 600 ( P = 0.02 ) remained as independent prognostic factor .\n\nDespite the clinical manifestation , the lymphocytes represented the predominant cell type in the tumor infiltrate .\n\nThe percentage of T-cells was higher in animals with MC-BMT without metastasis , while the percentage of B-lymphocytes was greater in animals with metastasized MC-BMT ( P < 0.05 ) .\n\nThe relative percentage of CD4+ T-cells was significantly greater in metastasized tumors ( both MC-BMT and MC ) , ( P < 0.05 ) while the proportion of CD8+ T-cells was higher in MC-BMT without metastasis .\n\nConsequently , the CD4+/CD8+ ratio was significantly increased in both groups with metastasis .\n\nRegardless of the tumor type , the animals with high proportions of CD4+ and low CD8+ T-cells had decreased survival rates .\n\nCONCLUSION The intensity of lymphocytic infiltrate and probably the relative abundance of the CD4+ and CD8+ T-lymphocytes may represent important survival prognostic biomarkers for canine mammary carcinomas .", "output": "Tumor promoting inflammation, Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "INTRODUCTION Anterior-gradient 2 ( AGR2 ) is an estrogen-responsive secreted protein .\n\nIts upregulation has been well documented in a number of cancers , particularly breast cancer , for which mixed data exist on the prognostic implications of AGR2 expression .\n\nAlthough emerging evidence indicates that AGR2 is associated with poor prognosis , its function and impact on cancer-relevant pathways have not been elucidated in breast cancer .\n\nMETHODS To investigate the biologic role of AGR2 in breast cancer , AGR2 was transiently knocked down , by using siRNA , in T47 D and ZR-75-1 ( estrogen receptor-alpha ( ER)-positive ) and MDA-MB-231 and SK-BR-3 ( ER-negative ) human breast cancer cell lines .\n\nThe impact of silencing AGR2 was evaluated in both anchorage-dependent and anchorage-independent growth ( soft agar , spheroid ) assays .\n\nCell-cycle profiles in ER-positive cell lines were determined with BrdU incorporation , and cell death was measured with Annexin V , JC-1 , and F7-26 staining .\n\nAfter transiently silencing AGR2 or stimulating with recombinant AGR2 , modulation of key regulators of growth and survival pathways was assessed with Western blot .\n\nCombination studies of AGR2 knockdown with the antiestrogens tamoxifen and fulvestrant were carried out and assessed at the level of anchorage-dependent growth inhibition and target modulation ( cyclin D1 , ER ) .\n\nRESULTS AGR2 knockdown inhibited growth in anchorage-dependent and anchorage-independent assays , with a more-pronounced effect in ER-positive cell lines .\n\nCyclin D1 levels and BrdU incorporation were reduced with AGR2 knockdown .\n\nConversely , cyclin D1 was induced with recombinant AGR2 .\n\nAGR2 knockdown induced cell death in ZR-75-1 and T47 D cells , and also downregulated survivin and c-Myc .\n\nEvidence of AGR2-ER crosstalk was demonstrated by a reduction of ER at the protein level after transiently silencing AGR2 .\n\nAGR2 knockdown in combination with fulvestrant or tamoxifen did not preclude the efficacy of the antiestrogens , but enhanced it .\n\nIn addition , p-Src , implicated in tamoxifen resistance , was downregulated with AGR2 knockdown .\n\nCONCLUSIONS Transiently silencing AGR2 in ER-positive breast cancer cell lines inhibited cell growth and cell-cycle progression and induced cell death .\n\nBreast cancer drivers ( ER and cyclin D1 ) as well as cancer-signaling nodes ( pSrc , c-Myc , and survivin ) were demonstrated to be downstream of AGR2 .\n\nCollectively , the data presented support the utility of anti-AGR2 therapy in ER-positive breast cancers because of its impact on cancer-relevant pathways .", "output": "Sustaining proliferative signaling" }, { "input": "Autophagy is activated in response to cellular stressors and mediates lysosomal degradation and recycling of cytoplasmic material and organelles as a temporary cell survival mechanism .\n\nDefective autophagy is implicated in human pathology , as disruption of protein and organelle homeostasis enables disease-promoting mechanisms such as toxic protein aggregation , oxidative stress , genomic damage , and inflammation .\n\nWe previously showed that autophagy-defective immortalized mouse mammary epithelial cells are susceptible to metabolic stress , DNA damage , and genomic instability .\n\nWe now report that autophagy deficiency is associated with endoplasmic reticulum ( ER ) and oxidative stress , and with deregulation of p62-mediated keratin homeostasis in mammary cells , allograft tumors , and mammary tissues from genetically engineered mice .\n\nIn human breast tumors , high phospho(Ser73)-K8 levels are inversely correlated with Beclin 1 expression .\n\nThus , autophagy preserves cellular fitness by limiting ER and oxidative stress , a function potentially important in autophagy-mediated suppression of mammary tumorigenesis .\n\nFurthermore , autophagy regulates keratin homeostasis in the mammary gland via a p62-dependent mechanism .\n\nHigh phospho(Ser73)-K8 expression may be a marker of autophagy functional status in breast tumors and , as such , could have therapeutic implications for breast cancer patients .", "output": "Resisting cell death" }, { "input": "Hypoxia is one of the inevitable circumstances of various tumors .\n\nIt controls various levels of regulation in tumor progression and results in tumor resistance to radiotherapy and chemotherapy .\n\nHere we investigated a synthetic TPZ derivative , N-ethoxymethyl-3-amino-1,2,4-benzotriazine-1,4-dioxide ( XQ2 ) , a novel compound that induced anti-cancer effects both in normoxia and in hypoxia , cell proliferation assay found that XQ2 exhibited a potent inhibitory effect on the tested cancer cell lines both in normoxia and in hypoxia .\n\nFlow cytometry and western blot studies indicated that XQ2 induces G2/M arrest and a caspase-dependent apoptosis in A549 cells .\n\nAdditionally , intracellular reactive oxygen species ( ROS ) appear to play a key role in the anticancer effect of XQ2 in hypoxia .\n\nTaken together , our data suggest that XQ2 exerted anticancer action by suppressing the ROS level and triggering cell-cycle arrest and the caspase-dependent pathway , which is associated with apoptosis .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "The deregulation of Met/hepatocyte growth factor ( HGF ) receptor tyrosine kinase signaling constitutes a common event in colorectal cancers .\n\nHowever , the physiopathological functions of such a deregulation remain poorly understood .\n\nIn the present study , we investigated the role of the deregulation of Met receptor in the neoplastic transformation of intestinal epithelial cells .\n\nTo do so , the normal , well-established and characterized rat intestinal epithelial IEC-6 cells were transduced with a retrovirus carrying the oncogenic constitutive active form of Met receptor , Tpr-Met .\n\nHerein , we show that compared with control IEC-6 cells , Tpr-Met-IEC-6 cells exhibit enhanced proliferation , loss of growth-contact inhibition , cell morphological alterations , actin cytoskeletal reorganization , loss of E-cadherin expression and anchorage-independent growth .\n\nMoreover , Tpr-Met-IEC-6 cells are conferred the capacity to produce the proangiogenic factor VEGF and to reduce the potent antiangiogenic factor thrombospondin-1 .\n\nOf significance , Tpr-Met-IEC-6 cells are endowed with the ability to elicit angiogenic responses and to form tumors and metastases in vivo .\n\nHence , our study demonstrates for the first time that the sole oncogenic engagement of Met receptor in normal intestinal epithelial cells is sufficient to induce a wide array of cancerous biological processes that are fundamental to the initiation and malignant progression of colorectal cancers .", "output": "Inducing angiogenesis, Evading growth suppressors" }, { "input": "Magnetite iron nanoparticles have been widely used as contrast agents and in thermal therapy for cancer .\n\nHowever , their adverse effects on human health have not been fully investigated .\n\nIn this study , iron oxide nanoparticles were prepared using inorganic iron chloride ( size : 5.3+/-3.6 nm in phosphate buffered saline , surface charge : 23.14 mV ) , and their inflammatory responses were investigated .\n\nWhen mice were treated with iron oxide nanoparticles ( 250 microg/kg , 500 microg/kg , and 1mg/kg ) by a single intratracheal instillation , the level of intracellular reduced glutathione ( GSH ) was decreased in the cells of bronchoalveolar lavage ( BAL ) fluid .\n\nThe arrest of cell cycles in G1 phase was observed , but S-phase was significantly decreased .\n\nThe concentrations of pro-inflammatory cytokines ( IL-1 , TNF-alpha , and IL-6 ) were dose-dependently increased at day 1 after instillation in the BAL fluid and in the blood .\n\nDuring the experimental period of 28 days , pro-inflammatory cytokines ( IL-1 , TNF-alpha , and IL-6 ) , Th0 cytokine ( IL-2 ) , Th1 type cytokine ( IL-12 ) , Th2 type cytokines ( IL-4 and IL-5 ) , TGF-beta , and IgE were also elevated .\n\nExpressions of many genes related with inflammation or tissue damage such as heat shock protein , matrix metalloproteinase , tissue inhibitors of metalloproteinases , and serum amyloid A were significantly induced .\n\nFormation of microgranuloma , which is one of the indicators for chronic inflammatory response , was observed in the alveolar space .\n\nIn addition , distribution of B cell and CD8+ T cell in blood lymphocytes was increased at day 28 .\n\nBased on the result , iron oxide nanoparticles may subchronic induce inflammatory responses via oxidative stress in mice by a single intratracheal instillation .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Infection by Opisthorchis viverrini , a risk factor for cholangiocarcinoma ( CCA ) may act through chronic inflammation , oxidative stress and lipid peroxidation ( LPO)-related damage and growth stimuli. 1,N6-etheno-2'-deoxyadenosine ( epsilondA ) , and 3,N4-etheno-2'-deoxycytidine ( epsilondC ) , markers for LPO-derived DNA damage were highly increased in white blood cell and urine of O. viverrini-infected Thai patients .\n\nIn order to investigate tissue specificity etheno adducts were measured in a cholangiocarcinogenesis model , in O. viverrini-infected hamsters that had received N-nitrosodimethylamine ( NDMA , 12.5 ppm in dw ) for 2 months. epsilondA- and epsilondC-levels were analyzed in paraffin-embedded liver sections by a novel immunohistochemical method , from 21 up to 180 days post-O. viverrini-infection .\n\nIn inflamed areas of the liver , etheno adducts were localized in the nuclei of inflammatory cells and in the epithelial lining of the bile duct .\n\nSemi-quantitative image analysis showed higher adduct levels in the liver of O. viverrini-infected hamsters , treated with or w/o NDMA when compared with untreated controls .\n\nLevels were found highest in the liver of O. viverrini-infected plus NDMA-treated hamsters .\n\nAdducts increased in an age-dependent manner from O. viverrini-infection until CCA development .\n\nIncreased adduct formation paralleled histopathological changes in plasma alkaline phosphatase ( ALP ) activity , bile duct hyperplasia , dysplasia , precancerous lesions , and CCA appearance .\n\nAlso elevated expression of alkyladenine DNA glycosylase ( AAG ) , which excises 1,N6-ethenoadenine ( epsilonA ) was linked to higher adduct formation , suggesting imbalanced repair .\n\nOur results implicate accumulation of inflammation-related , promutagenic DNA damage in target tissue and possibly imbalanced repair in the onset of cholangiocarcinogenesis .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Contact inhibition is the process by which cells switch from a motile growing state to a passive and stabilized state upon touching their neighbors .\n\nWhen two cells touch , an adhesion link is created between them by means of transmembrane E-cadherin proteins .\n\nSimultaneously , their actin filaments stop polymerizing in the direction perpendicular to the membrane and reorganize to create an apical belt that colocalizes with the adhesion links .\n\nHere , we propose a detailed quantitative model of the role of cytoplasmic beta-catenin and alpha-catenin proteins in this process , treated as a reaction-diffusion system .\n\nUpon cell-cell contact the concentration in alpha-catenin dimers increases , inhibiting actin branching and thereby reducing cellular motility and expansion pressure .\n\nThis model provides a mechanism for contact inhibition that could explain previously unrelated experimental findings on the role played by E-cadherin , beta-catenin , and alpha-catenin in the cellular phenotype and in tumorigenesis .\n\nIn particular , we address the effect of a knockout of the adenomatous polyposis coli tumor suppressor gene .\n\nPotential direct tests of our model are discussed .", "output": "Evading growth suppressors" }, { "input": "Pancreatic adenocarcinoma is an aggressive cancer with a greater than 95% mortality rate and short survival after diagnosis .\n\nChemotherapeutic resistance hinders successful treatment .\n\nThis resistance is often associated with mutations in codon 12 of the K-Ras gene ( K-Ras 12 ) , which is present in over 90% of all pancreatic adenocarcinomas .\n\nCodon 12 mutations maintain Ras in a constitutively active state leading to continuous cellular proliferation .\n\nOur study determined if TRAIL resistance in pancreatic adenocarcinomas with K-Ras 12 mutations could be overcome by first sensitizing the cells with Benzyl isothiocyanate ( BITC ) .\n\nBITC is a component of cruciferous vegetables and a cell cycle inhibitor .\n\nBxPC3 , MiaPaCa2 and Panc-1 human pancreatic adenocarcinoma cell lines were examined for TRAIL resistance .\n\nOur studies show BITC induced TRAIL sensitization by dual activation of both the extrinsic and intrinsic apoptotic pathways .", "output": "Resisting cell death" }, { "input": "Proliferation of non-transformed cells is regulated by cell-cell contacts , which are referred to as contact-inhibition .\n\nVice versa , transformed cells are characterised by a loss of contact-inhibition .\n\nDespite its generally accepted importance for cell-cycle control , little is known about the intracellular signalling pathways involved in contact-inhibition .\n\nUnravelling the molecular mechanisms of contact-inhibition and its loss during tumourigenesis will be an important step towards the identification of novel target genes in tumour diagnosis and treatment .\n\nTo better understand the underlying molecular mechanisms we identified the transcriptional programme of contact-inhibition in NIH3T3 fibroblast using high-density microarrays .\n\nSetting the cut off : >or=1.5-fold , P or=2-fold , P 0.05 ) and Cty-c protein expression as well ( P > 0.05 ) .\n\nHowever , Nec-1 could reduce Caspase-8 activity significantly ( P < 0.05 , P < 0.01 ) and increase NF-kappa B protein expression ( P < 0.05 , P < 0.01 ) and finally decrease Caspase-3 activity ( P < 0.05 ) .\n\nCONCLUSION Nec-1 could reduce cell apoptosis induced by Al , through Caspase-8 pathway , and up-regulate the expression of NF-kappa B protein .", "output": "Resisting cell death" }, { "input": "Bisphosphonates ( BPs ) are the current standard of care for bone lesions in patients with multiple myeloma ( MM ) but they are associated with a number of side effects such as osteonecrosis of the jaw .\n\nThe exact mechanisms of osteonecrosis are not elucidated , and its physiopathology is based on several hypotheses such as a decrease in bone remodeling or an inhibitory effect on angiogenesis .\n\nThe aim of our study was to investigate the mechanism involved in the pathogenesis of osteonecrosis .\n\nWe examined the apoptosis of circulating endothelial progenitor cells in MM subjects before and after BP treatment and in osteonecrosis patients using a flow-cytometric analysis .\n\nOur data showed an increase in endothelial cell apoptosis in MM patients after BP administration and in osteonecrosis subjects .\n\nOur study seems in agreement with the hypothesis that BPs can inhibit angiogenesis interfering with endothelial cell proliferation and survival , leading to loss of blood vessels and avascular necrosis .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "Indoleamine 2,3-dioxygenase ( IDO ) is generally considered to be immunosuppressive but recent findings suggest this characterization oversimplifies its role in disease pathogenesis .\n\nRecently , we showed that IDO is essential for tumor outgrowth in the classical two-stage model of inflammatory skin carcinogenesis .\n\nHere , we report that IDO loss did not exacerbate classical inflammatory responses .\n\nRather , IDO induction could be elicited by environmental signals and tumor promoters as an integral component of the inflammatory tissue microenvironment even in the absence of cancer .\n\nIDO loss had limited impact on tumor outgrowth in carcinogenesis models that lacked an explicit inflammatory tumor promoter .\n\nIn the context of inflammatory carcinogenesis where IDO was critical to tumor development , the most important source of IDO was radiation-resistant non-hematopoietic cells , consistent with evidence that loss of the IDO regulatory tumor suppressor gene Bin1 in transformed skin cells facilitates IDO-mediated immune escape by a cell autonomous mechanism .\n\nTaken together , our results identify IDO as an integral component of ' cancer-associated ' inflammation that tilts the immune system toward tumor support .\n\nMore generally , they promote the concept that mediators of immune escape and cancer-associated inflammation may be genetically synonymous .", "output": "Tumor promoting inflammation" }, { "input": "PURPOSE Overproduction of reactive oxygen species ( ROS ) intermediates above the functional capability of cellular antioxidants may result in instability of important macromolecules and represents the molecular basis of many diseases including inflammation processes , cardiovascular alterations , cancer etc .\n\nThe purpose of this study was to determine plasma level of superoxide anion , hydrogen-peroxide and malondialdehyde ( MDA ) as markers of oxidative stress and activities of superoxide dismutase ( SOD ) , catalase ( CAT ) and glutathione peroxidase ( GPx ) as antioxidant enzymes in B-chronic lymphocytic leukemia ( B-CLL ) patients .\n\nMETHODS The study included 29 untreated B-CLL patients in stage A , and 21 in stages B and C , classified according to the Binet system ; 31 healthy volunteers formed the control group .\n\nAfter centrifugation of heparinized peripheral blood , plasma levels of all investigated parameters were determined using spectrophotometric methods .\n\nRESULTS Plasma CAT activity was increased in B-CLL patients compared with control subjects ; also , progression of disease was related with significantly higher plasma activity of CAT .\n\nAlso , B-CLL patients showed significantly higher plasma concentration of MDA compared with controls .\n\nNo statistically significant differences of superoxide anion and hydrogen peroxide as well as plasma activity of SOD and GPx between the tested groups were noted .\n\nCONCLUSION Increase of CAT activity in B-CLL patients indicates that there is stimulation of the antioxidant enzyme system , while the increase of MDA concentration shows increased lipid peroxidation level .\n\nAccording to these results it could be concluded that an imbalance exists between oxidants and antioxidants in the plasma of B-CLL patients .", "output": "Tumor promoting inflammation" }, { "input": "The biological activities of tocotrienols are receiving increasing attention .\n\nHerein , we report the efficacy of a mixed-tocotrienol diet against prostate tumorigenesis in the transgenic adenocarcinoma mouse prostate ( TRAMP ) mouse model .\n\nMale TRAMP mice , 8 wk old , were fed 0.1% , 0.3% , or 1% mixed tocotrienols in AIN-76A diet up to 24 wk old .\n\nLikewise , a positive control group consisting of male TRAMP mice and a negative control group consisting of wild-type nontransgenic mice were fed regular AIN-76A diet up to 24 wk old .\n\nOur results show that mixed-tocotrienol-fed groups had a lower incidence of tumor formation along with a significant reduction in the average wet weight of genitourinary apparatus .\n\nFurthermore , mixed tocotrienols significantly reduced the levels of high-grade neoplastic lesions as compared to the positive controls .\n\nThis decrease in levels of high-grade neoplastic lesions was found to be associated with increased expression of proapoptotic proteins BAD ( Bcl(2) antagonist of cell death ) and cleaved caspase-3 and cell cycle regulatory proteins cyclin dependent kinase inhibitors p21 and p27 .\n\nIn contrast , the expression of cyclins A and E were found to be decreased in mixed-tocotrienol groups .\n\nTaken together , our results show that by modulating cell cycle regulatory proteins and increasing expression of proapoptotic proteins , mixed tocotrienols suppress prostate tumorigenesis in the TRAMP mice .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Cellular senescence is a mechanism to inhibit the growth of mammalian cells after oncogenic activation , or in response to damage or stress .\n\nWe describe here the identification of a novel gene , SENEX , that regulates stress induced premature senescence pathways in endothelial cells ( ECs ) involving p16(INK4a) and retinoblastoma protein activation .\n\nEndogenous levels of SENEX remain unchanged during replicative senescence but are regulated by H(2)O(2)-mediated stress .\n\nIn contrast to that previously described for senescence in other cell types , the SENEX induced senescent ECs are profoundly anti-inflammatory .\n\nThe cells are resistant to tumor necrosis factor ( TNF)\u03b1-induced apoptosis , adhesion of neutrophils and mononuclear cells , and the surface ( but not cytoplasmic ) expression of endothelial leukocyte adhesion molecule 1 and vascular cell adhesion molecule 1 .\n\nFurthermore they are resistant to thrombin induced vascular leak .\n\nSenescent ECs such as those lining atherosclerotic lesions may therefore function to limit the inflammatory response .\n\nSENEX is also essential for EC survival since depletion either ectopically by siRNA or by high- dose H(2)O(2) treatment causes apoptosis .\n\nTogether , these findings expand our understanding of the role of senescence in the vasculature and identify SENEX as a fulcrum for driving the resultant phenotype of the endothelium after activation .", "output": "Tumor promoting inflammation, Enabling replicative immortality, Resisting cell death" }, { "input": "AIMS To screen the genes and possible signal transduction pathways with which nucleostemin ( NS ) interacts and explore the mechanism of NS in prostate cancer .\n\nMETHODS NS-specific short-hairpin RNA expression plasmid was used to downregulate the NS level in PC-3 cells and the changes of cell cycle were studied .\n\nAfter that , oligonucleotide DNA microarray was used to screen the genome changes in PC-3 cells and quantitative real-time PCR was used to further confirm the differentially expressed genes .\n\nRESULTS Detection of cell cycle showed a decrease of S stage and an increase of G1 stage after downregulation of NS. 219 differentially expressed genes were found and these genes were involved in cell cycle , cell proliferation , signal transduction , cell apoptosis and cell differentiation , and so on .\n\nGenes related to cell cycle were discussed emphatically .\n\nINK4 family genes ( P15 , P16 , P18 ) were upregulated while cyclin D1 HDAC1 were downregulated .\n\nThese genes were tightly related to CDK4/6-cyclin D and pRb-E2F1 complexes .\n\nCONCLUSION NS is an important G1/S checkpoint regulator and it could regulate cell cycles via a p53-independent pathway in prostate cancer .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "AIM To compare the value of intravenous contrast-enhanced ultrasonography ( US ) , intravenous contrast-enhanced computed tomography ( CT ) , and magnetic resonance imaging ( MRI ) in the diagnosis of hepatic hemangiomas .\n\nMATERIAL AND METHODS The study enrolled 48 patients , aged between 20 and 79 years ( 35 [ 72.9% ] women , 13 [ 27.1% ] men ; mean age , 53.5+/-12.855 years ) , who were examined and treated in the Departments of Gastroenterology , Surgery , and Oncology , Hospital of Kaunas University of Medicine , in the year 2007 .\n\nAll patients underwent intravenous contrast-enhanced US , intravenous contrast-enhanced CT , and MRI and were diagnosed with hepatic hemangioma according to the findings of these examinations .\n\nRESULTS The size of hemangiomas was < or =2.0 cm in 20 cases ( 41.7% ) and >2.0 cm in 28 ( 58.3% ) .\n\nNo association between hepatic hemangioma and patient's age was found ( chi(2)=0.547 , df=2 , P=0.761 ) .\n\nNearly one-third of hemangiomas were located in the segment IV of the left hepatic lobe .\n\nThere were a few complicated hemangiomas in the study sample : 2 with calcification and 1 with necrosis .\n\nThe sensitivity of CT in the diagnosis of hepatic hemangioma was 76.92% ; specificity , 33.3% ; positive prognostic value , 83.3% ; and negative prognostic value , 25.0% .\n\nThe sensitivity of intravenous contrast-enhanced US in the diagnosis of hepatic hemangioma was 77.8% ; specificity , 100% ; positive prognostic value , 100% ; and negative prognostic value , 23.1% .\n\nCONCLUSIONS Intravenous contrast-enhanced US is more specific than intravenous contrast-enhanced CT in the diagnosis of hepatic hemangioma ( P=0.0005 ) and has a higher positive prognostic value ( P=0.001 ) .", "output": "Resisting cell death" }, { "input": "A novel compound 2-arylidene-4,7-dimethyl indan-1-one synthesized was screened for anticancer effect against the human breast adenocarcinoma cell line , MCF-7 .\n\nAn IC50 value of > or = 80 microM , nontoxic to the normal breast cell line HBL-100 , showed complete inhibition of the MCF-7 cells .\n\nAnalysis of mechanisms showed nuclear fragmentation and DNA laddering in gel electrophoresis .\n\nGSH and GR levels were found to be reduced after the compound treatment .\n\nCell cycle analysis using fluorescent cytometry revealed G2/M phase arrest , which indicates the compound deserves consideration for further studies against cancer .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Radiation-induced carcinogenesis is a major concern both for astronauts on long-term space missions and for cancer patients being treated with therapeutic radiation .\n\nExposure to radiation induces oxidative stress and chronic inflammation , which are critical initiators and promoters of carcinogenesis .\n\nMany studies have demonstrated that non-steroidal anti-inflammatory drugs and antioxidants can reduce the risk of radiation-induced cancer .\n\nIn this study , we found that a synthetic triterpenoid , CDDO-Me ( bardoxolone methyl ) , was able to protect human colon epithelial cells ( HCECs ) against radiation-induced transformation .\n\nHCECs that were immortalized by ectopic expression of hTERT and cdk4 and exhibit trisomy for chromosome 7 ( a non-random chromosome change that occurs in 37% of premalignant colon adenomas ) can be transformed experimentally with one combined exposure to 2 Gy of protons at 1 GeV/nucleon followed 24 h later by 50 cGy of ( 56)Fe ions at 1 GeV/nucleon .\n\nTransformed cells showed an increase in proliferation rate and in both anchorage-dependent and independent colony formation ability .\n\nA spectrum of chromosome aberrations was observed in transformed cells , with 40% showing loss of 17p ( e.g. loss of one copy of p53 ) .\n\nPretreatment of cells with pharmacological doses of CDDO-Me , which has been shown to induce antioxidative as well as anti-inflammatory responses , prevented the heavy-ion-induced increase in proliferation rate and anchorage-dependent and independent colony formation efficiencies .\n\nTaken together , these results demonstrate that experimentally immortalized human colon epithelial cells with a non-random chromosome 7 trisomy are valuable premalignant cellular reagents that can be used to study radiation-induced colorectal carcinogenesis .\n\nThe utility of premalignant HCECs to test novel compounds such as CDDO-Me that can be used to protect against radiation-induced neoplastic transformation is also demonstrated .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "INTRODUCTION The homeobox-containing transcription factor muscle segment homeobox 2 ( Msx2 ) plays an important role in mammary gland development .\n\nHowever , the clinical implications of Msx2 expression in breast cancer are unclear .\n\nThe aims of this study were to investigate the potential clinical value of Msx2 as a breast cancer biomarker and to clarify its functional role in vitro .\n\nMETHODS Msx2 gene expression was first examined in a well-validated breast cancer transcriptomic dataset of 295 patients .\n\nMsx2 protein expression was then evaluated by immunohistochemistry in a tissue microarray ( TMA ) containing 281 invasive breast tumours .\n\nFinally , to assess the functional role of Msx2 in vitro , Msx2 was ectopically expressed in a highly invasive breast tumour cell line ( MDA-MB-231 ) and an immortalised breast cell line ( MCF10a ) , and these cell lines were examined for changes in growth rate , cell death and cell signalling .\n\nRESULTS Examination of Msx2 mRNA expression in a breast cancer transcriptomic dataset demonstrated that increased levels of Msx2 were associated with good prognosis ( P = 0.011 ) .\n\nEvaluation of Msx2 protein expression on a TMA revealed that Msx2 was detectable in both tumour cell nuclei and cytoplasm .\n\nCytoplasmic Msx2 expression was associated with low grade tumours ( P = 0.012 ) and Ki67 negativity ( P = 0.018 ) .\n\nNuclear Msx2 correlated with low-grade tumours ( P = 0.015 ) , estrogen receptor positivity ( P = 0.038 ) , low Ki67 ( P = 0.005 ) and high cyclin D1 expression ( P = 0.037 ) .\n\nIncreased cytoplasmic Msx2 expression was associated with a prolonged breast cancer-specific survival ( P = 0.049 ) , recurrence-free survival ( P = 0.029 ) and overall survival ( P = 0.019 ) .\n\nEctopic expression of Msx2 in breast cell lines resulted in radically decreased cell viability mediated by induction of cell death via apoptosis .\n\nFurther analysis of Msx2-expressing cells revealed increased levels of p21 and phosphorylated extracellular signal-regulated kinase ( ERK ) and decreased levels of Survivin and the ' split ends ' ( SPEN ) protein family member RBM15 .\n\nCONCLUSIONS We conclude that increased Msx2 expression results in improved outcome for breast cancer patients , possibly by increasing the likelihood of tumour cell death by apoptosis .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "The NOTCH signaling pathway plays important role in the development of multicellular organisms , as it regulates cell proliferation , survival , and differentiation .\n\nIn adults , it is essential for the T- or B-lymphocyte lineage commitment .\n\nNOTCH1 and FBXW7 mutations both lead the activation of the NOTCH1 pathway and are found in the majority of T-ALL patients .\n\nIn this study , the mutation analysis of NOTCH1 and FBXW7 genes was performed in 87 pediatric T-ALLs who were treated on the ALL-BFM protocols .\n\nIn 19 patients ( 22% ) , activating NOTCH1 mutations were observed either in the heterodimerization domain or in the PEST domain and 7 cases ( 10% ) demonstrated FBXW7 mutations ( 2 cases had both NOTCH1 and FBXW7 mutations ) .\n\nWe also analyzed the relationship of the mutation data between the clinical and biological data of the patients .\n\nNOTCH1 and FBXW7 , NOTCH1 alone were found correlated with lower initial leucocyte counts which was independent from the sex and T- cell immunophenotype .\n\nHowever , NOTCH1 and FBXW7 mutations were not predictive of outcome in the overall cohort of pediatric T-ALLs .", "output": "Genomic instability and mutation" }, { "input": "This study aimed to investigate the mechanism by which the human lung cancer drug resistance-related gene BC006151 regulates chemosensitivity by down-regulating BC006151 expression via antisense gene transfer in H446/(C)DDP cells .\n\nA retroviral vector containing the antisense BC006151 sequence was constructed and transfected into H446/(C)DDP cells .\n\nTransfection of the empty vector served as a negative control .\n\nThe two groups of transfected cells were treated with various chemotherapeutic agents , after which morphological changes in cell ultrastructure were compared by transmission electron microscopy , cell proliferation and chemosensitivity to particular chemotherapeutic agents were compared by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method , the effects of chemotherapy on cell cycle and apoptosis were compared by flow cytometry , and Bcl-2 was evaluated by immunohistochemistry and Western blot analysis .\n\nResults showed that apoptotic body-like structures were identified by transmission electron microscopy in the antisense gene-transfected cells .\n\nMTT founded that these cells exhibited a significantly lower level of proliferation than the control cells ( p<0.01 ) , together with a markedly increased sensitivity to various chemotherapeutic agents ( p<0.01 ) .\n\nFlow cytometry analysis revealed that a G1 phase arrest accounted for the reduction in proliferation in the antisense gene-transfected cells ; increased apoptosis was also detected ( p<0.01 ) .\n\nBoth immunohistochemistry and western blot analysis confirmed that Bcl-2 expression was significantly down-regulated in the antisense gene-transfected cells compared to controls .\n\nIn a word , down-regulation of BC006151 can significantly inhibit proliferation and increase apoptosis of H446/(C)DDP cells after chemotherapy , and this gene may play an important role in the development of multidrug resistance in lung cancer .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "The expression of \" growth arrest and DNA damage inducible genes 45 and 153 \" is related to apoptotic induction of cells .\n\nGADD45 is an effector gene of the tumor suppressor p53 , and GADD153 is associated with cellular function of cancer prevention .\n\nCurcumin , isolated from the plant Curcuma longa ( LINN ) , has been investigated as a promising cancer preventive in food because curcumin , a phenolic and coloring compound , is widely ingested in the Indian subcontinent .\n\nHowever , the exact mechanisms of action of curcumin have not yet been clearly elucidated .\n\nBased on our successful results with green tea catechins as cancer preventive , we studied the relationship between the expression of GADD45 and 153 and apoptotic induction in human lung cancer cell line PC-9 .\n\nIn our study curcumin increased the expression of GADD45 and 153 in a p53-independent manner .\n\nCurcumin also inhibited the growth of PC-9 cells and induced G(1)/S arrest of the cell-cycle followed by strong induction of apoptosis .\n\nTreatment with GADD45 and 153 small interfering RNAs ( siRNAs ) inhibited the apoptotic induction in PC-9 cells by curcumin .\n\nMoreover , curcumin induced the expression of cyclin dependent kinase inhibitor genes p21 and p27 , while it inhibited the expression of numerous genes , including Bcl-2 , cyclin D1 , CDK2 , CDK4 and CDK6 .\n\nAll the results with PC-9 cells suggest that the up-regulation of GADD45 and 153 by curcumin is a prime mechanism in the anticancer activity of curcumin .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Genipin is a metabolite of geniposide isolated from an extract of Gardenia fructus .\n\nSome observations suggested that genipin could induce cell apoptosis in hepatoma cells and PC3 human prostate cancer cells .\n\nHowever , the effects of genipin on HeLa human cervical carcinoma cells are still unknown .\n\nIn this study , we provided evidences that genipin induced the death of HeLa cells through apoptotic pathway in a dose-dependent manner .\n\nGenipin could remarkably induce cytotoxicity in HeLa cells and inhibit its proliferation .\n\nInduction of the apoptosis by genipin was confirmed by analysis of DNA fragmentation and induction of sub-G(1) peak through flow cytometry .\n\nThe results also showed that genipin-treated HeLa cells cycle was arrested at G(1) phase .\n\nWestern blot analysis revealed that the phosphorylated c-Jun NH(2)-terminal kinase ( JNK ) protein , phospho-Jun protein , p53 protein and bax protein significantly increased in a dose-dependent manner after treatment of genipin for 24 h , and to our knowledge , the activation of JNK maybe result in the increase of the p53 protein level , and the increase of the p53 protein led to the accumulation of bax protein , bax protein further induced cell apoptotic death eventually .\n\nTaken all these together , it is possible to develop genipin as an anti-cancer drug .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "The authors examined nutritional risk factors for prostate cancer among 9,559 participants in the Prostate Cancer Prevention Trial ( United States and Canada , 1994-2003 ) .\n\nThe presence or absence of cancer was determined by prostate biopsy , which was recommended during the trial because of an elevated prostate-specific antigen level or an abnormal digital rectal examination and was offered to all men at the trial's end .\n\nNutrient intake was assessed using a food frequency questionnaire and a structured supplement-use questionnaire .\n\nCancer was detected in 1,703 men ; 127 cancers were high-grade ( Gleason score 8-10 ) .\n\nThere were no associations of any nutrient or supplement with prostate cancer risk overall .\n\nRisk of high-grade cancer was associated with high intake of polyunsaturated fats ( quartile 4 vs. quartile 1 : odds ratio = 2.41 , 95% confidence interval ( CI ) : 1.33 , 4.38 ) .\n\nDietary calcium was positively associated with low-grade cancer but inversely associated with high-grade cancer ( for quartile 4 vs. quartile 1 , odds ratios were 1.27 ( 95% CI : 1.02 , 1.57 ) and 0.43 ( 95% CI : 0.21 , 0.89 ) , respectively ) .\n\nNeither dietary nor supplemental intakes of nutrients often suggested for prostate cancer prevention , including lycopene , long-chain n-3 fatty acids , vitamin D , vitamin E , and selenium , were significantly associated with cancer risk .\n\nHigh intake of n-6 fatty acids , through their effects on inflammation and oxidative stress , may increase prostate cancer risk .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND The most deadly form of cancer is not lung or colon , breast or prostate ; it is any cancer that has become metastatic .\n\nMortality due to metastatic melanoma , one of the most aggressive and deadly cancers , has increased steadily over the last several decades .\n\nUnfortunately , the arsenal of chemotherapeutic agents available today is most often unsuccessful at extending and improving the life expectancy of afflicted individuals .\n\nWe sought to identify an effective and nontoxic agent against metastatic melanoma .\n\nMETHODOLOGY/PRINCIPAL FINDINGS We chose to study Cloudman S-91 mouse melanoma cells ( sub-clone M3 , CCL53.1 ) because these cells are highly aggressive and metastatic , representing one of the deadliest types of cancer .\n\nMelanoma cells also had an experimental advantage because their morphology , which is easily monitored , relates to the physiology of metastatic cells and normal melanocytes .\n\nWe chose to test methyl sulfone as a chemotherapeutic agent for two reasons .\n\nBecause of its chemical structure , we speculated a potential anti-cancer activity by targeting microtubules .\n\nEqually important , methyl sulfone has a well-established safety profile in humans .\n\nSurprisingly , we found that malignant melanoma cells exposed to methyl sulfone demonstrated the loss of phenotypes characteristic of malignant cells , and the reemergence of phenotypes characteristic of healthy melanocytes .\n\nBriefly , over time methyl sulfone induced contact inhibition , loss of ability to migrate through an extracellular matrix , loss of anchorage-independent growth , proper wound healing followed by contact inhibition , irreversible senescence followed by arborization with melanosomes in arbors as seen in normal melanocytes .\n\nCONCLUSIONS/SIGNIFICANCE Methyl sulfone may have clinical potential as a non-toxic agent effective against metastatic melanoma .\n\nAdditionally , methyl sulfone has promise as a tool to explore molecular mechanisms of metastatic transformation as well as fundamental processes such as cell migration , contact inhibition , wound healing and cellular senescence .", "output": "Enabling replicative immortality, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "INTRODUCTION HER2 and estrogen receptor ( ER ) are important in breast cancer and are therapeutic targets of trastuzumab ( Herceptin ) and tamoxifen , respectively .\n\nRetinoids inhibit breast cancer growth , and modulate signaling by HER2 and ER .\n\nWe hypothesized that treatment with retinoids and simultaneous targeting of HER2 and/or ER may have enhanced anti-tumor effects .\n\nMETHODS The effects of retinoids combined with trastuzumab or tamoxifen were examined in two human breast cancer cell lines in culture , BT474 and SKBR3 .\n\nAssays of proliferation , apoptosis , differentiation , cell cycle distribution , and receptor signaling were performed .\n\nRESULTS In HER2-overexpressing/ER-positive BT474 cells , combining all-trans retinoic acid ( atRA ) with tamoxifen or trastuzumab synergistically inhibited cell growth , and altered cell differentiation and cell cycle .\n\nOnly atRA/trastuzumab-containing combinations induced apoptosis .\n\nBT474 and HER2-overexpressing/ER-negative SKBR3 cells were treated with a panel of retinoids ( atRA , 9-cis-retinoic acid , 13-cis-retinoic acid , or N-(4-hydroxyphenyl) retinamide ( fenretinide ) ( 4-HPR) ) combined with trastuzumab .\n\nIn BT474 cells , none of the single agents except 4-HPR induced apoptosis , but again combinations of each retinoid with trastuzumab did induce apoptosis .\n\nIn contrast , the single retinoid agents did cause apoptosis in SKBR3 cells ; this was only modestly enhanced by addition of trastuzumab .\n\nThe retinoid drug combinations altered signaling by HER2 and ER .\n\nRetinoids were inactive in trastuzumab-resistant BT474 cells .\n\nCONCLUSIONS Combining retinoids with trastuzumab maximally inhibits cell growth and induces apoptosis in trastuzumab-sensitive cells .\n\nTreatment with such combinations may have benefit for breast cancer patients .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND/AIMS by reducing the number of ATP molecules produced via aerobic glycolysis , the inhibition of lactic dehydrogenase ( LDH ) should hinder the growth of neoplastic cells without damaging the normal cells which do not rely on this metabolic pathway for their energetic needs .\n\nHere , we studied the effect of oxamic and tartronic acids , 2 inhibitors of LDH , on aerobic glycolysis and cell replication of HepG2 and PLC/PRF/5 cells , 2 lines from human hepatocellular carcinomas .\n\nMETHODS aerobic glycolysis was measured by calculating the amounts of lactic acid formed .\n\nThe effect on replication was assessed by culturing the cells in both standard conditions and glucose-deprived medium , which was used to shut down aerobic glycolysis .\n\nRESULTS the oxamic and tartronic acids inhibited aerobic glycolysis , impaired the growth of both cell lines and also induced an increased expression of p53-upregulated modulator of apoptosis , a signal of cell death .\n\nA strong impairment of cell replication by oxamic acid was only found when the cells were cultured in the presence of glucose , indicating that it was for the most part owing to inhibition of aerobic glycolysis .\n\nCONCLUSIONS inhibition of aerobic glycolysis achieved by blocking LDH could be useful in the treatment of human hepatocellular carcinomas .\n\nWithout interfering with glucose metabolism in normal cells , it could hinder cell growth by itself and could also enhance the chemotherapeutic index of associated anticancer agents by decreasing the levels of ATP selectively in neoplastic cells .", "output": "Cellular energetics" }, { "input": "BACKGROUND The Mutyh DNA glycosylase is involved in the repair of oxidized DNA bases .\n\nMutations in the human MUTYH gene are responsible for colorectal cancer in familial adenomatous polyposis .\n\nSince defective DNA repair genes might contribute to the increased cancer risk associated with inflammatory bowel diseases , we compared the inflammatory response of wild-type and Mutyh(-/-) mice to oxidative stress .\n\nMETHODOLOGY/PRINCIPAL FINDINGS The severity of colitis , changes in expression of genes involved in DNA repair and inflammation , DNA 8-oxoguanine levels and microsatellite instability were analysed in colon of mice treated with dextran sulfate sodium ( DSS ) .\n\nThe Mutyh(-/-) phenotype was associated with a significant accumulation of 8-oxoguanine in colon DNA of treated mice .\n\nA single DSS cycle induced severe acute ulcerative colitis in wild-type mice , whereas lesions were modest in Mutyh(-/-) mice , and this was associated with moderate variations in the expression of several cytokines .\n\nEight DSS cycles caused chronic colitis in both wild-type and Mutyh(-/-) mice .\n\nLymphoid hyperplasia and a significant reduction in Foxp3(+) regulatory T cells were observed only in Mutyh(-/-) mice .\n\nCONCLUSIONS The findings indicate that , in this model of ulcerative colitis , Mutyh plays a major role in maintaining intestinal integrity by affecting the inflammatory response .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "PURPOSE The cytotoxic effect of the combination treatment of TNF-alpha and hyperthermia on L929 and TNF-alpha-resistant L929 ( rL929 ) cells was investigated .\n\nMATERIALS AND METHODS L929 cells were treated with TNF-alpha ( 5 ng/mL ) , heating at 43 degrees C or the combination of TNF-alpha and heating .\n\nThe cells were harvested at different time within the 24-hour period .\n\nThe viability and the type of cell death of the harvested cells were examined .\n\nRESULTS When L929 cells were treated with a combination of TNF-alpha and heating the cells died quickly and apoptosis increased to an overwhelming extent , especially in the group pre-treated with TNF-alpha for 1 h prior to heating .\n\nAlthough rL929 cells were resistant to TNF-alpha alone , the cells became sensitive to TNF-alpha treatment when combined with heating .\n\nSimilar to the L929 cell , the cells also died rapidly and exhibited apoptosis to a higher extent .\n\nUsing an Annexin-V-FITC kit and flow cytometer , we found that both necrosis and apoptosis occurred .\n\nAgarose gel electrophoresis of DNA extracted from treated cells showed that the DNA fragments were multiples of approximately 200 bp .\n\nFurthermore , by studying the kinetics of cell death and apoptosis , we found that the loss of cell membrane integrity preceded the DNA fragmentation in both L929 and rL929 cells .\n\nCONCLUSION The results suggested that hyperthermia may enhance the necrotic and apoptotic effects of TNF-alpha on some tumour cells and overcome the resistance of some tumour cells to TNF-alpha .", "output": "Resisting cell death" }, { "input": "Chromosomal instability and the subsequent genetic mutations are considered to be critical factors in the development of the majority of solid tumors , but the mechanisms by which a stable diploid cell loses the ability to maintain genomic integrity are not well characterized .\n\nWe have approached this critical issue through the use of high-throughput screens in untransformed diploid epithelial cells .\n\nIn a screen of a cDNA library , we identified 13 kinases whose overexpression leads to increased ploidy .\n\nIn a series of shRNA screens , we identified 16 kinases whose loss leads to increased ploidy .\n\nIn both cDNA and shRNA screens , the majority of hits have not been linked previously to genomic stability .\n\nWe further show that sustained loss of the shRNA screening hits leads to multipolar spindles and heterogeneous chromosome content , two characteristics of chromosomal instability .\n\nLoss of several of the kinases leads to loss of contact inhibition and to anchorage-independent growth , vital traits acquired during tumor development .\n\nWe anticipate that this work will serve as a template for the comprehensive identification of pathways whose dysregulation can drive tumorigenesis through impaired karyotypic maintenance .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "Taxol ( Paclitaxel ) is an important natural product for the treatment of solid tumors such as ovarian , breast , non-small-cell lung tumors , and some head and neck carcinomas .\n\nDifferent concentrations of taxol trigger distinct effects on cell death forms .\n\nIn present study , cell counting kit ( CCK-8 ) assay , confocal fluorescence microscopy imaging , flow cytometry ( FCM ) and western blotting ( WB ) analysis were used to analyze the characteristics of cell death induced by low ( 35 nM ) and high ( 70 microM ) concentration of taxol respectively in human lung adenocarcinoma ( ASTC-a-1 ) cells .\n\nOur results showed that low concentration of taxol induced cell death dominantly in apoptotic fashion associated with nuclear fragmentation , protein synthesis , phosphatidylserine ( PS ) externalization , G2/M cell cycle arrest , Bax translocation into mitochondria and caspase-3 activation , whereas high concentration of this drug induced significant cytoplasm vacuolization , mitochondria swelling and paraptosis-like cell death form without protein synthesis that is necessary for paraptosis .\n\nAlthough the mechanism of high concentration of taxol-induced paraptosis-like cell death has not been clear , this finding might have a potential implication for cancer therapy , especially for apoptosis-resistant cancer .", "output": "Resisting cell death" }, { "input": "BACKGROUND Current treatments have a modest impact on survival of pancreatic cancer patients and this study investigates the interaction between sorafenib and gemcitabine and the molecular pharmacodynamics of this combination .\n\nMETHODS The pancreatic cancer cells were treated with sorafenib and gemcitabine , alone or in combination .\n\nThe effects of treatments were evaluated on cell proliferation , cell cycle , apoptosis , phosphorylation of Akt , c-Kit , ERK and VEGFR2 , and expression of genes related to drug activity .\n\nRESULTS Gemcitabine and sorafenib synergistically interacted on the inhibition of cell proliferation , and assessment of apoptosis demonstrated that drug associations increased the apoptotic index .\n\nSorafenib reduced c-Kit , ERK and VEGFR2 activation and on the other hand , gemcitabine inhibited Akt phosphorylation .\n\nMoreover , quantitative PCR showed that sorafenib modulated the expression of genes related to gemcitabine activity , while gemcitabine induced the expression of RKIP .\n\nCONCLUSION These data demonstrate that gemcitabine and sorafenib combination displays a synergistic effect in pancreatic cancer cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The main objective of the present investigation was to study the urinary neopterin excretion in the context of the activation of the adaptive cellular immune system at the tumor site .\n\nFor this purpose , we compared pre-treatment urinary neopterin levels measured in 92 ovarian cancer patients , with intratumoral levels of mRNA transcripts from factors either involved in the adaptive antitumor immune defense ( CD3 , IFN-\u03b3 , IRF-1 , IRF-2 , SOCS1 and iNOS ) or immune tolerance ( FoxP3 ) .\n\nThis study did not reveal an association between urinary neopterin and one of these investigated \" on tumor site transcripts \" .\n\nFrom all the factors reflecting the magnitude of the local adaptive antitumor response , intratumoral IRF-1 expression above the edge of the 25th percentile was found to predict most reliably favorable progression-free ( median 34 months vs. 10 months ; p < 0.001 ) and overall ( median 52 months vs. 16 months ; p < 0.001 ) survival .\n\nIn contrast , pre-treatment urinary neopterin excretion above 275 \u03bcmol/mol creatinine , which indicates an unspecific activation of the innate immune system , was associated with a very poor overall survival with a median of only 11 months when compared with a median overall survival of 40 months in patients with lower urinary neopterin excretion ( p = 0.021 ) .\n\nInterestingly , the considerable survival benefit in patients with high IRF-1-expressing cancers was completely abrogated as well for progression-free as for overall survival when urinary neopterin concentrations were found to be concomitantly elevated .\n\nThese findings demonstrate that in ovarian carcinomas the unspecific \" cancer-related inflammation \" contributes to a significant subversion of the adaptive antitumor immune defense mounted at the tumor site .", "output": "Tumor promoting inflammation" }, { "input": "Single-nucleotide polymorphisms in genes involved in DNA-damage-induced responses are reported frequently to be a risk factor in various cancer types .\n\nHere we analysed polymorphisms in 5 genes involved in DNA repair ( XPD Asp312Asn and Lys751Gln , XRCC1 Arg399Gln , APE1 Asp148Glu , NBS1 Glu185Gln , and XPA G-4A ) and in a gene involved in regulation of the cell-cycle ( CCND1 A870G ) .\n\nWe compared their frequencies in groups of colon , head and neck , and breast cancer patients , and 2 healthy control groups : ( 1 ) matched healthy Polish individuals and ( 2 ) a NCBI database control group .\n\nHighly significant differences in the distribution of genotypes of the APE1 , XRCC1 and CCND1 genes were found between colon cancer patients and healthy individuals .\n\nThe 148Asp APE1 allele and the 399Gln XRCC1 allele apparently increased the risk of colon cancer ( OR = 1.9-2.3 and OR = 1.5-2.1 , respectively ) .\n\nAdditionally , frequencies of XPD genotypes differed between healthy controls and patients with colon or head and neck cancer .\n\nImportantly , no differences in the distribution of these polymorphisms were found between healthy controls and breast cancer patients .\n\nThe data clearly indicate that the risk of colon cancer is associated with single-nucleotide polymorphism in genes involved in base-excision repair and DNA-damage-induced responses .", "output": "Genomic instability and mutation" }, { "input": "Evidence suggests that stem-like cells are responsible for initiation , maintenance and recurrence of solid tumors , including Glioblastoma Multiforme ( GBM ) .\n\nGBM is an intractable , highly lethal tumor of the central nervous system .\n\nAlthough epidermal growth factor receptor ( EGFR ) is highly expressed in many GBMs , anti-EGFR therapies have been unsuccessful as treatment .\n\nFew studies have examined EGFR activation in GBM stem cells ( GSCs ) to determine if patient-specific GSCs are amenable to anti-EGFR therapy pre-clinically .\n\nWe hypothesized that EGFR activation in GSCs varied between patients and was an important determinant of responsiveness to anti-EGFR therapy .\n\nCell cycle and apoptosis analysis was performed on tumor-spheres by immuncytochemistry in the presence and absence of the AG1478 .\n\nSecond messenger pathways operative in these processes were elucidated by immunoblotting .\n\nEGFR activated AKT and inactivated GSK3beta in EGFR+/PTEN+ GSCs .\n\nAG1478 and erlotinib significantly decreased the total number of tumor-spheres that EGFR+/ PTEN+ GSCs generated and the rate of sphere formation .\n\nInhibition of EGFR signaling by AG1478 increased GSC senescence and apoptosis , likely via inhibition of AKT and activation of GSK3beta .\n\nSphere formation by EGFR-/ PTEN- GSCs was independent of EGF stimulation , but dependant on B27 growth supplement .\n\nOur data suggest that EGFR+/PTEN+ GSCs are susceptible to anti-EGFR therapy in vitro .", "output": "Sustaining proliferative signaling, Enabling replicative immortality, Resisting cell death" }, { "input": "n-3 Polyunsaturated fatty acids ( PUFA ) have a chemopreventive effect while n-6 PUFA promote carcinogenesis .\n\nThe effect of these essential fatty acids may be related to oxidative stress .\n\nTherefore , the study was designed to evaluate the effect of different ratios of fish oil ( FO ) and corn oil ( CO ) in the prevention of colon cancer .\n\nMale Wistar rats were divided into control , dimethylhydrazine dihydrochloride ( DMH ) treated , FO + CO ( 1:1 ) and FO + CO ( 2.5:1 ) .\n\nAll the groups , except the control received a weekly injection of DMH for 4 weeks .\n\nThe animals were sacrificed either 48 h later ( initiation phase ) or kept for 16 weeks ( post initiation phase ) .\n\nDMH treatment in the initiation phase animals showed mild to moderate inflammation , decreased ROS and TrxR activity , increased antioxidants , apoptosis and ACF multiplicity .\n\nThe post initiation study showed severe inflammation with hyperplasia , increased ACF multiplicity and ROS levels , a decrease in antioxidants and apoptosis .\n\nThe FO + CO ( 1:1 ) treated animals showed severe inflammation , a decrease in ROS , an increase in antioxidants and apoptosis in the initiation phase .\n\nFO + CO ( 1:1 ) in the post initiation phase and FO + CO ( 2.5:1 ) in the initiation showed mild inflammation , increased ROS , apoptosis and decreased antioxidants .\n\nThere was a decrease in ACF multiplicity and ROS levels , increased antioxidants and apoptosis in the post initiation phase study .\n\nThe present study suggests that FO has a dose- and time-dependent chemopreventive effect in colon cancer mediated through oxidative stress and apoptosis .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "BACKGROUND In this report , we explored the role of PKCalpha and PKCe as mediators of phorbol 12-myristate13-acetate ( PMA)-induced proliferation in pituitary tumor GH3B6 cells , and determined if the ERK1/2 and Akt pathways were activated .\n\nMETHODS The GH3B6 cell proliferation was estimated by BrdU incorporation and the cell cycle progression by flow cytometric cell cycle analysis .\n\nWe determined the expression of PKCalpha and PKCe in membrane and cytosolic fractions by western blotting .\n\nThe subcellular redistribution of both PKC isozymes was analyzed by confocal microscopy .\n\nRESULTS Incubation with PMA for 15 min stimulated PKCalpha and PKCe activation , which was correlated with the phosphorylation of ERK1/2 but not Akt .\n\nThe activation of both these PKC isozymes was closely associated with the stimulation of proliferation and the cell cycle progression induced by PMA in GH3B6 cells , an effect that was blocked by the inhibitors of PKCalpha ( G\u00f66976 ) and PKCe ( eV1-2 ) .\n\nIn addition , the pretreatment with the inhibitor of ERK1/2 ( PD98059 ) prevented the mitogenic activity induced by treatment with PMA for 15 min .\n\nCONCLUSION We demonstrated that the activation of PKCalpha and PKCe by phorbol ester in tumor pituitary GH3B6 cells led to cell proliferation and cell cycle progression , effects that involved ERK1/2 activation .", "output": "Sustaining proliferative signaling" }, { "input": "In spite of numerous advances , the 5-year survival rate for head and neck squamous cell cancer has remained largely stagnant and few new anti-tumor drugs have been developed .\n\nPCH4 , a derivative of n-butylidenephthalide , has been investigated for its anti-tumor effects on oral squamous cell carcinoma ( OSCC ) .\n\nThe aim of this study was to investigate the anti-tumor mechanism of a potential target gene , Nur77 , in OSCC cells , which can be induced by PCH4 treatment .\n\nData show that PCH4 promoted Nur77 translocation from the nucleus to the cytoplasm and induced cell apoptosis in OSCC cells .\n\nWhen Nur77 translocation was blocked , the degree of tumor apoptosis caused by PCH4 was significantly inhibited ( p\u2009<\u20090.05 ) .\n\nWithin the MAPK pathway , PCH4 only induced JNK phosphorylation .\n\nFurthermore , treatment with a JNK inhibitor significantly reduced PCH4-induced apoptosis ( p\u2009<\u20090.05 ) and decreased PCH4-induced Nur77 expression ( p\u2009<\u20090.05 ) .\n\nIn a xenograft animal model , administration of PCH4 also showed anti-tumor effects .\n\nWe have demonstrated that OSCC cells are sensitive to PCH4 and that Nur77 protein translocation from the nucleus to the cytoplasm might be associated with the induction of apoptosis by PCH4 .\n\nThese results indicate that PCH4 may serve as a potential anti-tumor drug for OSCC therapy .", "output": "Resisting cell death" }, { "input": "INTRODUCTION Natural herbal compounds with novel actions different from existing breast cancer ( BCa ) treatment modalities are attractive for improving therapeutic efficacy and safety .\n\nWe have recently shown that penta-1,2,3,4,6-O-galloyl-\u03b2-D-glucose ( PGG ) induced S-phase arrest in prostate cancer ( PCa ) cells through inhibiting DNA replicative synthesis and G(1) arrest , in addition to inducing cell death at higher levels of exposure .\n\nWe and others have shown that PGG through intraperitoneal ( i.p. ) injection exerts a strong in vivo growth suppression of human PCa xenograft models in athymic nude mice .\n\nThis study aims to test the hypothesis that the novel targeting actions of PGG are applicable to BCa cells , especially those lacking proven druggable targets .\n\nMETHODS Mono-layer cell culture models of p53-wild type estrogen receptor ( ER)-dependent MCF-7 BCa cells and p53-mutant ER-/progesterone receptor ( PR)- and Her2-regular ( triple-negative ) MDA-MB-231 BCa were exposed to PGG for a comprehensive investigation of cellular consequences and molecular targets/mediators .\n\nTo test the in vivo efficacy , female athymic mice inoculated with MDA-MB-231 xenograft were treated with 20mg PGG/kg body weight by daily gavage starting 4 days after cancer cell inoculation .\n\nRESULTS Exposure to PGG induced S-phase arrest in both cell lines as indicated by the lack of 5-bromo2'-deoxy-uridine ( BrdU ) incorporation into S-phase cells as well as G(1) arrest .\n\nHigher levels of PGG induced more caspase-mediated apoptosis in MCF-7 , in strong association with induction of P53 Ser(15) phosphorylation , than in MDA-MB-231 cells .\n\nThe cell cycle arrests were achieved without an induction of cyclin dependent kinase ( CDK ) inhibitory proteins P21(Cip1) and P27(Kip1) .\n\nPGG treatment led to decreased cyclin D1 in both cell lines and over-expressing cyclin D1 attenuated G(1) arrest and hastened S arrest .\n\nIn serum-starvation synchronized MCF-7 cells , down-regulation of cyclin D1 was associated with de-phosphorylation of retinoblastoma ( Rb ) protein by PGG shortly before G(1)-S transition .\n\nIn vivo , oral administration of PGG led to a greater than 60% inhibition of MDA-MB231 xenograft growth without adverse effect on host body weight .\n\nCONCLUSIONS Our in vitro and in vivo data support PGG as a potential drug candidate for breast cancer with novel targeting actions , especially for a triple negative BCa xenograft model .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "The NF-\u03baB is best known for its role in inflammation .\n\nHere we show that constitutive NF-\u03baB activity in cancer cells promotes the biosynthesis of redox scavenger glutathione ( GSH ) , which in turn confers resistance to oxidative stress .\n\nInhibition of NF-\u03baB significantly decreases GSH in several lines of human leukemia and prostate cancer cells possessing high or moderate NF-\u03baB activities .\n\nConcomitantly , NF-\u03baB inhibition by pharmacological and molecular means sensitizes \" NF-\u03baB positive \" cancer cells to chemically-induced oxidative stress and death .\n\nWe propose that inhibition of NF-\u03baB can reduce intracellular GSH in \" NF-\u03baB-positive \" cancers thereby improving the efficacy of oxidative stress-based anti-cancer therapy .", "output": "Tumor promoting inflammation" }, { "input": "Maintaining the integrity of the cell cycle is critical for ensuring that cells only undergo DNA replication and proliferation under controlled conditions in response to discrete stimuli .\n\nOne mechanism by which the fidelity of this process is guaranteed is through the activation of cell cycle checkpoints .\n\nThe mitotic spindle checkpoint , which is regulated by Aurora B kinase , ensures proper kinetochore attachment to chromosomes leading to equal distribution of chromosomes to daughter cells .\n\nWe demonstrated that the mitogen-activated protein kinase ( MAPK ) cascade regulates mitotic progression and the spindle checkpoint .\n\nAs demonstrated by immunofluorescence at kinetochores , depletion of Raf Kinase Inhibitory Protein ( RKIP ) , an inhibitor of Raf/MEK/ERK signaling , causes an increase in MAPK activity that inhibits Aurora B kinase activity .\n\nBy monitoring mitotic index and transit time from nuclear envelope breakdown to anaphase , we demonstrated that RKIP depletion leads to a defective spindle checkpoint and genomic instability , particularly in response to drugs that disrupt microtubule function .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Verbascum thapsus commonly known as ' mullein ' is part of a large family of Scrophulariaceae consisting of more than 360 species .\n\nFrom antiquity Verbascum thapsus has been used as a medicinal herb , it contains diverse polysaccharides , iroid glycosides , flavonoids , saponins , volatile oils and phenylentanoids .\n\nInducible nitric oxide synthase ( iNOS ) represents one of the three isoforms that produce nitric oxide using L-arginine as a substrate in response to an increase in superoxide anion activated by NF-kB .\n\nIt is implicated in different pathophysiological events and its expression increases greatly during an inflammatory process , due to oxidative stress and the activation of the enzymes of the antioxidant network such as SOD , CAT and GPx.In this study an inflammatory state was reproduced by treating THP-1 cells ( human myelomonocytic leukaemia ) with pro-inflammatory stimuli , such as LPS and IFN-gamma , obtaining an up-regulation both in the expression and in the activity of iNOS .\n\nThe aim of the work was to investigate the antiinflammatory action of verbascoside using a concentration of 100 mum .\n\nThe results show a significant decrease of the expression and activity of iNOS , extracellular O(2) ( - ) production , SOD , CAT and GPx activity when the cells were treated with verbascoside .\n\nBased on these results it is hypothesized that verbascoside has antiinflammatory properties since it reduces the production of superoxide radicals and consequently reduces the activity of iNOS .", "output": "Tumor promoting inflammation" }, { "input": "PURPOSE Idarubicin is a synthetic anthracycline anticancer drug widely used in the treatment of some hematological malignancies .\n\nThe studies in our laboratory have clearly demonstrated that idarubicin can undergo reductive bioactivation by NADPH-cytochrome P450 reductase to free radicals with resulting formation of DNA strand breaks , which can potentially contribute to its genotoxic effects [ Celik , H. , Arin\u00e7 , E. , Bioreduction of idarubicin and formation of ROS responsible for DNA cleavage by NADPH-cytochrome P450 reductase and its potential role in the antitumor effect .\n\nJ Pharm Pharm Sci , 11(4):68-82 , 2008 ] .\n\nIn the current study , our aim was to investigate the possible protective effects of several phenolic antioxidants , quercetin , rutin , naringenin , resveratrol and trolox , against the DNA-damaging effect of idarubicin originating from its P450 reductase-catalyzed bioactivation .\n\nMETHODS DNA damage was measured by detecting single-strand breaks in plasmid pBR322 DNA using a cell-free agarose gel method .\n\nRESULTS Our results indicated that , among the compounds tested , quercetin was the most potent antioxidant in preventing DNA damage .\n\nQuercetin significantly decreased the extent of DNA strand breaks in a dose-dependent manner ; 100 microM of quercetin almost completely inhibited the DNA strand breakage .\n\nUnlike quercetin , its glycosidated conjugate rutin , failed to provide any significant protection against idarubicin-induced DNA strand breaks except at the highest concentration tested ( 2 mM ) .\n\nThe protective effects of other antioxidants were significantly less than that of quercetin even at high concentrations .\n\nQuercetin was found to be also an effective protector against DNA damage induced by mitomycin C. CONCLUSION We conclude that quercetin , one of the most abundant flavonoids in the human diet , is highly effective in reducing the DNA damage caused by the antitumor agents , idarubicin and mitomycin C , following bioactivation by P450 reductase .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Cisplatin chemotherapy often causes acute kidney injury in cancer patients .\n\nThe causative mechanisms of cisplatin-induced acute kidney injury include renal inflammation , activation of p53 tumour suppressor protein and tubular apoptosis .\n\nLuteolin , a flavone found in medicinal herbs and plants , has been reported to exhibit anti-inflammatory , antioxidant and anticarcinogenic activities .\n\nThe purpose of this study was to investigate the anti-apoptotic effect of luteolin on cisplatin-induced acute kidney injury and the molecular mechanism .\n\nMETHODS C57BL/6 mice were treated with cisplatin ( 20 mg/kg ) with or without treatment with luteolin ( 50 mg/kg for 3 days ) .\n\nRenal function , histological changes , degree of oxidative stress and tubular apoptosis were examined .\n\nThe effects of luteolin on cisplatin-induced expression of renal p53 , PUMA-\u03b1 and Bcl-2 family proteins were evaluated .\n\nRESULTS Treatment of mice with cisplatin resulted in renal damage , showing an increase in blood urea nitrogen and creatinine levels , tubular damage , oxidative stress and apoptosis .\n\nTreatment of cisplatin-treated mice with luteolin significantly improved renal dysfunction , reducing tubular cell damage , oxidative stress and apoptosis .\n\nExamination of molecules involving apoptosis of the kidney revealed that treatment of cisplatin increased the levels of p53 and its phosphorylation , PUMA-\u03b1 , Bax and caspase-3 activity that were significantly decreased by treatment with luteolin .\n\nCONCLUSION These results indicate that cisplatin induces acute kidney injury by regulation of p53-dependent renal tubular apoptosis and that luteolin ameliorates the cisplatin-mediated nephrotoxicity through down-regulation of p53-dependent apoptotic pathway in the kidney .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "BACKGROUND Oxidative stress and inflammation are important steps in the pathogenesis of atherosclerosis .\n\nWe postulated that therapeutic concentrations of aspirin and pravastatin , especially in combination , may suppress oxidative stress and inflammation in endothelial cells , and this concept was examined in human coronary artery endothelial cells ( HCAECs ) .\n\nMETHODS Human coronary artery endothelial cells were cultured and treated with oxidized-low density lipoprotein ( ox-LDL , 60 microg/ml for 24 hours ) alone , or pre-treated with aspirin ( 1 , 2 or 5 mmol/L ) , pravastatin ( 1 , 5 or 10 micromol/L ) or their combination ( 1 mmol/L aspirin and 5 micromol/L pravastatin ) , followed by ox-LDL treatment .\n\nAfter respective treatment , superoxide anion production , p38 mitogen activated protein kinase and transcription factor NF-kappaB activation , protein expression of lectin-like ox-LDL receptor-1 ( LOX-1 ) and adhesion molecules , and monocyte adhesion were measured .\n\nRESULTS Ox-LDL treatment greatly elicited its receptor LOX-1 expression , superoxide anion production and inflammatory response , which were minimally affected by low concentration of aspirin ( 1 mmol/L ) or pravastatin ( 5 micromol/L ) , but were markedly decreased by their combination .\n\nActivation of p38 mitogen activated protein kinase and NF-kappaB , the expression of intercellular adhesion molecule-1 and monocyte chemotactic protein-1 , which were only mildly affected by aspirin or pravastatin alone , were significantly attenuated by their combination .\n\nAs a consequence , monocyte adhesion to endothelial cells was markedly attenuated by the combination of the two agents .\n\nWell-known anti-oxidants alpha-tocopherol and gamma-tocopherol had similar inhibitory effects on ox-LDL-mediated oxidative stress and LOX-1 expression as well as monocyte adhesion as did the combination of aspirin and pravastatin .\n\nCONCLUSIONS These studies point to a positive interaction between aspirin and pravastatin with regard to endothelial biology .\n\nAnti-oxidant and subsequent anti-inflammatory effect may be one of the potential underling mechanisms .", "output": "Tumor promoting inflammation" }, { "input": "Application of adenovirus vectors ( Adv ) in metastatic cancer treatment is limited .\n\nWe previously demonstrated that covalent conjugation of polyethleneglycol ( PEG ) to Adv enhances therapeutic effects and decreases toxic side-effects after systemic administration , but the level of immune response to PEGylated Adv ( PEG-Ad ) was not examined .\n\nHere , we examined the effect of PEGylation of Adv on the production of anti-Adv antibodies and antitumor response .\n\nWe constructed a set of PEG-Ad using 5-kDa PEG , with modification rates of 30% , 45% and 90% .\n\nAfter systemic administration of Advs to rats , we examined the level of anti-Adv immunoglobulin ( Ig)G and IgM in serum .\n\nThe levels of anti-Adv IgG and anti-Adv IgM in rats treated with unmodified Adv were higher than those in control group .\n\nRats treated with PEG-Ad that had a 90% modification rate showed lower level of anti-Adv IgG and anti-Adv IgM than those treated with unmodified Adv , whereas rats treated with PEG-Ad that had a 30% or 45% modification rate showed a similar level of anti-Adv IgG and IgM to those treated with unmodified Adv. Systemic administration of PEG-Ad that had a 90% modification rate , and expressed tumor necrosis factor-alpha , significantly reduced the number of metastatic colonies in the lung compared to unmodified Adv , with negligible side effects .\n\nThese results suggest that systemic administration of PEG-Ad with an appropriate PEG modification rate has the potential to reduce the production of antibodies against Adv and increase the therapeutic response against metastatic cancer .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "BACKGROUND Advancing age is associated with substantial increases in the incidence rates of common diseases affecting the prostate gland including benign prostatic hyperplasia ( BPH ) and prostate carcinoma .\n\nThe prostate is comprised of a functional secretory epithelium , a basal epithelium , and a supporting stroma comprised of structural elements , and a spectrum of cell types that includes smooth muscle cells , fibroblasts , and inflammatory cells .\n\nAs reciprocal interactions between epithelium and stromal constituents are essential for normal organogenesis and serve to maintain normal functions , discordance within the stroma could permit or promote disease processes .\n\nIn this study we sought to identify aging-associated alterations in the mouse prostate microenvironment that could influence pathology .\n\nMETHODOLOGY/PRINCIPAL FINDINGS We quantitated transcript levels in microdissected glandular-adjacent stroma from young ( age 4 months ) and old ( age 20-24 months ) C57BL/6 mice , and identified a significant change in the expression of 1259 genes ( p<0.05 ) .\n\nThese included increases in transcripts encoding proteins associated with inflammation ( e.g. , Ccl8 , Ccl12 ) , genotoxic/oxidative stress ( e.g. , Apod , Serpinb5 ) and other paracrine-acting effects ( e.g. , Cyr61 ) .\n\nThe expression of several collagen genes ( e.g. , Col1a1 and Col3a1 ) exhibited age-associated declines .\n\nBy histology , immunofluorescence , and electron microscopy we determined that the collagen matrix is abundant and disorganized , smooth muscle cell orientation is disordered , and inflammatory infiltrates are significantly increased , and are comprised of macrophages , T cells and , to a lesser extent , B cells .\n\nCONCLUSION/SIGNIFICANCE These findings demonstrate that during normal aging the prostate stroma exhibits phenotypic and molecular characteristics plausibly contributing to the striking age associated pathologies affecting the prostate .", "output": "Tumor promoting inflammation" }, { "input": "A 51-year-old previously healthy man , an ex-smoker , was admitted to the authors ' medical department with a 3-month history of dry cough ; intermittent fever ; painless , ulcerated cutaneous lesions over the trunk and limbs ( Figure 1 ) ; and progressive weight loss .\n\nHe was of Greek descent .\n\nHis medical history was remarkable for nasal polyps , which were surgically removed 15 years earlier .\n\nInitially , he had been treated with antibiotics , without improvement .\n\nSeveral days before admission , chest radiography revealed pulmonary infiltrates in the left lower lobe .\n\nOn admission , physical examination revealed a well-orientated man in mild distress , with inspiratory rhonchi at the lower part of the left lung and scattered erythematous nodules of variable size , some of which were ulcerated .\n\nLaboratory values were notable for leukopenia , 3.3 x 10(9)/L ; total protein , 5.9 g/dL ; globulin , 2.2 g/dL ; serum glutamic oxaloacetic transaminase , 86 IU/L ; serum glutamic pyruvic transaminase , 71 IU/L ; and lactate dehydrogenase , 519 U/L .\n\nComputed tomograph ( CT ) of the chest showed multiple alveolar opacities bilaterally ( Figure 2 ) .\n\nFiberoptic bronchoscopy did not reveal any important pathologic findings .\n\nResults of bronchial biopsy , cytology of bronchoalveolar lavage , washing , brushing , and sputum following bronchoscopy were negative .\n\nCT of the brai and sinonasal area revealed an abnormal low-density mass in the left nasal area .\n\nCT findings of the abdomen were negative , as were results of a bone marrow biopsy .\n\nThere was no evidence of immunosuppression .\n\nThe differential diagnosis , considering the evidence described , included granulomatous or infectious diseases , angiocentric lymphoproliferative lesions , and lymphomas .\n\nBiopsy of a skin lesion showed lymphoproliferative infiltration of the dermis with a follicular and angiocentric growth pattern and regional epidermal necrosis .\n\nImmunohistochemical stains showed that the tumor cell were positive for CD56 and CD3 ( cytoplasmic positivity ) and expressed the cytotoxic proteins T-cell intracellular antigen and granzyme B ( Figure 3 ) They lacked TdT , CD34 , CD7 , CD8 , TCL-1 , and CD123 .\n\nFindings from an in situ hybridization study for Epstein-Barr virus were negative .\n\nGive this result , molecular analysis ofT-cell receptor ( TCR ) gene rearrangements was performed using polymerase chain reaction-based TCR-gamma gene , wit negative results .\n\nThe morphology and the immunophenotype were consistent with natural killer/T-cell lymphoma , nasal-type .\n\nNasal involvement must be first excluded to proceed to the diagnosis of nasal-type natural killer-cell lymphoma .\n\nIndeed , histologic examination of the nasal mass revealed its polypoid nature .\n\nThus , the authors were led to the diagnosis of extranodal extranasal natural killer/T-cell lymphoma , nasal-type , CD56-positive , Ep stein-Barr virus-negative , TCR-negative .\n\nThe patient received combination chemotherapy and completed 4 cycles of cyclophosphamide , doxorubicin vincristine , and prednisone every 14 days for 2 months .\n\nSkin lesions improved , and there was no fever soon after the initiation of therapy .\n\nReevaluatio after the fourth cycle , however , disclosed pulmonary infiltrations as well as leukemic infiltration of the central nervous system .\n\nThe patient had receive systemic salvage chemotherapy and intrathecal infusions of methotrexate .\n\nAlthough the lung lesions had diminished at that time , the patient develope paraplegia , his clinical course rapidly deteriorated , and he eventually died .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "Progression of breast cancer is associated with remodeling of the extracellular matrix , often involving a switch from estrogen dependence to a dependence on EGF receptor ( EGFR)/HER-2 and is accompanied by increased expression of the main binding protein for insulin-like growth factors ( IGFBP-3 ) .\n\nWe have examined the effects of IGFBP-3 on EGF responses of breast epithelial cells in the context of changes in the extracellular matrix .\n\nOn plastic and laminin with MCF-10A normal breast epithelial cells , EGF and IGFBP-3 each increased cell growth and together produced a synergistic response , whereas with T47D breast cancer cells IGFBP-3 alone had no effect , but the ability of EGF to increase cell proliferation was markedly inhibited in the presence of IGFBP-3 .\n\nIn contrast on fibronectin with MCF-10A cells , IGFBP-3 alone inhibited cell growth and blocked EGF-induced proliferation .\n\nWith the cancer cells , IGFBP-3 alone had no effect but enhanced the EGF-induced increase in cell growth .\n\nThe insulin-like growth factor-independent effects of IGFBP-3 alone on cell proliferation were completely abrogated in the presence of an EGFR , tyrosine kinase inhibitor , Iressa .\n\nAlthough IGFBP-3 did not affect EGFR phosphorylation [ Tyr(1068) ] , it was found to modulate receptor internalization and was associated with activation of Rho and subsequent changes in MAPK phosphorylation .\n\nThe levels of fibronectin and IGFBP-3 within breast tumors may determine their dependence on EGFR and their response to therapies targeting this receptor .", "output": "Sustaining proliferative signaling" }, { "input": "The AP-1 transcription factor c-Jun is essential for cellular proliferation in many cell types , but the molecular link between growth factors and c-Jun activation has been enigmatic .\n\nIn this study we identify a previously uncharacterized RING-domain-containing protein , RACO-1 ( RING domain AP-1 co-activator-1 ) , as a c-Jun co-activator that is regulated by growth factor signalling .\n\nRACO-1 interacted with c-Jun independently of amino-terminal phosphorylation , and was both necessary and sufficient for c-Jun/AP-1 activation .\n\nGrowth factor-mediated stimulation of AP-1 was attributable to MEK/ERK-dependent stabilization of RACO-1 protein .\n\nStimulation of the MEK/ERK pathway strongly promoted Lys 63-linked ubiquitylation of RACO-1 , which antagonized Lys 48-linked degradative auto-ubiquitylation of the same Lys residues .\n\nRACO-1 depletion reduced cellular proliferation and decreased expression of several growth-associated AP-1 target genes , such as cdc2 , cyclinD1 and hb-egf .\n\nMoreover , transgenic overexpression of RACO-1 augmented intestinal tumour formation triggered by aberrant Wnt signalling and cooperated with oncogenic Ras in colonic hyperproliferation .\n\nThus RACO-1 is a co-activator that links c-Jun to growth factor signalling and is essential for AP-1 function in proliferation .", "output": "Sustaining proliferative signaling" }, { "input": "AIMS Treatment decisions are difficult in clinically localised prostate cancer and further biomarkers of aggressive behaviour are required .\n\nWe investigated the hypothesis that the tissue expression of three cell cycle markers , Rb , p21 and p16 , would provide helpful prognostic information in a well characterised series of prostate cancers which were clinically localised and treated conservatively .\n\nMETHODS The immunohistochemical staining expression of these markers was assessed in tissue microarrays and correlated with 10 year prostate cancer survival and overall survival and then compared with pathological data including contemporary Gleason score , age , measures of tumour extent and initial serum prostate specific antigen ( PSA ) level .\n\nRESULTS Rb overexpression did not show any significant association with Gleason score or prostate cancer survival. p21 protein expression showed a significant association with prostate cancer survival ( p\u2009=\u20090.02 ) and overall survival ( p\u2009=\u20090.01 ) in a univariate model but not in a multivariate model with pathological and serum PSA data .\n\nThere was a significant association between p16 cytoplasmic expression and prostate cancer survival ( HR\u2009=\u20092.52 , 95%CI\u2009= 1.79-3.55 , p\u2009<\u20090.001 ) and overall survival ( HR\u2009= 1.54 , 95% CI\u2009=\u20091.20-1.98 , p\u2009=\u20090.001 ) in a univariate model. p16 expression remained an independent prognostic factor for prostate cancer survival ( HR\u2009=\u20091.50 , 95%CI\u2009=\u20091.05-2.14 , p\u2009=\u20090.03 ) .\n\nCONCLUSION We conclude that p16 cytoplasmic expression can be used as a predictor of outcome in conservatively treated prostate cancer .\n\nRb and p21 show no independent association with outcome and therefore further research is not warranted .", "output": "Evading growth suppressors" }, { "input": "Cytokeratin ( CK ) 19-positive hepatocellular carcinoma ( HCC ) has been reported to have a poor prognosis .\n\nThe mechanism of the development of CK19-positive HCC remains to be studied .\n\nTo clarify this , in vitro experiments were performed using human HCC cell lines ( PLC-5 , HepG2 ) , and the phenotypic changes after stimulation with several growth factors were examined using quantitative reverse transcriptase PCR , western blotting , and immunofluorescence staining .\n\nIn vivo experiments using human HCC specimens obtained from a total of 78 patients and clinicopathological analysis were also performed .\n\nAmong the growth factors tested , epidermal growth factor ( EGF ) had prominent effects on inducing CK19 expression in PLC-5 and HepG2 , which was accompanied by the reduced expression of \u03b1-fetoprotein in PLC-5 .\n\nThe induction of CK19 expression after EGF stimulation was accompanied by the phosphorylation of c-Jun-N-terminal kinase ( JNK)/stress-activated protein kinase , which was blocked by the addition of JNK inhibitors .\n\nEGF also increased proliferative abilities and invasive properties of the HCC cell lines .\n\nIn vivo , 9 ( 12% ) of 78 HCC cases showed positive immunohistochemical staining of CK19 .\n\nThe extent of positive immunohistochemical signals of EGF , EGF receptor ( EGFR ) , and JNK expression was significantly intense in CK-19-positive HCC than those of CK19-negative HCC .\n\nClinicopathological analysis showed that CK19-positive HCC had a high incidence of portal vein invasion , extrahepatic metastasis and an early relapse , which was associated with the worsened 2-year disease free survival .\n\nThese results indicate that the activation of the EGF-EGFR signaling pathway is associated with the development of CK19-positive HCC , and the EGF-induced increase in growth abilities of HCC may account for the poor prognosis of the patients .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "In this study , we compared the effects of SKI-606 with Iressa , Src/Abl and EGF-R kinase inhibitors , respectively , on selected parameters in HeLa and SiHa cervical cancer cell lines , which express E6/E7 oncoproteins of high-risk HPV types 18 and 16 , respectively .\n\nOur results show that SKI-606 and Iressa inhibit cell proliferation and provoke G(0)-G(1) cell cycle arrest and reduction of S and G(2)-M phase using 2 and 5\u2009\u03bcM concentrations of these inhibitors .\n\nIn contrast , SKI-606 induces differentiation to an epithelial phenotype \" mesenchymal-epithelial transition \" ; thus SKI-606 causes a dramatic decrease in cell motility and invasion abilities of HeLa and SiHa cancer cells , in comparison to untreated cells and Iressa-treated cells in which these parameters are only slightly affected .\n\nThese changes are accompanied by a regulation of the expression patterns of E-cadherin and catenins .\n\nThe molecular pathway analysis of Src/Abl inhibitor revealed that SKI-606 blocks the phosphorylation of \u03b2-catenin and consequently converts its role from a transcriptional regulator to a cell-cell adhesion molecule .\n\nOur findings indicate that SKI-606 inhibits signaling pathways involved in regulating tumor cell migration and invasion genes via \u03b2-catenin alteration , suggesting that Src inhibitor , in comparison to EGF-R , is a promising therapeutic agent for human cervical cancer .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "INTRODUCTION I\u03baB Kinase \u03b5 ( IKK\u03b5 ) is a member of the IKK family which plays an important role in the activation of nuclear factor-\u03baB ( NF-\u03baB ) .\n\nOverexpressed in over 30% of breast cancers , IKK\u03b5 has been recently identified as a potential breast cancer oncogene .\n\nThe purpose of this study is to examine the therapeutic potential of IKK\u03b5 siRNA on human breast cancer cells .\n\nMETHODS Eight siRNAs targeting different regions of the IKK\u03b5 mRNA were designed , and the silencing effect was screened by quantitative real time RT-PCR .\n\nThe biological effects of synthetic siRNAs on human breast cancer cells were investigated by examining the cell proliferation , migration , invasion , focus formation , anchorage-independent growth(via soft agar assay ) , cell cycle arrest , apoptosis ( via annexing binding ) , NF-\u03baB basal level , and NF-\u03baB related gene expressions upon the IKK\u03b5 silencing .\n\nRESULTS Silencing of IKK\u03b5 in human breast cancer cells resulted in decrease of focus formation potential and clonogenicity as well as in vitro cell migration/invasion capabilities .\n\nMoreover , knockdown of IKK\u03b5 suppressed cell proliferation .\n\nCell cycle assay showed that the anti-proliferation effect of IKK\u03b5 siRNA was mediated by arresting cells in G(0)/G(1) phase , which was caused by down-regulation of cyclin D(1) .\n\nFurthermore , we demonstrated that silencing of IKK\u03b5 inhibited the NF-\u03baB basal activity as well as the Bcl-2 expression .\n\nSignificant apoptosis was not observed in breast cancer cells upon the silencing of IKK\u03b5 .\n\nThe present study provided the first evidence that silencing IKK\u03b5 using synthetic siRNA could inhibit the invasiveness properties and proliferation of breast cancer cells .\n\nCONCLUSIONS Our results suggested that silencing IKK\u03b5 using synthetic siRNA may offer a novel therapeutic strategy for breast cancer .", "output": "Sustaining proliferative signaling, Resisting cell death, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Unlike the growth factor dependence of normal cells , cancer cells can maintain growth factor-independent glycolysis and survival through expression of oncogenic kinases , such as BCR-Abl .\n\nAlthough targeted kinase inhibition can promote cancer cell death , therapeutic resistance develops frequently , and further mechanistic understanding is needed .\n\nCell metabolism may be central to this cell death pathway , as we have shown that growth factor deprivation leads to decreased glycolysis that promotes apoptosis via p53 activation and induction of the proapoptotic protein Puma .\n\nHere , we extend these findings to show that elevated glucose metabolism , characteristic of cancer cells , can suppress protein kinase C\u03b4 ( PKC\u03b4)-dependent p53 activation to maintain cell survival after growth factor withdrawal .\n\nIn contrast , DNA damage-induced p53 activation was PKC\u03b4 independent and was not metabolically sensitive .\n\nBoth stresses required p53 Ser(18) phosphorylation for maximal activity but led to unique patterns of p53 target gene expression , showing distinct activation and response pathways for p53 that were differentially regulated by metabolism .\n\nConsistent with oncogenic kinases acting to replace growth factors , treatment of BCR-Abl-expressing cells with the kinase inhibitor imatinib led to reduced metabolism and p53- and Puma-dependent cell death .\n\nAccordingly , maintenance of glucose uptake inhibited p53 activation and promoted imatinib resistance .\n\nFurthermore , inhibition of glycolysis enhanced imatinib sensitivity in BCR-Abl-expressing cells with wild-type p53 but had little effect on p53-null cells .\n\nThese data show that distinct pathways regulate p53 after DNA damage and metabolic stress and that inhibiting glucose metabolism may enhance the efficacy of and overcome resistance to targeted molecular cancer therapies .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Cellular energetics" }, { "input": "Aberrant expression and polymorphism of fibroblast growth factor receptor 4 ( FGFR4 ) has been linked to tumor progression and anticancer drug resistance .\n\nWe describe here a novel mechanism of tumor progression by matrix degradation involving epithelial-to-mesenchymal transition in response to membrane-type 1 matrix metalloproteinase ( MT1-MMP , MMP-14 ) induction at the edge of tumors expressing the FGFR4-R388 risk variant .\n\nBoth FGFR4 and MT1-MMP were upregulated in tissue biopsies from several human cancer types including breast adenocarcinomas , where they were partially coexpressed at the tumor/stroma border and tumor invasion front .\n\nThe strongest overall coexpression was found in prostate carcinoma .\n\nStudies with cultured prostate carcinoma cell lines showed that the FGFR4-R388 variant , which has previously been associated with poor cancer prognosis , increased MT1-MMP-dependent collagen invasion .\n\nIn this experimental model , knockdown of FGFR4-R388 or MT1-MMP by RNA interference blocked tumor cell invasion and growth in collagen .\n\nThis was coupled with impaired phosphorylation of FGFR substrate 2 and Src , upregulation of E-cadherin , and suppression of cadherin-11 and N-cadherin .\n\nThese in vitro results were substantiated by reduced MT1-MMP content and in vivo growth of prostate carcinoma cells after the FGFR4-R388 gene silencing .\n\nIn contrast , knockdown of the alternative FGFR4-G388 allele enhanced MT1-MMP and invasive tumor cell growth in vivo and within three-dimensional collagen .\n\nThese results will help to explain the reported association of the FGFR4-R388 variant with the progression and poor prognosis of certain types of tumors .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Current palliative chemotherapy ( CT ) regimens achieve clinical benefits in less than 50% of patients treated for metastatic gastric cancers , and long-term survivals are anecdotical .\n\nGenetic polymorphisms and differences at the level of transcription in genes involved in biological processes of drug metabolism , DNA repair and drug resistance can explain the observed individual differences in response to drugs , in survival and in different susceptibility to the toxic effects of CT .\n\nThe possibility to classify patients on the basis of genetic signatures could help in choosing the CT regimen .\n\nWe present herein an analysis of genetic and expression profiling of three patients affected by metastatic gastric cancer , treated with CT and alive , disease-free , at 66-82 months .\n\nFour patients with typical clinical outcome represented the control group .\n\nExpression profiling from paraffin-embedded tumor tissues was performed on an ad hoc set of genes involved in drug metabolism and resistance , DNA repair , cell cycle regulation and growth factors signalling .\n\nGenetic polymorphism analysis on DNA extracted from peripheral blood was done by pyrosequencing of genetic markers predictive of drug response .\n\nExpression analysis in long-term survivors revealed a significant upregulation of PTEN , TP63 , GADD45a and MAPK1 genes .\n\nWe found also an upregulation of CYP1A1 , CYP3A4 and ERBB4 genes .\n\nEGF was found to be down-regulated in long-term survivors .\n\nERCC1 C8092A polymorphism seems to be associated with survival in our set of patients .\n\nThe present study shed light on a set of genes , which could have a predictive role in survival of patients with metastatic gastric tumors .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Cancer cells undergo multi-step processes in obtaining the ability to metastasize , and are constantly exposed to signals that induce apoptosis .\n\nAcquisition of anti-apoptotic properties by cancer cells is important for metastasis , and recent studies suggest that transforming growth factor ( TGF)-\u03b2 promotes the survival of certain types of cancer cells .\n\nHere , we found that in highly metastatic breast cancer cells , JygMC(A) , JygMC(B) and 4T1 , TGF-\u03b2 ligands were produced in autocrine fashion .\n\nPharmacological inhibition of endogenous TGF-\u03b2 signalling by a TGF-\u03b2 type I receptor kinase inhibitor in serum-free conditions increased the expression of BH3-only protein , Bim ( also known as Bcl2-like 11 ) in JygMC(A) and JygMC(B) cells , and caused apoptotic cell death .\n\nWe also found that induction of Bim by TGF-\u03b2 was not observed in Foxc1 knocked-down cancer cells .\n\nThese findings suggest that TGF-\u03b2 plays a crucial role in the regulation of survival of certain types of cancer cells through the TGF-\u03b2-Foxc1-Bim pathway , and that specific inhibitors of TGF-\u03b2 signalling might be useful as apoptosis inducers in breast cancer cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Hypoxia\u2013reoxygenation ( HR ) is a primary driver of angiogenesis in both atherogenesis and tumorigenesis .\n\nThe main target of hypoxia-driven proangiogenic signaling is adherens junctions responsible for contact inhibition of endothelial cells .\n\nWe analyzed the effects of hypoxia ( 8\u201312 hours ) followed by a brief period of reoxygenation ( 2 hours ) ( HR ) on angiogenesis and integrity of adherens junction in cultured human umbilical vein endothelial cells as well as the effects of aspirin on modulation of human umbilical vein endothelial cells ' response to HR .\n\nCells exposed to HR displayed considerable enhancement of tube formation ( angiogenesis ) on matrigel .\n\nImmunocytostaining of near-confluent cells revealed that HR caused disruption of adherens junctions and internalization of their components VE-cadherin , p120 catenin , and b-catenin .\n\nAdditionally , HR resulted in the appearance of binucleated cells , and VE-cadherin in colocalization with b-catenin was found to be positioned between the separating nuclei .\n\nPresence of aspirin ( acetylsalicylic acid , 1 mM ) resulted in preservation of adherens junctions on the cellular membrane and prevented angiogenesis as well as mitosis .\n\nHR caused upregulation LOX-1 , the p47(phox) subunit of NADPH , while reducing transcription of endothelial nitric oxide synthase .\n\nAspirin had no effect on endothelial nitric oxide synthase and canceled the transcriptional activation of the LOX-1 and p47(phox) subunit of NADPH oxidase .\n\nBased on these data , we hypothesize that aspirin preserves the integrity of adherens junctions and thus blunts angiogenic response to HR through downregulation of LOX-1 and the LOX-1-mediated p47(phox) component of NADPH oxidase transcription , thus preventing NADPH oxidase assembly and function .", "output": "Inducing angiogenesis" }, { "input": "Myxoid leiomyosarcoma is an extremely rare variant of leiomyosarcoma , masquerading almost to perfection as a benign lesion .\n\nFor , indeed , the tumor lacks the defining features of high mitotic activity , cellular atypia or necrosis , and the microscopic picture is dominated by abundant myxoid stroma containing sparse spindle cells .\n\nWe report here such a case occurring in the uterus and discuss the differential diagnosis .\n\nThe relevant literature is reviewed .", "output": "Resisting cell death" }, { "input": "Estrogen receptor \u03b1 ( ER\u03b1 ) expression in breast cancer is predictive of response to endocrine therapy ; however , resistance is common in ER\u03b1-positive tumors that overexpress the growth factor receptor ERBB2 .\n\nEven in the absence of estrogen , ER\u03b1 can be activated by growth factors , including the epidermal growth factor ( EGF ) .\n\nEGF induces a transcriptional program distinct from estrogen ; however , the mechanism of the stimulus-specific response is unknown .\n\nHere we show that the EGF-induced ER\u03b1 genomic targets , its cistromes , are distinct from those induced by estrogen in a process dependent on the transcription factor AP-1 .\n\nThe EGF-induced ER\u03b1 cistrome specifically regulates genes found overexpressed in ERBB2-positive human breast cancers .\n\nThis provides a potential molecular explanation for the endocrine therapy resistance seen in ER\u03b1-positive breast cancers that overexpress ERBB2 .\n\nThese results suggest a central role for ER\u03b1 in hormone-refractory breast tumors dependent on growth factor pathway activation and favors the development of therapeutic strategies completely antagonizing ER\u03b1 , as opposed to blocking its estrogen responsiveness alone .", "output": "Sustaining proliferative signaling" }, { "input": "Designed from a high throughput screened hit compound , novel 2-amino-1-thiazolyl imidazoles were synthesized and demonstrated cytotoxicity against human cancer cells. 1-(4-Phenylthiazol-2-yl)-4-(thiophen-2-yl)-1H-imidazol-2-amine ( compound 2 ) , a 2-amino-1-thiazolyl imidazole , inhibited tubulin polymerization , interacted with the colchicine-binding sites of tubulins , and caused cell cycle arrest at the G(2)/M phase in human gastric cancer cells .\n\nDisruption of the microtubule structure in cancer cells by compound 2 was also observed .\n\nCompound 2 concentration-dependently inhibited the proliferation of cancer cells in histocultured human gastric and colorectal tumors .\n\nGiven orally , compound 2 prolonged the lifespans of leukemia mice intraperitoneally inoculated with the murine P388 leukemic cells .\n\nWe report 2-amino-1-thiazolyl imidazoles as a novel class of orally active microtubule-destabilizing anticancer agents .", "output": "Evading growth suppressors" }, { "input": "OBJECTIVE The goal of this study was to investigate the relationship between plasma levels of insulin-like growth factors-1 ( IGF-1 ) and IGF-binding protein-3 ( IGFBP-3 ) and the risk for cervical intraepithelial neoplasia ( CIN ) and cervical cancer .\n\nMETHODS Plasma levels of IGF-1 and IGFBP-3 of 44 cervical cancer patients , 82 CIN patients and 40 neoplasm-free patients were investigated .\n\nThen the associations of the plasma levels of IGF-1 and IGFBP-3 with cervical neoplasm or its clinicopathologic parameters were analyzed .\n\nRESULTS The mean IGF-1 concentrations were significantly different among the control , CIN , and cervical cancer groups ; the levels were higher in the CIN group compared to the controls .\n\nAccording to the quartile category , the plasma IGF-1 level was significantly higher ( p=0.0015 ) in the CIN group than in the controls .\n\nThe IGFBP-3 level showed no association between the controls and CIN groups ( p=0.842 ) .\n\nAlthough the mean IGF-1/IGFBP-3 molar ratio had borderline significance ( p=0.08 ) among the study population , the quartile comparison showed a significantly higher IGF-1/IGFBP-3 molar ratio in the CIN group compared to the control group ( p=0.041 ) .\n\nCONCLUSION Plasma levels of IGF-1 and the IGF-1/IGFBP-3 molar ratio might be useful for the development early detection of cervical lesions and used as an adjuvant diagnostic tool for cervical neoplasia after more larger scale research .", "output": "Sustaining proliferative signaling" }, { "input": "The aim of this study was to determine whether isorhamnetin , an immediate 3'-O-methylated metabolite of quercetin , affects proliferation , cell death , and the cell cycle of human colon carcinoma ( HCT-116 ) cells .\n\nIsorhamnetin was found to be a potent antiproliferative agent in a dose- and time-dependent manner , with an IC50 of 72 \u03bcM after 48 h of incubation as estimated by MTT assay .\n\nFlow cytometry and fluorescence microscopy analysis showed that isorhamnetin exerted a stimulatory effect on apoptosis and necrosis .\n\nIsorhamnetin also increased the number of cells in G2/M phase .\n\nSerum deprivation appeared to potentiate the effects of isorhamnetin on cell death and facilitated cell cycle progression to G0/G1 phase .\n\nThese results suggest that isorhamnetin might mediate inhibition of HCT-116 cell growth through the perturbation of cell cycle progression and are consistent with the notion that G2/M checkpoints could be a conserved target for flavonoids in human colon cancer cells , leading to apoptotic and necrotic death .\n\nThese antiproliferative , apoptotic , necrotic , and cell cycle effects suggest that isorhamnetin may have clinically significant therapeutic and chemopreventive capabilities .\n\nTo our knowledge , this is the first report of the effect of isorhamnetin on human colon cancer cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Cetuximab is a chimeric antibody registered for the therapy of advanced colorectal carcinoma .\n\nCancer and anticancer therapy are associated with oxidative stress , and disorders of antioxidant balance may be involved in the toxicity associated with anticancer treatment .\n\nThe aim of the present study was to investigate the changes of serum retinol , alpha-tocopherol and C-reactive protein during the first month of treatment with cetuximab and chemotherapy .\n\nTwenty-five consecutive patients with metastatic colorectal carcinoma treated with a combination of chemotherapy and cetuximab were included in the present study .\n\nSerum retinol and alpha-tocopherol were determined by high-performance liquid chromatography and serum C-reactive protein was determined using commercial kits .\n\nSignificant correlation was observed between baseline concentrations of retinol and C-reactive protein ( r(s)=-0.54 , p<0.01 ) .\n\nMedian survival of patients who had baseline serum retinol below 1.25 \u00b5mol/L was 10 mo compared to 18 mo for patients who had serum retinol equal or above 1.25 \u00b5mol/L ( p<0.05 ) ; median survival of patients who had serum C-reactive protein below 24 mg/L was significantly longer compared to patients with C-reactive protein levels equal or above 24 mg/L ( 18 vs. 7 mo , p<0.05 ) , but no difference in survival was observed based on alpha-tocopherol levels .\n\nTwenty-two patients had evaluation of retinol , alpha-tocopherol and C-reactive protein at least once during the follow up .\n\nSerum concentration of alpha-tocopherol decreased significantly during the therapy , but retinol and C-reactive protein concentrations remained unchanged .\n\nIn conclusion , a significant correlation was observed between serum retinol and C-reactive protein .\n\nSerum alpha-tocopherol decreased significantly during the first month of combination therapy with cetuximab .\n\nLow retinol and high C-reactive protein concentrations were predictive of poor prognosis in this patient population .", "output": "Tumor promoting inflammation" }, { "input": "Spindle cell oncocytoma ( SCO ) of the pituitary gland is a relatively recently established , very rare subtype of adenohypophysis tumours that was introduced as a distinct clinicopathological entity in the fourth edition of WHO classification of the central nervous system tumours ( 2007 ) .\n\nIt is non-endocrine neoplasm of the anterior pituitary that occurs in adults and usually follows a benign clinical course , corresponding to WHO grade I. Up to now , pituitary SCO have been reported occasionally and only 14 cases of SCO have been documented in the literature .\n\nBecause of their rarity , the pathogenesis and natural history of these tumours have not been fully characterized .\n\nWe report two additional cases of SCO occurring in females aged 63 years ( Case 1 ) and 65 years ( Case 2 ) , who presented with pan-hypopituitarism , headache and visual field defect .\n\nIn both cases , the magnetic resonance imaging showed solid sellar mass of moderate size with suprasellar extension .\n\nThe clinical and radiological features suggested non-functioning pituitary macroadenomas without evidence of invasive growth .\n\nOne patient presented with tumour recurrence 3 years after undergoing the previous surgical removal of tumour , which was initially misdiagnosed as schwannoma .\n\nThe first tumour was removed by transsphenoidal surgery and the second one by frontal craniotomy .\n\nHistologically and immunohistochemically , both tumours displayed the features typical for SCO of the pituitary .\n\nThey were composed of interwoven fascicles of spindle cells exhibiting abundant eosinophilic cytoplasm of oncocytic or granular appearance .\n\nMitoses were rarely observed and necrosis was absent .\n\nIn one case , the advanced lymphocytic infliltration was observed within neoplastic tissue .\n\nThe tumour cells exhibited immunoreactivity for S-100 protein , galectin-3 , vimentin and epithelial membrane antigen but they were negative for GFAP , anterior pituitary neuroendocrine markers ( prolactin , growth hormone , TSH , ACTH , FSH , LH ) , chromogranin , synaptophysin , cytokeratin CK ( AE1/AE3 ) , smooth muscle actin , desmin , CD34 and CD68 .\n\nMIB1 labeling index did not exceed 10% .\n\nUltrastructurally , the tumour cells were rich in mitochondria with lamellar cristae .\n\nMoreover , in Case 2 some tumour cells showed a number of giant mitochondria with severely destructed internal matrix .\n\nSpindle cell oncocytoma of the anterior pituitary is often misdiagnosed entity of uncertain histogenesis .\n\nIt should be considered in the differential diagnosis of various sellar-region lesions of oncocytic morphology .", "output": "Resisting cell death" }, { "input": "BACKGROUND Apoptosis-stimulating protein of p53 ( ASPP ) family members can stimulate the apoptotic function of p53 but have no impact on its cell cycle arrest function .\n\nMATERIAL AND METHODS The expression pattern of the ASPP family consisting of ASPP1 , ASPP2 , and iASPP was examined by immunohistochemistry in 45 formalin-fixed and paraffin-embedded endometrial endometrioid adenocarcinoma ( EEA ) specimens and 26 normal endometrial tissue ( NET ) samples .\n\nRESULTS The expression rates of ASPP1 and ASPP2 in EEA were significantly lower than those in NET ( p < 0.05 ) .\n\nHowever , the iASPP expression rate in EEA was statistically higher in contrast to NET ( p < 0.05 ) .\n\nExpression of ASPP1 and iASPP in EEA had no correlation with any clinicopathological features ( p > 0.05). iASPP was associated with grade , invasion , and lymph node metastasis ( p < 0.05 ) .\n\nCONCLUSIONS It is a novel finding that the expression pattern of the ASPP family members has respective pathological and clinical implications in EEA , and iASPP might be a candidate target for EEA therapy .", "output": "Activating invasion and metastasis" }, { "input": "Reciprocity of inflammation , oxidative stress and neovascularization is emerging as an important mechanism underlying numerous processes from tissue healing and remodelling to cancer progression .\n\nWhereas the mechanism of hypoxia-driven angiogenesis is well understood , the link between inflammation-induced oxidation and de novo blood vessel growth remains obscure .\n\nHere we show that the end products of lipid oxidation , \u03c9-(2-carboxyethyl)pyrrole ( CEP ) and other related pyrroles , are generated during inflammation and wound healing and accumulate at high levels in ageing tissues in mice and in highly vascularized tumours in both murine and human melanoma .\n\nThe molecular patterns of carboxyalkylpyrroles are recognized by Toll-like receptor 2 ( TLR2 ) , but not TLR4 or scavenger receptors on endothelial cells , leading to an angiogenic response that is independent of vascular endothelial growth factor .\n\nCEP promoted angiogenesis in hindlimb ischaemia and wound healing models through MyD88-dependent TLR2 signalling .\n\nNeutralization of endogenous carboxyalkylpyrroles impaired wound healing and tissue revascularization and diminished tumour angiogenesis .\n\nBoth TLR2 and MyD88 are required for CEP-induced stimulation of Rac1 and endothelial migration .\n\nTaken together , these findings establish a new function of TLR2 as a sensor of oxidation-associated molecular patterns , providing a key link connecting inflammation , oxidative stress , innate immunity and angiogenesis .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "BACKGROUND Human Barrett's cancer cell lines have numerous , poorly-characterized genetic abnormalities and , consequently , those lines have limited utility as models for studying the early molecular events in carcinogenesis .\n\nCell lines with well-defined genetic lesions that recapitulate various stages of neoplastic progression in Barrett's esophagus would be most useful for such studies .\n\nMETHODOLOGY/PRINCIPAL FINDINGS To develop such model cell lines , we started with telomerase-immortalized , non-neoplastic Barrett's epithelial ( BAR-T ) cells , which are spontaneously deficient in p16 , and proceeded to knock down p53 using RNAi , to activate Ras by introducing oncogenic H-Ras(G12V) , or both .\n\nBAR-T cells infected with either p53 RNAi or oncogenic H-Ras(G12V) alone maintained cell-to-cell contact inhibition and did not exhibit anchorage-independent growth in soft agar .\n\nIn contrast , the combination of p53 RNAi knockdown with expression of oncogenic H-Ras(G12V) transformed the p16-deficient BAR-T cells , as evidenced by their loss of contact inhibition , by their formation of colonies in soft agar , and by their generation of tumors in immunodeficient mice .\n\nCONCLUSIONS/SIGNIFICANCE Through these experiments , we have generated a number of transformed and non-transformed cell lines with well-characterized genetic abnormalities recapitulating various stages of carcinogenesis in Barrett's esophagus .\n\nThese lines should be useful models for the study of carcinogenesis in Barrett's esophagus , and for testing the efficacy of chemopreventive and chemotherapeutic agents .", "output": "Evading growth suppressors" }, { "input": "Autophagy is one of the survival processes of cancer cells , especially in stressful conditions such as starvation , hypoxia and chemotherapeutic agents .\n\nHowever , its roles in tumor survival have not yet been fully elucidated .\n\nHere , we found for the first time that JAK2/STAT3 was activated in HeLa cells when they were starved or treated with rapamycin .\n\nSTAT3 activation was associated with autophagic processes , because it was completely inhibited by 3-methyladenine , partially inhibited by knockdown of molecules associated with autophagic processes and blocked by antioxidants , DPI , a Nox inhibitor and knockdown of p22 phox , indicating that ROS generated by Nox that was activated during autophagic processes activated JAK2/STAT3 pathway .\n\nActivated STAT3 directly bound to IL6 promoter and increased IL6 mRNA and protein secretion .\n\nFinally , the conditioned media , which included IL6 , from starved HeLa cells promoted cancer cell survival in both normal and starved conditions , confirmed by clonogenic , proliferation and cell death assays .\n\nThese data together indicate that the autophagic process in cancer cells can contribute to their survival by JAk2/STAT3 activation and subsequent secretion of growth factors .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Diallyl disulfide ( DADS ) is a major organo-sulfur compound derived from garlic ( Allium sativum ) , which inhibits the proliferation of various types of cancer cells .\n\nIn this study we investigated the effect of DADS on the induction of apoptosis , as well as its regulatory effect on the activation of transcription factors in B16F-10 melanoma cells .\n\nTreatment of B16F-10 cells with nontoxic concentrations of DADS resulted in the presence of apoptotic bodies and induced DNA fragmentation in a dose-dependent manner .\n\nCell-cycle analysis revealed that the occurrence of the sub-G1 peak was significantly elevated in DADS-treated cells .\n\nDADS treatment also down-reguated Bcl-2 expression and up-regulated p53 , caspase-9 , and caspase-3 expression in B16F-10 melanoma cells .\n\nThe study also reveals that DADS inhibited the activation and nuclear translocation of p65 , p50 , and c-Rel subunits of nuclear factor ( NF)-B and other transcription factors , such as c-fos , activated transcription factor-2 , and cyclic adenosine monophosphate response element-binding protein , in B16F-10 melanoma cells The pro-inflammatory cytokine production and gene expression of tumor necrosis factor ( TNF)-\u03b1 , interleukin ( IL)-1\u03b2 , IL-6 , and granulocyte-macrophage colony-stimulating factor ( GM-CSF ) were down-regulated in DADS-treated cells compared with control B16F-10 metastatic melanoma cells .\n\nDADS induces caspase-dependent apoptosis through a mitochondria-mediated intrinsic pathway in B16F-10 melanoma cells by activating p53 and caspase-3 gene expression and suppressing pro-inflammatory cytokines and NF-B-mediated Bcl-2 activation .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Ultraviolet ( UV ) light induces DNA-damage checkpoints and mutagenesis , which are involved in cancer protection and tumorigenesis , respectively .\n\nHow cells identify DNA lesions and convert them to checkpoint-activating structures is a major question .\n\nWe show that during repair of UV lesions in noncycling cells , Exo1-mediated processing of nucleotide excision repair ( NER ) intermediates competes with repair DNA synthesis .\n\nImpediments of the refilling reaction allow Exo1 to generate extended ssDNA gaps , detectable by electron microscopy , which drive Mec1 kinase activation and will be refilled by long-patch repair synthesis , as shown by DNA combing .\n\nWe provide evidence that this mechanism maybe stimulated by closely opposing UV lesions , represents a strategy to redirect problematic repair intermediates to alternative repair pathways , and may also be extended to physically different DNA damages .\n\nOur work has significant implications for understanding the coordination between repair of DNA lesions and checkpoint pathways to preserve genome stability .", "output": "Genomic instability and mutation" }, { "input": "Base analogs are powerful antimetabolites and dangerous mutagens generated endogenously by oxidative stress , inflammation , and aberrant nucleotide biosynthesis .\n\nHuman inosine triphosphate pyrophosphatase ( ITPA ) hydrolyzes triphosphates of noncanonical purine bases ( i.e. , ITP , dITP , XTP , dXTP , or their mimic : 6-hydroxyaminopurine ( HAP ) deoxynucleoside triphosphate ) and thus regulates nucleotide pools and protects cells from DNA damage .\n\nWe demonstrate that the model purine base analog HAP induces DNA breaks in human cells and leads to elevation of levels of ITPA .\n\nA human polymorphic allele of the ITPA , 94C->A encodes for the enzyme with a P32T amino-acid change and leads to accumulation of nonhydrolyzed ITP .\n\nThe polymorphism has been associated with adverse reaction to purine base-analog drugs .\n\nThe level of both spontaneous and HAP-induced DNA breaks is elevated in the cell line with the ITPA P32T variant .\n\nThe results suggested that human ITPA plays a pivotal role in the protection of DNA from noncanonical purine base analogs .", "output": "Genomic instability and mutation" }, { "input": "Overexpression of the melanoma differentiation associated gene-7 ( MDA-7)/IL-24 in vitro generally results in the growth suppression and induction of apoptosis of diverse human tumor cells .\n\nIn this study , we investigated the effects of overexpression of the MDA-7/IL-24 gene in human hepatocellular carcinoma ( HCC ) cells in vitro and in vivo .\n\nAdenovirus-mediated overexpression of MDA-7 facilitated the MDA-7/IL-24-induced apoptosis and G2/M arrest in HCC cells , but not in the normal liver cell line L02 , and the effect was independent of the p53 status .\n\nInhibition of metastasis and angiogenesis was correlated with decreasing expression of STAT3 , P-STAT3 , MMP-2 , VEGF , and TGF-beta genes , regulated by STAT3 in MHCCLM6 cells .\n\nWe also showed that Ad.mda-7 combined with doxorubicin ( ADM ) had significantly enhanced antitumor and antimetastatic effects in vivo , accompanied by the downregulation of VEGF , MMP-2 , and TGF-beta genes and the upregulation of E-cadherin genes .\n\nThese data suggested that MDA-7/IL-24 induces its selective antitumor properties in HCC cells by promoting apoptosis independent of p53 status , inhibiting subcutaneous tumor growth and metastasis , and increasing the effect of chemotherapeutic agents .\n\nMDA-7/IL-24 represents a new class of cancer suppressor genes that may be useful in the targeted therapy of HCC .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Evading growth suppressors, Inducing angiogenesis, Resisting cell death" }, { "input": "BACKGROUND Breast cancer is less common in China than in the United States and perinatal characteristics predict breast cancer risk in the offspring .\n\nWe determined levels of pregnancy hormones in Boston and Shanghai to identify those possibly involved in the intrauterine origin of breast cancer .\n\nParticipants and methods : We compared maternal and cord blood levels of estradiol , estriol , testosterone , progesterone , prolactin , insulin-like growth factors ( IGF ) 1 and 2 , insulin-like growth factor-binding protein 3 , adiponectin and sex hormone-binding globulin ( SHBG ) in 241 Caucasian and 295 Chinese women .\n\nRESULTS In both centers , hormone levels at the 16th were predictive of those at the 27th gestational week , but there was little correlation between maternal and cord blood levels .\n\nIn cord blood , we found significantly ( P < 0.01 ) higher levels of estradiol ( 44.2% ) , testosterone ( 54.5% ) , IGF-2 ( 22.7% ) and strikingly SHBG ( 104.6% ) in Shanghai women , whereas the opposite was true for IGF-1 ( -36.8% ) .\n\nCONCLUSIONS Taking into account the current understanding of the plausible biological role of the examined endocrine factors , those likely to be involved in the intrauterine origin of breast cancer are SHBG and IGF-2 , with higher cord blood levels among Chinese , and IGF-1 , with higher cord blood levels among Caucasian women .", "output": "Sustaining proliferative signaling" }, { "input": "A growing number of studies have demonstrated an association between serum levels of insulin-like growth factors ( IGFs ) and IGF binding protein-3 ( IGFBP-3 ) and increased risk for various cancers .\n\nThe aim of this study was to evaluate the relationship between levels of IGF-II or IGFBP-3 in cervical scrapes with cervical cancer and precancerous lesions : low-grade squamous intraepithelial lesion ( LSIL ) and high-grade squamous intraepithelial lesion ( HSIL). 4 groups of cases were examined : LSIL ( n=20 ) , HSIL ( n=28 ) , cervical cancer ( n=45 ) , and controls ( n=51 ) .\n\nControl subjects were women with normal , HPV DNA-negative Papanicolau ( Pap ) test .\n\nIGF-II and IGFBP-3 levels in cervical scrapes were measured by ELISA .\n\nResults show that median protein levels of IGF-II were significantly lower in cervical cancer cases vs. controls ( 446.5 ng/mg vs. 1,168.6 ng/mg , p<0.001 ) .\n\nSignificantly higher values of IGFBP-3 were found in HSIL vs. controls ( median : 549.5 ng/mg vs. 216 ng/mg ; p=0.018 ) , and were not affected by HR HPV infection , meanwhile no significant differences were observed in IGFBP-3 levels between LSIL or cervical cancer as compared to controls .\n\nThese data suggests that the progression to cervical cancer is associated with alterations in the IGF system and not affected by HR HPV infection .\n\nMore studies are needed to understand the possible role of IGFBP-3 in cervical carcinogenesis .", "output": "Sustaining proliferative signaling" }, { "input": "Mode of action ( MOA ) analysis provides a systematic description of key events leading to adverse health effects in animal bioassays for the purpose of informing human health risk assessment .\n\nUncertainties and data gaps identified in the MOA analysis may also be used to guide future research to improve understanding of the MOAs underlying a specific toxic response and foster development of toxicokinetic and toxicodynamic models .\n\nAn MOA analysis , consistent with approaches outlined in the MOA Framework as described in the Guidelines for Carcinogen Risk Assessment , was conducted to evaluate small intestinal tumors observed in mice chronically exposed to relatively high concentrations of hexavalent chromium ( Cr(VI) ) in drinking water .\n\nBased on review of the literature , key events in the MOA are hypothesized to include saturation of the reductive capacity of the upper gastrointestinal tract , absorption of Cr(VI) into the intestinal epithelium , oxidative stress and inflammation , cell proliferation , direct and/or indirect DNA modification , and mutagenesis .\n\nAlthough available data generally support the plausibility of these key events , several unresolved questions and data gaps were identified , highlighting the need for obtaining critical toxicokinetic and toxicodynamic data in the target tissue and in the low-dose range .\n\nExperimental assays that can address these data gaps are discussed along with strategies for comparisons between responsive and nonresponsive tissues and species .\n\nThis analysis provides a practical application of MOA Framework guidance and is instructive for the design of studies to improve upon the information available for quantitative risk assessment .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "BACKGROUND Ginkgolic acids ( GAs ) , extracted from the seed coat of Ginkgo biloba L. Our previous study has shown that GA monomer could inhibit the growth of Hep-2 significantly and induce the fragmentation of the chromosomal DNA .\n\nTo further assess the antitumor potential and turn it into a candidate new antitumor drug , the antitumor mechanism of GA was investigated .\n\nMETHOD The cytotoxicity and antitumor effect of GA monomer were assayed by MTT colorimetric assay with nontumorogenic MC-3T3-E1 as well as tumorogenic Hep-2 and Tac8113 cell lines .\n\nThe effect of GA monomer on the proliferation of tumor cell lines was analyzed with MTT colorimetric and CFSE labeled assay .\n\nCell cycle distribution and measurement of the percentage of apoptotic cells were performed by flow cytometry following stained with propidium iodide , annexin V-FITC .\n\nThe expression of apoptotic proteins Bcl-2 , Bax and caspase-3 was analyzed with Western blot .\n\nRESULT GA only inhibited the growth of tumorogenic cell lines in a both dose- and time-dependent manner .\n\nTumor cells were treated with GA for 72 h , 70.53 \u00b1 4.54% Hep-2 and 63.5 \u00b1 7.2% Tca8113 cells were retarded at GO/G1 phase , and the percentage of apoptosis was 40.4 \u00b1 1.58 and 38.4 \u00b1 1.7% , respectively .\n\nGA-treated activated caspase-3 downregulated the expression of anti-apoptotic Bcl-2 protein and upregulated the expression of pro-apoptotic Bax protein , eventually leading to a decrease in the Bcl-2/Bax ratio in tumor cells .\n\nCONCLUSIONS The antitumor action of GA was due to inhibiting the proliferation in a manner of inhibiting division , retarding the progress of cell cycle and inducing apoptosis , making GA a candidate as new antitumor drug .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "Histone deacetylase inhibitors induce cell cycle arrest and apoptosis in tumor cells and are , therefore , promising anti-cancer drugs .\n\nThe cyclin-dependent kinase inhibitor p21 is activated in histone deacetylase ( HDAC ) inhibitor-treated tumor cells , and its growth-inhibitory function contributes to the anti-tumorigenic effect of HDAC inhibitors .\n\nWe show here that induction of p21 by trichostatin A involves MAP kinase signaling .\n\nActivation of the MAP kinase signaling pathway by growth factors or stress signals results in histone H3 serine 10 phosphorylation at the p21 promoter and is crucial for acetylation of the neighboring lysine 14 and recruitment of activated RNA polymerase II in response to trichostatin A treatment .\n\nIn non-induced cells , the protein phosphatase PP2A is associated with the p21 gene and counteracts its activation .\n\nInduction of p21 is linked to simultaneous acetylation and phosphorylation of histone H3 .\n\nThe dual modification mark H3S10phK14ac at the activated p21 promoter is recognized by the phospho-binding protein 14-3-3\u03b6 , which protects the phosphoacetylation mark from being processed by PP2A .\n\nTaken together we have revealed a cross-talk of reversible phosphorylation and acetylation signals that controls the activation of p21 by HDAC inhibitors and identify the phosphatase PP2A as chromatin-associated transcriptional repressor in mammalian cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The multi-kinase inhibitor dasatinib induced a variable but significant decrease of viability in both p53(wild-type) ( EHEB , JVM-2 , JVM-3 ) and p53(mutated) ( MEC-1 , MEC-2 , BJAB ) prolymphocytic B leukemic cells , due to a combination of cell cycle block in G1 and apoptosis .\n\nAntibody phospho-kinase array analysis revealed that dasatinib inhibited the phosphorylation of various kinases , including ERK1/2 and p38/MAPK as well as of STAT3 transcription factors , in both p53(wild-type) and p53(mutated) cells .\n\nTherefore , dasatinib might offer a novel therapeutic strategy not only for p53(wild-type) , but also for p53(mutated) B malignancies that have the worst prognosis and urgently need innovative therapeutic approaches .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Ceramide induces cell cycle arrest and apoptotic cell death associated with increased levels of p27(kip1) .\n\nThe aim of this study was to examine the effects of ceramide on p27(kip1) protein levels as a measure of cell cycle arrest and apoptosis .\n\nResults showed that ceramide increased p27(kip1) protein levels through activation of protein phosphatase 2A ( PP2A ) in PC-3 prostate cancer cells .\n\nTreatment of cells with the PP2A inhibitor okadaic acid or with PP2A-C\u03b1 siRNA inhibited ceramide-induced enhanced p27(kip1) protein expression and Akt dephosphorylation , and prevented Skp2 downregulation .\n\nOverexpression of constitutively active Akt attenuated ceramide-induced Skp2 downregulation and p27(kip1) upregulation .\n\nIn addition , ceramide stimulated binding of the PP2A catalytic subunit PP2A-C\u03b1\u03b2 to Akt as assessed by immunoprecipitation experiments , indicating that PP2A is involved in the induction of p27(kip1) via inhibition of Akt pathway .\n\nFinally , whether PP2A can regulate p27(kip1) expression independently of Akt pathway was determined .\n\nKnockdown of PP2A-C\u03b1 with siRNA reduced p27(kip1) levels in the presence of Akt inhibitor .\n\nThese data reveal that PP2A is a regulator of ceramide-induced p27(kip1) expression via Akt-dependent and Akt-independent pathways .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "SET and MYND domain-containing protein 3 ( SMYD3 ) is a histone methyltransferase that plays an important role in transcriptional regulation in human carcinogenesis .\n\nIt can specifically methylate histone H3 at lysine 4 and activate the transcription of a set of downstream genes , including several oncogenes ( e.g. , N-myc , CrkL , Wnt10b , RIZ and hTERT ) and genes involved in the control of cell cycle ( e.g. , CyclinG1 and CDK2 ) and signal transduction ( e.g. , STAT1 , MAP3K11 and PIK3CB ) .\n\nTo determine the effects of SMYD3 over-expression on cell proliferation , we transfected SMYD3 into MDA-MB-231 cells and found that these cells showed several transformed phenotypes as demonstrated by colony growth in soft agar .\n\nBesides , we show here that down-regulation of SMYD3 could induce G1-phase cell cycle arrest , indicating the potent induction of apoptosis by SMYD3 knockdown .\n\nThese results suggest the regulatory mechanisms of SMYD3 on the acceleration of cell cycle and facilitate the development of strategies that may inhibit the progression of cell cycle in breast cancer cells .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "Signal transducers and activators of transcription 3 ( Stat3 ) is activated by cytokines and growth factors in lung cancers and regulates expression of genes implicated in cell growth , survival , and transformation .\n\nPreviously , we found that mice with a deletion of the G protein-coupled receptor , family C , group 5 , member a ( Gprc5a ) gene develop lung tumors , indicating that Gprc5a is a tumor suppressor .\n\nHerein , we show that epithelial cells from Gprc5a knockout mouse lung ( Gprc5a(-/-) cells ) survive better in vitro in medium deprived of exogenous growth factors and form more colonies in semisolid medium than their counterparts from wild-type mice ( Gprc5a(+/+) cells ) .\n\nStat3 tyrosine 705 phosphorylation and expression of several Stat3-regulated antiapoptotic genes were higher in Gprc5a(-/-) than in Gprc5a(+/+) cells .\n\nBoth cell types secreted leukemia inhibitory factor ( Lif ) ; however , whereas Stat3 activation was persistent in Gprc5a(-/-) cells , it was transient in Gprc5a(+/+) cells .\n\nLung adenocarcinoma cells isolated from Gprc5a(-/-) mice also exhibited autocrine Lif-mediated Stat3 activation .\n\nThe level of Socs3 , the endogenous Stat3 inhibitory protein , was higher in Gprc5a(+/+) than in Gprc5a(-/-) cells , and expression of the tumor suppressor stabilized Socs3 .\n\nInhibition of Stat3 signaling in Gprc5a(-/-) normal and cancer cells by the Janus-activated kinase 2 inhibitor AG490 or by a dominant negative Stat3(Y705F) increased starvation-induced apoptosis and inhibited colony formation .\n\nThese results show that persistent Stat3 activation is important for the survival and transformation of Gprc5a(-/-) lung cells and suggest that the tumor suppressive effects of Gprc5a are mediated , at least in part , by inhibition of Stat3 signaling through Socs3 stabilization .", "output": "Sustaining proliferative signaling" }, { "input": "INTRODUCTION During selective segregation of DNA , a cell asymmetrically divides and retains its template DNA .\n\nAsymmetric division yields daughter cells whose genome reflects that of the parents ' , simultaneously protecting the parental cell from genetic errors that may occur during DNA replication .\n\nWe hypothesized that long-lived epithelial cells are present in immortal , premalignant cell populations , undergo asymmetric division , retain their template DNA strands , and cycle both during allometric growth and during pregnancy .\n\nMETHODS The glands of 3-week old immune competent Balb/C female mice were utilized intact or cleared of host epithelium and implanted with ductal-limited , lobule-limited , or alveolar-ductal progenitor cells derived from COMMA-D1 pre-malignant epithelial cells. 5-bromo-2-deoxyuridine ( 5-BrdU ) was administered to identify those cells which retain their template DNA .\n\nNulliparous mice were then either injected with [ (3)H]-thymidine ( (3)H-TdR ) to distinguish 5-BrdU-label retaining cells that enter the cell cycle and euthanized , or mated , injected with ( 3)H-TdR , and euthanized at various days post-coitus .\n\nSections were stained for estrogen receptor-\u03b1(ER-\u03b1) or progesterone receptor ( PR ) via immunohistochemistry .\n\nCells labelled with both 5-BrdU and ( 3)H-TdR were indicative of label-retaining epithelial cells ( LREC ) .\n\nRESULTS Cells that retained a 5-BrdU label and cells labelled with [ (3)H]-thymidine were found in all mice and were typically detected along the branching epithelium of mature mouse mammary glands .\n\nCells containing double-labelled nuclei ( LREC ) were found in the intact mammary gland of both pregnant and nulliparous mice , and in mammary glands implanted with pre-malignant cells .\n\nDouble-labelled cells ( (3)H-TdR/5-BrdU ) represent a small portion of cells in the mammary gland that cycle and retain their template DNA ( 5-BrdU ) .\n\nSome label-retaining cells were also ER-\u03b1 or PR positive .\n\nLRECs distributed their second label ( (3)H-TdR ) to daughter cells ; and this effect persisted during pregnancy .\n\nLRECs , and small focal hyperplasia , were found in all immortalized premalignant mammary implant groups .\n\nCONCLUSIONS The results indicate that a subpopulation of long-lived , label-retaining epithelial cells ( LRECs ) is present in immortal premalignant cell populations .\n\nThese LRECs persist during pregnancy , retain their original DNA , and a small percentage express ER-\u03b1 and PR .\n\nWe speculate that LRECs in premalignant hyperplasia represent the long-lived ( memory ) cells that maintain these populations indefinitely .", "output": "Sustaining proliferative signaling" }, { "input": "INTRODUCTION The insulin-like growth factor I receptor ( IGF-IR ) pathway plays a major role in cancer growth , tumor cell survival and resistance to therapy .\n\nBACKGROUND Preclinical evidence that targeting the IGF-IR is effective in cancer treatment has been accumulating for almost 2 decades .\n\nEarly clinical trials revealed an acceptable safety profile together with pharmacodynamic evidence that the receptor can be targeted successfully .\n\nIt is premature to draw conclusions regarding the therapeutic potential of this class of compounds but well-documented single-agent activity was noted during phase I evaluations , and recent evidence from a phase-II study suggests that co-administration of an anti-IGF-1R antibody with chemotherapy for non-small-cell lung cancer ( NSCLC ) improves objective response rate and progression-free survival .\n\nVIEWPOINTS These early results are a strong indication for continued research on the targeting of IGF-R , particularly in the treatment of NSCLC .\n\nCONCLUSIONS Today , IGF-1R targeting appears a promising approach , more than two dozen compounds have been developed and clinical trials are underway .", "output": "Sustaining proliferative signaling" }, { "input": "Accumulating evidence from epidemiological studies indicates that chronic inflammation and oxidative stress play critical roles in neoplastic development .\n\nThe aim of this study was to investigate the anti-inflammatory , anti-oxidative stress activities , and differential regulation of Nrf2-mediated genes by tea Chrysanthemum zawadskii ( CZ ) and licorice Glycyrrhiza uralensis ( LE ) extracts .\n\nThe anti-inflammatory and anti-oxidative stress activities of hexane/ethanol extracts of CZ and LE were investigated using in vitro and in vivo approaches , including quantitative real-time PCR ( qPCR ) and microarray .\n\nAdditionally , the role of the transcriptional factor Nrf2 ( nuclear erythroid-related factor 2 ) signaling pathways was examined .\n\nOur results show that CZ and LE extracts exhibited potent anti-inflammatory activities by suppressing the mRNA and protein expression levels of pro-inflammatory biomarkers IL-1\u03b2 , IL-6 , COX-2 and iNOS in LPS-stimulated murine RAW 264.7 macrophage cells .\n\nCZ and LE also significantly suppressed the NO production of LPS-stimulated RAW 264.7 cells .\n\nAdditionally , CZ and LE suppressed the NF-\u03baB luciferase activity in human HT-29 colon cancer cells .\n\nBoth extracts also showed strong Nrf2-mediated antioxidant/Phase II detoxifying enzymes induction .\n\nCZ and LE induced NQO1 , Nrf2 , and UGT and antioxidant response element ( ARE)-luciferase activity in human hepatoma HepG2 C8 cells .\n\nUsing Nrf2 knockout [ Nrf2 ( -/-) ] and Nrf2 wild-type ( +/+ ) mice , LE and CZ showed Nrf2-dependent transactivation of Nrf2-mediated antioxidant and phase II detoxifying genes .\n\nIn summary , CZ and LE possess strong inhibitory effects against NF-\u03baB-mediated inflammatory as well as strong activation of the Nrf2-ARE-anti-oxidative stress signaling pathways , which would contribute to their overall health promoting pharmacological effects against diseases including cancer .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND/AIMS We investigated whether the anticancer drug Ukrain ( UK ) is able to modulate the expression of some of the key markers of tumor progression in pancreatic cell carcinoma , in order to assess its potential therapeutic effect .\n\nMETHODS Three cell lines ( HPAF-II , PL45 , HPAC ) were treated with UK ( 5 , 10 and 20 \u03bcM ) for 48 h , or left untreated .\n\nSecreted protein acidic and rich in cysteine ( SPARC ) mRNA levels were assessed by real-time PCR .\n\nMatrix metalloproteinases ( MMP)-2 and -9 activity was analyzed by SDS zymography ; SPARC protein levels in cell lysates and supernatants were determined by Western blot .\n\nCell cycle was determined by flow cytometric analysis , and invasion by matrigel invasion assay .\n\nRESULTS UK down-regulated MMP-2 and MMP-9 , suggesting that UK may decrease pancreatic cancer cell invasion , as confirmed by the matrigel invasion assay .\n\nSPARC protein down-regulation in supernatants points to an inhibition by UK of extracellular matrix remodeling in the tumor microenvironment .\n\nAt the same time , SPARC mRNA and cellular protein level up-regulation suggests that UK can affect cell proliferation by cell cycle inhibition , showing a cell cycle G2/M arrest in UK-treated cells .\n\nCONCLUSION Our results suggest that UK modulates two major aspects involved in tumorigenesis of pancreatic cancer cells , such as extracellular matrix remodeling and cell proliferation .", "output": "Evading growth suppressors, Activating invasion and metastasis" }, { "input": "PTEN , a phosphoinositide-3-phosphatase , serves dual roles as a tumor suppressor and regulator of cellular anabolic/catabolic metabolism .\n\nAdaptation of a redox-sensitive cysteinyl thiol in PTEN for signal transduction by hydrogen peroxide may have superimposed a vulnerability to other mediators of oxidative stress and inflammation , especially reactive carbonyl species , which are commonly occurring by-products of arachidonic acid peroxidation .\n\nUsing MCF7 and HEK-293 cells , we report that several reactive aldehydes and ketones , e.g. electrophilic \u03b1,\u03b2-enals ( acrolein , 4-hydroxy-2-nonenal ) and \u03b1,\u03b2-enones ( prostaglandin A(2) , \u039412-prostaglandin J(2) and 15-deoxy-\u0394-12,14-prostaglandin J(2) ) covalently modify and inactivate cellular PTEN , with ensuing activation of PKB/Akt kinase ; phosphorylation of Akt substrates ; increased cell proliferation ; and increased nuclear \u03b2-catenin signaling .\n\nAlkylation of PTEN by \u03b1,\u03b2-enals/enones and interference with its restraint of cellular PKB/Akt signaling may accentuate hyperplastic and neoplastic disorders associated with chronic inflammation , oxidative stress , or aging .", "output": "Tumor promoting inflammation" }, { "input": "INTRODUCTION The progressive growth of malignancies is accompanied by a decline in the immune response through mechanisms which are poorly understood .\n\nApoptosis and induction of inflammation by tumor released cytokines as tumor escape mechanisms have been proposed to play an important role in colorectal carcinogenesis .\n\nMETHODS Expression of Tumor necrosis factor-alpha ( TNF-\u03b1 ) was analyzed in colorectal cancer specimen and the cancer cell line HT-29 by immunohistochemistry and RT-PCR .\n\nTNF-\u03b1 expression on protein and mRNA level were correlated with clinical characteristics and impact on survival .\n\nTNFR-1 was co-labelled with TNF-\u03b1 and CD8+ cytotoxic T cells in immunofluorescence double staining experiments .\n\nRESULTS 94% ( n=98/104 ) of the patients with CRC expressed TNF-\u03b1 .\n\nHigh TNF-\u03b1 expression was significantly associated with positive lymph node stage and recurrence of the tumor .\n\nMultivariate analysis revealed high TNF-\u03b1 expression as an independent prognostic factor .\n\nImmunohistochemistry was correlated with RT-PCR results ( \u03c4=0.794 ) .\n\nImmunofluorescence double staining experiments revealed increased TNFR-1 expression by CD8+ cells .\n\nCONCLUSIONS TNF-\u03b1 expression by tumor cells may be an efficient immunological escape mechanism by inflammation-enhanced metastases and probably by induction of apoptosis in tumor-infiltrating CD8+ immune cells resulting in a down regulation of the tumoral immune response .\n\nOur data support the role of tumor-derived TNF-\u03b1 expression as an important promoter of tumoral immune escape mechanisms and malignant progression , and suggest that analysis on either protein ( immunohistochemistry ) or RNA level ( RT-PCR ) can be used effectively in this respect .\n\nTargeting TNF-\u03b1 may be a promising option , especially in cases with high TNF-\u03b1 expression and positive lymph node metastases .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "p38 kinases activated by growth factors , hormones , and environmental stresses exert diverse functions in regulating normal and malignant cell pathophysiology .\n\nEnhanced levels of activated p38 isoforms have been linked with poor prognosis in breast cancer , although the mechanistic basis for this association is poorly understood .\n\nIn this study , we report that p38 activation in cervical cancer cells is driven by osteopontin ( OPN ) , an extracellular matrix-associated cytokine that drives invasive progression .\n\nOPN regulates CD44-mediated p38 phosphorylation that induces NF-\u03baB activation and NF-\u03baB-dependent expression of furin , an extracellular protease implicated in human papilloma virus ( HPV ) processing that enhances cervical cancer cell motility .\n\nOPN induces CD44-mediated MKK3/6 phosphorylation which in turn phosphorylates p38 in these cells .\n\nOPN-induced furin expression and cell motility was impeded by blockades to MKK3/6 , p38\u03b1/\u03b2 or NF-\u03baB signaling .\n\nIn a mouse xenograft model of human cervical cancer , tumor growth was enhanced by OPN overexpression and blocked by short hairpin RNA ( shRNA)-mediated OPN silencing .\n\nFurin overexpression similarly augmented tumor growth in the model , whereas blocking MKK3/6 , p38 , or furin reduced OPN-induced cervical tumor growth .\n\nAnalysis of clinical specimens revealed that enhanced expression of OPN , phosphorylated NF-\u03baB , p65 , and furin correlated with cervical cancer progression , further strengthening the in vitro and in vivo results .\n\nIn summary , our findings offer a proof of concept for targeting OPN and its downstream p38 signaling as a novel therapeutic strategy to manage cervical cancer .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Though an increased efficacy of carmustine and temozolomide ( TMZ ) has been demonstrated by inactivation of O(6)-methylguanine-DNA methyltransferase ( MGMT ) with O(6)-benzyl-guanine ( BG ) in human pancreatic tumors refractive to alkylating agents , the regulatory mechanisms have not been explored .\n\nMETHODS The effects of TMZ and BG on apoptosis , cell growth , the mitotic index , cell cycle distribution , and protein expression were studied by TUNEL , cell counting , flow cytometry , and Western blot analysis , respectively .\n\nRESULTS The wt-p53 human pancreatic tumor cell line Capan-2 and p53-efficient mouse embryonic fibroblasts ( MEFs ) were more responsive to treatment with TMZ + BG than mutant p53 Capan-1 and p53-null MEFs .\n\nS phase delay with a subsequent G2/M arrest was observed in Capans in response to BG + TMZ .\n\nThe G1-to-S transition delay in Capan-2 was associated with p53-dependent apoptosis and was distinctly different from the presumed mismatch repair ( MMR ) killing operative during the G2/M arrest .\n\nThe effect of p53 on BG + TMZ toxicity was supported by a marked change in apoptosis when p53 function was restored/inactivated .\n\nThere was an early induction of MMR proteins in p53-efficient lines .\n\nCONCLUSION p53 provokes a classic proapoptotic response by delaying G1-to-S progression , but it may also facilitate cell killing by enhancing MMR-related cell cycle arrest and cell death .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Levels of regulatory T cells ( Tregs ) are increased in different cancer types as well as in inflammatory diseases , such as rheumatoid arthritis .\n\nTreg accumulation may result from aberrant proliferation and trafficking as well as greater resilience to oxidative stress compared with conventional T cells .\n\nThis enhanced antioxidative capacity of Tregs possibly serves as feedback inhibition during inflammation and prevents uncontrolled immune reactions by favoring survival of suppressor rather than effector cells .\n\nIn this study , we demonstrate that human Tregs express and secrete higher levels of thioredoxin-1 , a major antioxidative molecule .\n\nThioredoxin-1 has an essential role in maintaining their surface thiol density as the first line of antioxidative defense mechanisms and is sensitive to proinflammatory stimuli , mainly tumor necrosis factor-\u03b1 , in a nuclear factor-\u03baB-dependent fashion .\n\nThe antiapoptotic and oncogenic potential of ( secreted ) Trx-1 suggests that it may exert effects in Tregs beyond redox regulation .", "output": "Tumor promoting inflammation" }, { "input": "The purpose of this study was to evaluate the usefulness of high-resolution MRI ( HR-MRI ) and proton MR spectroscopy ( (1)H-MRS ) for monitoring the early therapeutic response to radiotherapy .\n\nTwenty rabbits with VX2 carcinoma were divided into control ( n = 8 ) and irradiation ( n = 12 ) groups .\n\nThe irradiation group underwent HR-MRI and ( 1)H-MRS using a microscopy coil at 1 , 3 , 7 or 14 days after irradiation .\n\nRabbits in the control group were subjected to HR-MRI and ( 1)H-MRS at the same time intervals .\n\nAll rabbits were killed after imaging and subjected to histopathologic examinations .\n\nThe diameter of necrosis by HR-MRI was then compared to that on the gross specimens .\n\nThe ratios of choline/creatine ( Cho/Cr ) and lactate/creatine ( Lac/Cr ) on the tumor and necrotic area detected by in vivo ( 1)H-MRS were compared between the control and irradiation groups , respectively .\n\nIn addition , the ratios of Cho/Cr and Lac/Cr were compared between the tumor and necrotic area in each irradiation group .\n\nA significant correlation was found between the diameter of necrosis in each sequence of HR-MRI and that in the gross specimens ( r = 0.84-0.91 , p = 0.03- < 0.003 ) .\n\nThe ratios of Lac/Cr in the tumors of the irradiation groups were significantly higher than those in the control groups after 1 day and 3 days of irradiation ( p = 0.04 , and p = 0.02 ) .\n\nHistological analysis showed necrosis and swelling of the endothelia of capillaries and arterioles at 1 day and 3 days after irradiation .\n\nIt was suggested that HR-MRI and ( 1)H-MRS are useful methods for monitoring the early therapeutic response to radiotherapy .", "output": "Resisting cell death" }, { "input": "OBJECTIVE Chronic generation of inflammatory cytokines and reactive oxygen species are implicated in atherosclerosis , aging , cancers , and other chronic diseases .\n\nWe hypothesized that zinc induces A20 in premonocytic , endothelial , and cancer cells , and A20 binds to tumor necrosis factor ( TNF)-receptor associated factor , and inhibits I\u03ba kinase-\u03b1 ( IKK-\u03b1)/nuclear factor-\u03baB ( NF-\u03baB ) , resulting in downregulation of TNF-\u03b1 and interleukin-1\u03b2 ( IL-1\u03b2 ) .\n\nMETHODS To test this hypothesis , we used HL-60 , human umbilical vein endothelial cells , and SW480 cell lines under zinc-deficient and zinc-sufficient conditions in this study .\n\nWe measured oxidative stress markers , inflammatory cytokines , A20 protein and mRNA , A20-FRAF-1 complex , and IKK-\u03b1/NF-\u03baB signaling in stimulated zinc-deficient and zinc sufficient cells .\n\nWe also conducted antisense A20 and siRNA studies to investigate the regulatory role of zinc in TNF-\u03b1 and IL-1\u03b2 via A20 .\n\nRESULTS We found that zinc increased A20 and A20-tumor necrosis factor-receptor associated factor-1 complex , decreased the IKK-\u03b1/NF-\u03baB signaling pathway , oxidative stress markers , and inflammatory cytokines in these cells compared with zinc-deficient cells .\n\nWe confirmed that zinc-induced A20 contributes to downregulation of TNF-\u03b1 and IL-1\u03b2 by antisense and short interfering RNA A20 studies .\n\nCONCLUSION Our studies suggest that zinc suppresses generation of NF-\u03baB-regulated inflammatory cytokines by induction of A20 .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND Apoptosis , a widely important mechanism that contributes to cell growth reduction , is reported to be induced by Crocus sativus in different cancer types .\n\nThe present study was designed to elucidate apoptosis induction by crocin , a main component of Crocus sativus in a human pancreatic cancer cell line ( BxPC-3 ) .\n\nMETHODS Cell viability was measured by MTT assay , Hoechest33258 staining was used to detect the chromatin condensation characteristic of apoptosis , and DNA fragmentation was assessed by gel electrophoresis and cell cycle analysis by flow cytometry .\n\nRESULTS Crocin induced apoptosis and G1-phase cell cycle arrest of BxPC-3 cells , while decreasing cell viability in a dose dependent and time dependent manner .\n\nCells treated with 10\u03bcg/L crocin exhibited apoptotic morphology ( brightly blue-fluorescent condensed nuclei on Hoechst 33258 staining ) and reduction of volume .\n\nDNA analysis revealed typical ladders as early as 12 hours after treatment indicative of apoptosis .\n\nCONCLUSION Our preclinical study demonstrated a pancreatic cancer cell line to be highly sensitive to crocin-mediated growth inhibition and apoptotic cell death .\n\nAlthough the molecular mechanisms of crocin action are not yet clearly understood , it appears to have potential as a therapeutic agent .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "Several studies have indicated that the cell-surface expressed nucleolin is implicated in tumorigenesis and angiogenesis , and represents an important target for cancer therapy .\n\nHere we show that treatment of rhabdoid tumor derived G401 cells with a nucleolin antagonist , the HB-19 pseudopeptide , could restore contact inhibition , impair anchorage-independent growth , and suppress tumor development in nude mice .\n\nG401 cells grow without contact inhibition , which is an in vitro characteristic property of malignant tumor cells .\n\nAt concentrations of HB-19 that does not affect cell viability and multiplication index , there is restoration of contact inhibition thus suggesting that HB-19 treatment causes reversion of the malignant phenotype .\n\nAccordingly , HB-19 pretreated G401 cells lose the capacity to form colonies in soft agar .\n\nWhen assayed for tumorigenicity in nude mice , only 50% of mice injected with HB-19 pretreated G401 cells developed tumors with the mean tumor weight of 0.32 g , compared to 100% of mice injected with control G401 cells with the mean tumor weight of 2.36 g .\n\nInterestingly , the restoration of contact inhibition in HB-19 treated G401 cells is concomitant with marked reduction of transcripts coding the Wilms ' tumor 1 gene , matrix metalloproteinase-2 , epithelial isoform of CD44 , and vascular endothelial growth factor , whereas no apparent modification is detected for transcripts coding the proto-oncogene c-Myc , anti-apoptotic Bcl-2 , pro-apoptotic Bax , tissue inhibitor of metalloproteinase TIMP-1 , angiogenesis inhibitor TSP-1 , and growth factor Midkine .\n\nThese findings indicate that the molecular mechanism of action of HB-19 on such highly malignant rhabdoid tumor cells is associated with a selective inhibitory effect on the expression of genes implicated in tumorigenesis and angiogenesis .", "output": "Evading growth suppressors" }, { "input": "Sarcomatoid renal cell carcinomas of the kidney are rare neoplasms constituting about 1-5% of all renal malignant neoplasms .\n\nThese are aggressive tumors and are commonly associated with conventional ( clear cell ) renal cell carcinomas , but cases associated with chromophobe renal cell carcinomas are sparse .\n\nCytological features of such lesions have rarely been reported .\n\nHere , we report a unique case of a 48-year-old male patient who presented with right flank lump and pain .\n\nA fine needle aspiration was performed from the lesion under ultrasound guidance and a cytological diagnosis of pleomorphic sarcoma was made .\n\nA right-sided radical nephrectomy was carried out and subsequent histopathology revealed a sarcomatoid renal cell carcinoma with wide areas of necrosis coexisting with chromophobe renal cell carcinoma with calcification .\n\nDifferentiation of pleomorphic sarcoma from a sarcomatoid renal cell carcinoma is , thus , challenging from cytopathology smears and the differential diagnoses should always be borne in mind while giving a cytopathological opinion .", "output": "Resisting cell death" }, { "input": "BACKGROUND AND AIMS breast reconstruction with silicone prosthesis following nipple-sparing mastectomy has become widely accepted as a reconstruction option in women requiring mastectomy for cancer .\n\nThe purpose of this study was to evaluate the incidence and some factors influencing early local complications in patients undergoing NSM with immediate implant reconstruction .\n\nMATERIAL AND METHODS prospective study was performed on a consecutive series of 214 breast reconstructions in 205 patients .\n\nAll complications during the six weeks after surgery were recorded. 42 prostheses were implanted after neoadjuvant chemotherapy , 27 patients previously had radiotherapy due to breast conserving surgery and in all other cases surgery was the pri-mary treatment for cancer .\n\nRESULTS the overall six-week complication rate was 16% ( 35 ) and included : major skin flap necrosis ( 4% , 9 procedures ) , minor skin necrosis ( 3% , 7 ) , major infection ( 2% , 5 ) , minor infection ( 3% , 7 ) , prolonged seroma formation ( 3% , 6 ) , haematoma ( 1% , 2 ) and epidermolysis ( 1% , 2 ) .\n\nIn 6% ( 12 ) reconstruction procedures explantation of prosthesis was done .\n\nNeoadjuvant chemo-therapy and radiotherapy were not associated with higher rate of complications .\n\nCONCLUSION nipple-sparing mastectomy with immediate implant reconstruction has acceptable morbidity rate in the hand of experienced oncoplastic surgeon and therefore should be considered as treatment option to women requiring mastectomy .", "output": "Resisting cell death" }, { "input": "Cancer development is associated with genetic instability .\n\nIdentification of specific loci altered during carcinogenesis in a particular tissue gives scope for early detection and predicting the progressive nature of the tissue pathology .\n\nInstability at microsatellite loci is widely attributed to mismatch repair errors due to epigenetic alterations .\n\nUsing three dinucleotide markers , D3S1313 , D9S171 , D17S250 and two mononucleotide markers BAT25 , BATRII , we evaluated MSI in 97 cases enrolled for endoscopy of upper GI tract with symptoms of dyspepsia , reflux or dysphagia .\n\nWe aimed at evaluating markers that reflect instability in esophageal malignancies , examine the prevalence of MSI in cancers and other pathologies of the esophagus , and determine the methylation status of hMLH1 gene in relation to MSI. 42% ( 21/50 ) cancers and 15.4%(2/13) precancers exhibited MSI where 85.7% cancers and 50% precancers with MSI , showed a hypermethylated hMLH1 promoter .\n\nIncreased number of cases with repair gene methylation were seen with increasing severity of the esophageal pathology suggesting epigenetic progression parallels histologic changes .\n\nBAT25 and D3S1313 markers exhibited instability frequently and cases with MSI using these markers showed an abnormal hMLH1 promoter .\n\nThus these markers were useful in identifying the mismatch repair phenotype .\n\nThese two markers may be useful to screen cases for early cancer related changes , after validation on a larger sample .", "output": "Genomic instability and mutation" }, { "input": "Many cells in mammals exist in the state of quiescence , which is characterized by reversible exit from the cell cycle .\n\nQuiescent cells are widely reported to exhibit reduced size , nucleotide synthesis , and metabolic activity .\n\nMuch lower glycolytic rates have been reported in quiescent compared with proliferating lymphocytes .\n\nIn contrast , we show here that primary human fibroblasts continue to exhibit high metabolic rates when induced into quiescence via contact inhibition .\n\nBy monitoring isotope labeling through metabolic pathways and quantitatively identifying fluxes from the data , we show that contact-inhibited fibroblasts utilize glucose in all branches of central carbon metabolism at rates similar to those of proliferating cells , with greater overflow flux from the pentose phosphate pathway back to glycolysis .\n\nInhibition of the pentose phosphate pathway resulted in apoptosis preferentially in quiescent fibroblasts .\n\nBy feeding the cells labeled glutamine , we also detected a \" backwards \" flux in the tricarboxylic acid cycle from \u03b1-ketoglutarate to citrate that was enhanced in contact-inhibited fibroblasts ; this flux likely contributes to shuttling of NADPH from the mitochondrion to cytosol for redox defense or fatty acid synthesis .\n\nThe high metabolic activity of the fibroblasts was directed in part toward breakdown and resynthesis of protein and lipid , and in part toward excretion of extracellular matrix proteins .\n\nThus , reduced metabolic activity is not a hallmark of the quiescent state .\n\nQuiescent fibroblasts , relieved of the biosynthetic requirements associated with generating progeny , direct their metabolic activity to preservation of self integrity and alternative functions beneficial to the organism as a whole .", "output": "Cellular energetics, Evading growth suppressors" }, { "input": "The vascular endothelial growth factor ( VEGF ) receptor tyrosine kinase inhibitor sunitinib has been approved for first-line treatment of patients with metastatic renal cancer and is currently being trialled in other cancers .\n\nHowever , the effectiveness of this anti-angiogenic agent is limited by the presence of innate and acquired drug resistance .\n\nBy screening a panel of candidate growth factors we identified fibroblast growth factor 2 ( FGF2 ) as a potent regulator of endothelial cell sensitivity to sunitinib .\n\nWe show that FGF2 supports endothelial proliferation and de novo tubule formation in the presence of sunitinib and that FGF2 can suppress sunitinib-induced retraction of tubules .\n\nImportantly , these effects of FGF2 were ablated by PD173074 , a small molecule inhibitor of FGF receptor signalling .\n\nWe also show that FGF2 can stimulate pro-angiogenic signalling pathways in endothelial cells despite the presence of sunitinib .\n\nFinally , analysis of clinical renal-cancer samples demonstrates that a large proportion of renal cancers strongly express FGF2 .\n\nWe suggest that therapeutic strategies designed to simultaneously target both VEGF and FGF2 signalling may prove more efficacious than sunitinib in renal cancer patients whose tumours express FGF2 .", "output": "Sustaining proliferative signaling" }, { "input": "Quercetin is a flavonoid with anticancer properties .\n\nIn this study , we examined the effects of quercetin on cell cycle , viability , and proliferation of cancer cells , either singly or in combination with the microtubule-targeting drugs taxol and nocodazole .\n\nAlthough quercetin induced cell death in a dose-dependent manner , 12.5-50 \u03bcM quercetin inhibited the activity of both taxol and nocodazole to induce G2/M arrest in various cell lines .\n\nQuercetin also partially restored drug-induced loss in viability of treated cells for up to 72 h .\n\nThis antagonism of microtubule-targeting drugs was accompanied by a delay in cell cycle progression and inhibition of the buildup of cyclin-B1 at the microtubule organizing center of treated cells .\n\nHowever , quercetin did not inhibit the microtubule targeting of taxol or nocodazole .\n\nDespite the short-term protection of cells by quercetin , colony formation and clonogenicity of HCT116 cells were still suppressed by quercetin or quercetin-taxol combination .\n\nThe status of cell adherence to growth matrix was critical in determining the sensitivity of HCT116 cells to quercetin .\n\nWe conclude that although long-term exposure of cancer cells to quercetin may prevent cell proliferation and survival , the interference of quercetin with cell cycle progression diminishes the efficacy of microtubule-targeting drugs to arrest cells at G2/M .", "output": "Sustaining proliferative signaling" }, { "input": "Numerous scientific studies have examined the relationship between coffee consumption and an array of medical conditions , including cancer , and yet the direct effect of commercially brewed coffee on cancer cells has not been evaluated .\n\nThe purpose of this study was to evaluate the antiproliferation effect of 4 different regular and decaffeinated coffee brews and 3 of coffee's bioactive ingredients-caffeine , chlorogenic acids , and caffeic acid-on 2 human ovarian cancer cell lines alone and in combination with cisplatin ( CDDP ) .\n\nAntiproliferation IC(50) for Brand A regular and decaffeinated coffee on A2780 cells was 1:70.79 \u00b1 5.66 and 1:55.68 \u00b1 2.00 dilution ( vol/vol ) in tissue culture medium ( mean \u00b1 standard error of the mean ; N = 12 ) , respectively , and slightly lower on A2780CP70 cells .\n\nThree other brands showed lower antiproliferation activity .\n\nAntiproliferation IC(50) concentrations of chlorogenic acids and caffeic acid are many folds lower than caffeine .\n\nIn combination with CDDP , both Brand A coffee brews , and the 3 bioactive compounds , showed additive antiproliferation effect on both cancer cell lines .\n\nFlow cytometry analysis showed that coffee treatment induced apoptosis of A2780 and A2780CP70 cells .\n\nTo our knowledge , this is the first report showing the antiproliferation activity and the additive effect with CDDP of commercially prepared coffee brews on human cancer cell lines .", "output": "Resisting cell death" }, { "input": "Over the past two decades , bioactive natural compounds have been shown to be a plausible adjunct to the treatment of breast cancer , the second leading cause of cancer death among American women .\n\nThis study was designed to investigate the effects of ursolic acid ( UA ) , a pentacyclic triterpene found in many foods and herbs , in a model of postmenopausal breast cancer .\n\nOvariectomized C57BL/6 mice ( n = 40 ) were randomized to receive control diet ( AIN-93G ) or diet supplemented with UA at 1 of 3 doses ( wt/wt ) : 0.05% , 0.10% , or 0.25% ( \u224854 , 106 , or 266 mg/kg body weight/day , respectively ) .\n\nAfter 3 wk , syngeneic MMTV-Wnt-1 mammary tumor cells were injected in the mammary fat pad , and mice continued on their respective diets for 5 more wk .\n\nAll UA doses decreased tumor cell proliferation , as assessed by Ki67 immunostaining ; nevertheless , UA at 0.10% was most effective in inhibiting tumor take and decreasing tumor final tumor size .\n\nModulation of Akt/mTOR signaling and induction of apoptosis appeared to mediate these effects on tumor growth .\n\nUA potently disrupted cell cycle progression and induced necrosis in a clonal MMTV-Wnt-1 mammary tumor cell line in vitro .\n\nThis study supports the potential of UA as an antitumorigenic agent .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Cell division and apoptosis are two crucial components of tumor biology and the importance of increased cell proliferation and reduced cell death have made them valid therapeutic targets .\n\nThe plant kingdom is a relatively underexploited cache of novel drugs , and crude extracts of plants are known for their synergistic activity .\n\nThe present study assessed the anti-proliferative activity of the medicinal plant Centrosema pubescens Benth .\n\nCentrosema pubescens dichloromethane extract ( CPDE ) inhibited the proliferation of HL-60 ( promyelocytic acute leukaemia ) cells with an IC\u2085\u2080 value of 5 \u03bcg/ml .\n\nFurther studies also showed that CPDE induces growth arrest at the G1 phase and specifically down-regulates the expressions of cyclin E and CDK2 and up-regulates p27(CKI) levels .\n\nThese events apparently lead to the induction of apoptosis , which was demonstrated qualitatively by a DNA fragmentation assay and propidium iodide staining .\n\nQuantitative assessment of the effective arrest of the cell cycle and of apoptosis was confirmed by flow cytometry .\n\nCPDE exhibited negligible cytotoxicity even at the highest dose tested ( 100 \u03bcg/ml ) in both normal peripheral blood mononuclear cells and in an in vitro model ( HL-60 ) .\n\nOur results strongly suggest that CPDE arrests the cell cycle at the G1 phase and triggers apoptosis by caspase activation .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "PURPOSE Steroid hormones and growth factors affect lung cancer , and it is possible they act in concert to influence patient outcome .\n\nEXPERIMENTAL DESIGN Primary lung tumors and normal lung tissue were analyzed for expression and localization of estrogen receptor \u03b1 and \u03b2-1 ( ER\u03b1 and ER\u03b2 ) , aromatase , progesterone receptor ( PR ) , and epidermal growth factor receptor ( EGFR ) .\n\nRESULTS Tumors expressed higher levels of ER\u03b2 compared to matched normal lung , whereas the reverse was true of PR .\n\nHigh cytoplasmic ER\u03b2 expression was identified as an independent negative prognostic predictor of overall survival ( OS ; HR = 1.67 ) , and low total PR was identified as an independent negative predictor of time to progression ( TTP ; HR = 1.59 ) .\n\nAfter adjusting for stage , age , sex , and smoking , combined high cytoplasmic ER\u03b2 and low total PR showed enhanced effects on OS ( HR = 2.64 ) and on TTP ( HR = 6.02 ) .\n\nFurther effects on OS were observed when EGFR expression was included ( HR = 5.32 ) .\n\nPatients with low cytoplasmic ER\u03b2 , low aromatase , low EGFR , and high total PR had shorter OS than patients with the opposite pattern ( HR = 6.60 ) .\n\nContribution of these markers to survival showed no significant sex differences in a multivariable model .\n\nER\u03b1 was elevated in tumors but was not predictive of survival , and appears to represent a variant ER\u03b1 protein that is only recognized by a C-terminal antibody .\n\nCONCLUSIONS Hormonal and EGFR pathways together may contribute to lung cancer prognosis .\n\nLung tumors with high ER\u03b2-1/low PR may define patients with aggressive biology .\n\nA validation study is necessary to fully assess the predictive value of these markers .", "output": "Sustaining proliferative signaling" }, { "input": "Although the p16(INK4a) and p21Waf1/Cip1 cyclin-dependent kinase ( CDK ) inhibitors are known to play key roles in cellular senescence in vitro , their roles in senescence remain rather poorly understood in vivo .\n\nThis situation is partly due to the possibility of compensatory effect(s) between p16INK4a and p21Waf1/Cip1 or to the upregulation of functionally related CDK inhibitors .\n\nTo directly address the cooperative roles of p16INK4a and p21Waf1/Cip1 in senescence in vivo , we generated a mouse line simply lacking both p16INK4a and p21Waf1/Cip1 genes [ double-knockout ( DKO) ] .\n\nMouse embryonic fibroblasts ( MEF ) derived from DKO mice displayed no evidence of cellular senescence when cultured serially in vitro .\n\nMoreover , DKO MEFs readily escaped Ras-induced senescence and overrode contact inhibition in culture .\n\nThis was not the case in MEFs lacking either p16INK4a or p21Waf1/Cip1 , indicating that p16(INK4a) and p21Waf1/Cip1 play cooperative roles in cellular senescence and contact inhibition in vitro .\n\nNotably , we found the DKO mice to be extremely susceptible to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-induced skin carcinogenesis that involves oncogenic mutation of the H-ras gene .\n\nMechanistic investigations suggested that the high incidence of cancer in DKO mice likely reflected a cooperative effect of increased benign skin tumor formation caused by p21Waf1/Cip1 loss , with increased malignant conversion of benign skin tumors caused by p16(INK4a) loss .\n\nOur findings establish an intrinsic cooperation between p16INK4a and p21Waf1/Cip1 in the onset of cellular senescence and tumor suppression in vivo .", "output": "Genomic instability and mutation, Enabling replicative immortality, Evading growth suppressors" }, { "input": "Genotoxic stress induces cell cycle arrest and DNA repair which may enable tumor cells to survive radiation therapy .\n\nHere , we defined the role of Ca(2+) signaling in the cell cycle control and survival of chronic myeloid leukemia ( CML ) cells subjected to ionizing radiation ( IR ) .\n\nTo this end , K562 erythroid leukemia cells were irradiated ( 0-10 Gy ) .\n\nTumor survival was analyzed by clonogenic survival assay and cell cycle progression via flow cytometry .\n\nPlasma membrane cation conductance was assessed by patch-clamp whole-cell recording and the cytosolic free Ca(2+) concentration ( [ Ca(2+)](i) ) was measured by fura-2 Ca(2+) imaging .\n\nNuclear activity of Ca(2+)/calmodulin-dependent kinase II ( CaMKII ) was defined by Western blotting .\n\nIn addition , the effect of IR ( 5 Gy ) on the cation conductance of primary CML cells was determined .\n\nThe results indicated that IR ( 10 Gy ) induced a G(2)/M cell cycle arrest of K562 cells within 24 h post-irradiation ( p.i. ) and decreased the clonogenic survival to 0.5 % of that of the control cells .\n\nIn K562 cells , G(2)/M cell cycle arrest was preceded by activation of TRPV5/6-like nonselective cation channels in the plasma membrane 1-5 h p.i. , resulting in an elevated Ca(2+) entry as evident from fura-2 Ca(2+) imaging .\n\nSimilarly , IR stimulated a Ca(2+)-permeable nonselective cation conductance in primary CML cells within 2-4 h p.i. .\n\nCa(2+) entry , into K562 cells was paralleled by an IR-induced activation of nuclear CaMKII .\n\nThe IR-stimulated accumulation in G(2) phase was delayed upon buffering [ Ca(2+)](i) with the Ca(2+) chelator BAPTA-AM or inhibiting CaMKII with KN93 ( 1 nM ) .\n\nIn addition , KN93 decreased the clonogenic survival of irradiated cells but not of control cells .\n\nIn conclusion , the data suggest that IR-stimulated cation channel activation , Ca(2+) entry and CaMKII activity participate in control of cell cycle progression and survival of irradiated CML cells .", "output": "Evading growth suppressors" }, { "input": "Periplocin is one of cardenolides isolated from cortex periplocae which is used for treatment of rheumatoid arthritis and reinforcement of bones and tendons in traditional medicine .\n\nHere , we investigated the anti-tumor activity of periplocin against lung cancer cells bothin vitro and in vivo , and explored its anti-cancer mechanism .\n\nPeriplocin inhibited the growth of lung cancer cells and induced their apoptosis in time- and dose-dependent manners by cell cycle arrest in G0/G1 phase .\n\nPeriplocin exhibited anti-tumor activity both in human ( A549 ) and mouse ( LL/2 ) lung cancer xenograft models .\n\nImmunohistochemical analysis revealed that intratumoral angiogenesis was significantly suppressed .\n\nFurthermore , anti-cancer activity mediated by periplocin was associated with decreased level of phosphorylated AKT and ERK both in vitro and in vivo , which were important for cell growth and survival .\n\nMoreover , periplocin induced apoptosis by downregulating Bcl-2 and upregulating Bax , leading to activation of caspase-3 and caspase-9 .\n\nThese findings suggested that periplocin could inhibit the growth of lung cancer both in vitro and in vivo , which could be attributed to the inhibition of proliferation and the induction of apoptosis signaling pathway , such as AKT and ERK .\n\nThese observations provide further evidence on the anti-tumor effect of periplocin , and it may be of importance to further explore its potential role as a therapeutic agent for cancer .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Landmark studies of the status of DNA damage checkpoints and associated repair functions in preneoplastic and neoplastic cells has focused attention on importance of these pathways in cancer development , and inhibitors of repair pathways are in clinical trials for treatment of triple negative breast cancer .\n\nCancer heterogeneity suggests that specific cancer subtypes will have distinct mechanisms of DNA damage survival , dependent on biological context .\n\nIn this study , status of DNA damage response ( DDR)-associated proteins was examined in breast cancer subtypes in association with clinical features ; 479 breast cancers were examined for expression of DDR proteins \u03b3H2AX , BRCA1 , pChk2 , and p53 , DNA damage-sensitive tumor suppressors Fhit and Wwox , and Wwox-interacting proteins Ap2\u03b1 , Ap2\u03b3 , ErbB4 , and correlations among proteins , tumor subtypes , and clinical features were assessed .\n\nIn a multivariable model , triple negative cancers showed significantly reduced Fhit and Wwox , increased p53 and Ap2\u03b3 protein expression , and were significantly more likely than other subtype tumors to exhibit aberrant expression of two or more DDR-associated proteins .\n\nDisease-free survival was associated with subtype , Fhit and membrane ErbB4 expression level and aberrant expression of multiple DDR-associated proteins .\n\nThese results suggest that definition of specific DNA repair and checkpoint defects in subgroups of triple negative cancer might identify new treatment targets .\n\nExpression of Wwox and its interactor , ErbB4 , was highly significantly reduced in metastatic tissues vs. matched primary tissues , suggesting that Wwox signal pathway loss contributes to lymph node metastasis , perhaps by allowing survival of tumor cells that have detached from basement membranes , as proposed for the role of Wwox in ovarian cancer spread .", "output": "Genomic instability and mutation, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "The incidence of oral squamous cell carcinoma ( SCC ) is increasing but the long-term survival rate remains low .\n\nAn animal model would therefore be helpful for evaluation of new treatment modalities for oral SCC .\n\nHamster is small animal , therefore , the cancer of hamster cheek pouch is not optimal for tumor imaging .\n\nThe VX2 cell line has been used in many carcinoma-related studies , including oral SCC research , but it is derived from cutaneous tissue and not mucosa .\n\nWe chemically induced tongue squamous cell carcinoma in rabbits and subsequently established a rabbit squamous cell line .\n\nThe cells grew in multiple layers without contact inhibition for 60 passages over 2 years and were positive for cytokeratin ( CK ) .\n\nElectron microscopy revealed that cells were polygonal with rich microvilli on the surface , and there were desmosomes between cells and bundles of tonofibril beside the cell membrane .\n\nThe chromosome number ranged from 71 to 272 , with a modal value of 145 ( 12.4% ) .\n\nThe cells were transplantable into nude mice subcutaneously or rabbit submucosally and produced carcinomas in all the animals .\n\nThe cell line should be a useful tool for the study of the biological characteristics of oral SCC , especially tongue SCC .", "output": "Evading growth suppressors" }, { "input": "Rhabdoid tumors have been reported in many different anatomic sites as an aggressive tumor and usually present with a rhabdoid tumor component ( a composite tumor ) rather than a pure rhabdoid tumor .\n\nRhabdoid tumor in the prostate has been described only once in the prostatic region as a possible epithelial origin .\n\nRhabdoid features in prostatic stromal sarcomas ( PSSs ) have never been described in the literature .\n\nHere , we report a case of a PSS with rhabdoid features .\n\nA 31-year-old man presented with a 4-month history of voiding difficulty and anal pain .\n\nComputed tomography of the abdomen revealed an ovoid mass in the prostate invading rectum and urinary bladder .\n\nA needle biopsy was diagnosed as an unclassified spindle cell sarcoma , and 2 cycles of adriamycin-based neoadjuvant chemotherapy were given , followed by radical prostatectomy .\n\nThe prostatectomy specimen revealed a high-grade sarcoma with fascicles of highly cellular spindle cells and numerous mitoses with hemorrhage and necrosis .\n\nIn areas , the tumor also contained sheets of loosely cohesive epithelioid cells with rhabdoid tumor component .\n\nBoth spindle and rhabdoid tumor cells were positive for vimentin , CD34 , and progesterone receptor and negative for desmin and cytokeratin immunostainings .\n\nThe rhabdoid tumor cells retained INI1 expression .\n\nThe tumor recurred in the bladder , and the patient died of sepsis .\n\nTo the best of our knowledge , this is the first case of PSS with rhabdoid features .\n\nThe tumor showed an aggressive clinical behavior with a short-term survival ( 7 months after diagnosis ) .", "output": "Resisting cell death" }, { "input": "HGF signaling induces epithelial cells to disassemble cadherin-based adhesion and increase cell motility and invasion , a process termed epithelial-mesenchymal transition ( EMT ) .\n\nEMT plays a major role in cancer metastasis , allowing individual cells to detach from the primary tumor , invade local tissue , and colonize distant tissues with new tumors .\n\nWhile invasion of vascular and lymphatic networks is the predominant route of metastasis , nerves also can act as networks for dissemination of cancer cell to distant sites in a process termed perineual invasion ( PNI ) .\n\nSignaling between nerves and invasive cancer cells remains poorly understood , as does cellular decision making that selects the specific route of invasion .\n\nHere we examine how HGF signaling contributes to PNI using reductionist culture model systems .\n\nWe find that TGF\u03b2 , produced by PC12 cells , enhances scattering in response to HGF stimulation , increasing both cell-cell junction disassembly and cell migration .\n\nFurther , gradients of TGF\u03b2 induce migratory mesenchymal cells to undergo chemotaxis towards the source of TGF\u03b2 .\n\nInterestingly , VEGF suppresses TGF\u03b2-induced enhancement of scattering .\n\nThese results have broad implications for how combinatorial growth factor signaling contributes to cancer metastasis , suggesting that VEGF and TGF\u03b2 might modulate HGF signaling to influence route selection during cancer progression .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Metastatic cancer cells typically fail to halt migration on contact with non-cancer cells .\n\nThis invasiveness is in contrast to normal mesenchymal cells that retract on contact with another cell .\n\nWhy cancer cells are defective in contact inhibition of locomotion is not understood .\n\nHere , we analyse the dynamics of prostate cancer cell lines co-cultured with fibroblasts , and demonstrate that a combinatorial code of Eph receptor activation dictates whether cell migration will be contact inhibited .\n\nThe unimpeded migration of metastatic PC-3 cells towards fibroblasts is dependent on activation of EphB3 and EphB4 by ephrin-B2 , which we show activates Cdc42 and cell migration .\n\nKnockdown of EphB3 and EphB4 restores contact inhibition of locomotion to PC-3 cells .\n\nConversely , homotypic collisions between two cancer cells results in contact inhibition of locomotion , mediated by EphA-Rho-Rho kinase ( ROCK ) signalling .\n\nThus , the migration of cancer cells can switch from restrained to invasive , depending on the Eph-receptor profile of the cancer cell and the reciprocal ephrin ligands expressed by neighbouring cells .", "output": "Evading growth suppressors, Activating invasion and metastasis" }, { "input": "An epithelial cell line , referred to as A163 , was established from breast carcinoma derived from a patient with a strong family history of breast cancer but no known breast cancer susceptibility mutation .\n\nA163 was propagated in a serum-free culture medium including the epidermal growth factor .\n\nImmunophenotypic characterization demonstrated a mixed luminal and basal-like phenotype .\n\nWhen epidermal growth factor was excluded from the culture medium , A163 entered a quiescent period followed by a period of increased cell proliferation in a subpopulation of the cells .\n\nThe epidermal growth factor-independent subpopulation retained the basal-like phenotype of the parental cell line .\n\nKaryotype and fluorescent in situ hybridization analysis showed an amplification of epidermal growth factor receptor on 7q in A163-S1 only , resulting in high expression of total and phosphorylated epidermal growth factor receptor .\n\nThe A163-S1 sub-line piles up in culture , indicating a loss of contact inhibition .\n\nWhen grown on transwell filters , A163 shows basal expression of P63 and cytokeratin 14 , whereas A163-S1 expresses P63 ubiquitously , and has lost the basal specific expression of cytokeratin 14 , indicating a loss of polarity .\n\nFurthermore , when cultured in reconstituted basement membrane matrix , A163 form polarized normal like acini .\n\nIn contrast , A163-S1 form large disorganized structures with lack of polarity .\n\nThese cell lines may prove useful to understand molecular changes in breast cancer progression , in particular basal-like breast cancer subtype with bad prognosis and no current treatment options .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "C-reactive protein is produced in response to cytokines such as interleukin ( IL)-6 .\n\nIt is known that increased plasma IL-6 levels induce increased hepatic and intratumoral production of C-reactive protein .\n\nCyclooxygenase enzyme-2 is induced by various stimuli , including inflammation and various growth factors .\n\nExpression of these two markers has not been well studied in clear cell renal cell carcinoma .\n\nThe objective of this study is to correlate the expression of C-reactive protein and cyclooxygenase enzyme-2 in clear cell renal cell carcinoma with pathologic parameters .\n\nA search of the surgical pathology and consultation files at our institution was performed for nephrectomy specimens with clear cell renal cell carcinoma from 2007 to 2008 .\n\nImmunohistochemical stains for C-reactive protein and cyclooxygenase enzyme-2 were performed .\n\nStaining intensity was graded as 0 , 1+ , 2+ , and 3+ .\n\nThe staining intensity was then correlated with pathologic stage and Fuhrman nuclear grade for each case .\n\nA total of 110 cases were identified .\n\nStrong expression of C-reactive protein was associated with higher Fuhrman nuclear grade and pathologic stage , and the strength of correlation was statistically significant ( p\u2009=\u20090.01 and p\u2009=\u20090.001 ) , respectively .\n\nHowever , cyclooxygenase enzyme-2 expression did not show statistically significant correlation with both pathologic stage and Fuhrman nuclear grade ( p\u2009=\u20090.1 and p\u2009=\u20090.15 ) , respectively .\n\nTo our knowledge , this is the largest study to date correlating the expression of both C-reactive protein and cyclooxygenase enzyme-2 in tissue with pathologic parameters in patients with clear cell renal cell carcinoma , which could have significant prognostic and therapeutic implications .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND Adult human mesenchymal stem cells ( hMSC ) have been shown to home to sites of carcinoma and affect biological processes , including tumour growth and metastasis .\n\nPrevious findings have been conflicting and a clear understanding of the effects of hMSCs on cancer remains to be established .\n\nTherefore , we set out to investigate the impact of hMSCs on the oestrogen receptor positive , hormone-dependent breast carcinoma cell line MCF-7 .\n\nRESULTS In this study , we show the effects of hMSCs on cancer cells are mediated through a secreted factor(s) which are enhanced by cancer cell-hMSC contact/communication .\n\nIn addition to enhanced proliferation when in co-culture with hMSCs , MCF-7 cells were found to have increased migration potential in vitro .\n\nInhibition of ER signalling by the pure anti-oestrogen ICI 182,780 decreased the effect of hMSCs on MCF-7 cell proliferation and migration supporting a role for ER signalling in the hMSC/MCF-7 cell interaction .\n\nAdditionally , hMSCs have been shown to secrete a wide variety of growth factors and chemokines including stromal cell-derived factor-1 ( SDF-1 ) .\n\nThis coupled with the knowledge that SDF-1 is an ER-mediated gene linked with hormone-independence and metastasis led to the investigation of the SDF-1/CXCR4 signalling axis in hMSC-MCF-7 cell interaction .\n\nExperiments revealed an increase in SDF-1 gene expression both in vivo and in vitro when MCF-7 cells were cultured with hMSCs .\n\nSDF-1 treatment of MCF-7 cells alone increased proliferation to just below that seen with hMSC co-culture .\n\nAdditionally , blocking SDF-1 signalling using a CXCR4-specific inhibitor decreased hMSC induced proliferation and migration of MCF-7 .\n\nHowever , the combined treatment of ICI and AMD3100 reduced MCF-7 cell proliferation and migration below control levels , indicating targeting both the ER and CXCR4 pathways is effective in decreasing the hMSCs induction of MCF-7 cell proliferation and migration .\n\nCONCLUSIONS The sum of these data reveals the relationship between tumour microenvironment and tumour growth and progression .\n\nBetter understanding of the mechanisms involved in this tumour stroma cell interaction may provide novel targets for the development of treatment strategies for oestrogen receptor positive , hormone-independent , and endocrine-resistant breast carcinoma .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "BACKGROUND Human T-cell leukemia virus type I ( HTLV-I ) has efficiently adapted to its host and establishes a persistent infection characterized by low levels of viral gene expression and slow proliferation of HTLV-I infected cells over decades .\n\nWe have previously found that HTLV-I p30 is a negative regulator of virus expression .\n\nRESULTS In this study we show that p30 targets multiple cell cycle checkpoints resulting in a delayed entry into S phase .\n\nWe found that p30 binds to cyclin E and CDK2 and prevents the formation of active cyclin E-CDK2 complexes .\n\nIn turn , this decreases the phosphorylation levels of Rb and prevents the release of E2F and its transcriptional activation of genes required for G1/S transition .\n\nOur studies also show that HTLV-II p28 does not bind cyclin E and does not affect cell cycle progression .\n\nCONCLUSIONS In contrast to HTLV-I , the HTLV-II-related retrovirus is not oncogenic in humans .\n\nHere we report that the HTLV-I p30 delays cell cycle progression while its homologue , HTLV-II p28 , does not , providing evidence for important differences between these two related retrovirus proteins .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Multiple myeloma is characterized by the clonal expansion of malignant plasma cells ( multiple myeloma cells [ MMCs] ) , in the bone marrow .\n\nOsteolytic bone lesions are detected in 80% of patients because of increased osteoclastic bone resorption and reduced osteoblastic bone formation .\n\nMMCs are found closely associated with sites of increased bone resorption .\n\nOsteoclasts strongly support MMC survival in vitro .\n\nTo further elucidate the mechanisms involved in osteoclast/MMC interaction , we have identified 552 genes overexpressed in osteoclasts compared with other bone marrow cell subpopulations .\n\nOsteoclasts express specifically genes coding for 4 CCR2-targeting chemokines and genes coding for MMC growth factors .\n\nAn anti-CCR2 monoclonal antibody blocked osteoclast chemoattractant activity for MMC , and CCR2 chemokines are also MMC growth factors , promoting mitogen-activated protein kinase activation in MMC .\n\nAn anti-insulin growth factor-1 receptor monoclonal antibody completely blocked the osteoclast-induced survival of MMC suppressing both osteoclast and MMC survival .\n\nSpecific a proliferation-inducing ligand or IL-6 inhibitors partially blocked osteoclast-induced MMC survival .\n\nThese data may explain why newly diagnosed patients whose MMC express high levels of CCR2 present numerous bone lesions .\n\nThis study displays additional mechanisms involved in osteoclast/MMC interaction and suggests using CCR2 and/or insulin growth factor-1 targeting strategies to block this interaction and prevent drug resistance .", "output": "Sustaining proliferative signaling" }, { "input": "Many tumors contain heterogeneous populations of cells , only some of which exhibit increased tumorigenicity and resistance to anticancer therapies .\n\nEvidence suggests that these aggressive cancer cells , often termed \" cancer stem cells \" or \" cancer stem-like cells \" ( CSCs ) , rely upon developmental signaling pathways that are important for survival and expansion of normal stem cells .\n\nHere we report that , in analogy to embryonic mammary epithelial biology , estrogen signaling expands the pool of functional breast CSCs through a paracrine FGF/FGFR/Tbx3 signaling pathway .\n\nEstrogen or FGF9 pretreatment induced CSC properties of breast cancer cell lines and freshly isolated breast cancer cells , whereas cotreatment of cells with tamoxifen or a small molecule inhibitor of FGFR signaling was sufficient to prevent the estrogen-induced expansion of CSCs .\n\nFurthermore , reduction of FGFR or Tbx3 gene expression was able to abrogate tumorsphere formation , whereas ectopic Tbx3 expression increased tumor seeding potential by 100-fold .\n\nThese findings demonstrate that breast CSCs are stimulated by estrogen through a signaling pathway that similarly controls normal mammary epithelial stem cell biology .", "output": "Sustaining proliferative signaling" }, { "input": "Osteoblastic bone metastases are the most common metastases produced by human prostate cancers ( PCa ) .\n\nDeregulated activity of Wnt growth factors resulting from overexpression of the Wnt inhibitor Dickkopf-1 ( DKK-1 ) is known to contribute to formation of the osteoblastic component of PCa skeletal bone metastases .\n\nIn this study , we report that DKK-1 knockdown in osteolytic human PCa cells unexpectedly delays the development of both soft tissue and osseous lesions .\n\nPCa cells deficient in DKK-1 expression did not increase canonical Wnt signaling in target osteoblast cell lines ; however , DKK-1 knockdown PCa cells exhibited increased expression of the CDK inhibitor p21(CIP1/WAF1) and a 32% increase in G(1) arrest compared with control cells .\n\nAblating p21(CIP1/WAF1) in PCa cells deficient in DKK-1 was sufficient to rescue tumor growth .\n\nCollectively , our findings demonstrate that DKK-1 overexpression supports tumor growth in part by restricting expression of p21(CIP1/WAF1) through a mechanism independent of canonical Wnt signaling .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND To identify potential tumor suppressor genes , genome-wide data from exome and transcriptome sequencing were combined to search for genes with loss of heterozygosity and allele-specific expression .\n\nThe analysis was conducted on the breast cancer cell line HCC1954 , and a lymphoblast cell line from the same individual , HCC1954BL .\n\nRESULTS By comparing exome sequences from the two cell lines , we identified loss of heterozygosity events at 403 genes in HCC1954 and at one gene in HCC1954BL .\n\nThe combination of exome and transcriptome sequence data also revealed 86 and 50 genes with allele specific expression events in HCC1954 and HCC1954BL , which comprise 5.4% and 2.6% of genes surveyed , respectively .\n\nMany of these genes identified by loss of heterozygosity and allele-specific expression are known or putative tumor suppressor genes , such as BRCA1 , MSH3 and SETX , which participate in DNA repair pathways .\n\nCONCLUSIONS Our results demonstrate that the combined application of high throughput sequencing to exome and allele-specific transcriptome analysis can reveal genes with known tumor suppressor characteristics , and a shortlist of novel candidates for the study of tumor suppressor activities .", "output": "Genomic instability and mutation" }, { "input": "It has been demonstrated that growth factors produced by breast cancer cells stimulate aromatase expression in both breast cancer and adjacent adipose fibroblasts and stromal cells .\n\nHowever , whether these growth factors affect aromatase activity by other mechanisms still remain unclear .\n\nIn the current study , MCF-7aro and T47Daro aromatase transfected breast carcinoma cells were used to explore the mechanisms of post-transcriptional regulation of aromatase activity by growth factor pathways .\n\nOur study reveals that PI3K/Akt and MAPK inhibitors suppressed aromatase activity in MCF-7aro cells .\n\nHowever , PI3K/Akt pathway inhibitors stimulated aromatase activity in T47Daro cells .\n\nThis is due to enhanced MAPK phosphorylation as compensation after the PI3K/Akt pathway has been blocked .\n\nIGF-1 treatment increased aromatase activity in both breast cancer cell lines .\n\nIn addition , LTEDaro cells ( long-term estrogen deprived MCF-7aro cells ) which have enhanced MAPK activity , show higher aromatase activity compared to parental MCF-7aro cells , but the aromatase protein level remains the same .\n\nThese results suggest that aromatase activity could be enhanced by growth factor signaling pathways via post-transcriptional mechanisms .", "output": "Sustaining proliferative signaling" }, { "input": "Hepatocellular carcinoma ( HCC ) remains a common malignant cancer worldwide .\n\nThere is an urgent need to identify new molecular targets for the development of novel therapeutic approaches .\n\nHerein , we review the structure , function and biology of glypican-3 ( GPC3 ) and its role in human cancer with a focus on its potential as a therapeutic target for immunotherapy .\n\nGPC3 is a cell-surface protein that is over-expressed in HCC .\n\nLoss-of-function mutations of GPC3 cause Simpson-Golabi-Behmel syndrome ( SGBS ) , a rare X-linked overgrowth condition .\n\nGPC3 binds Wnt and Hedgehog ( Hh ) signalling proteins .\n\nGPC3 is also able to bind basic growth factors such as fibroblast growth factor 2 through its heparan sulphate glycan chains .\n\nGPC3 is a promising candidate for liver cancer therapy given that it shows high expression in HCC .\n\nAn anti-GPC3 monoclonal antibody has shown anti-cancer activity in mice and its humanised IgG molecule is currently undergoing clinical evaluation in patients with HCC .\n\nThere is also evidence that soluble GPC3 may be a useful serum biomarker for HCC .", "output": "Sustaining proliferative signaling" }, { "input": "A better understanding of the underlying mechanisms of DNA repair after exposure to ionizing radiation represents a research priority aimed at improving the outcome of clinical radiotherapy .\n\nBecause of the close association with DNA double strand break ( DSB ) repair , phosphorylation of the histone H2AX protein ( \u03b3H2AX ) , quantified by immunodetection , has recently been used as a method to study DSB induction and repair at low and clinically relevant radiation doses .\n\nHowever , the lack of consistency in literature points to the need to further validate the role of H2AX phosphorylation in DSB repair and the use of this technique to determine intrinsic radiosensitivity .\n\nIn the present study we used human mammary epithelial MCF10A cells , characterized by a radiosensitive phenotype due to reduced levels of the Ku70 and Ku80 repair proteins , and investigated whether this repair-deficient cell line displays differences in the phosphorylation pattern of H2AX protein compared to repair-proficient MCF10A cells .\n\nThis was established by measuring formation and disappearance of \u03b3H2AX foci after irradiating synchronized cell populations with ( 60)Co \u03b3-rays .\n\nOur results show statistically significant differences in the number of \u03b3H2AX foci between the repair-deficient and -proficient cell line , with a higher amount of \u03b3H2AX foci present at early times post-irradiation in the Ku-deficient cell line .\n\nHowever , the disappearance of those differences at later post-irradiation times questions the use of this assay to determine intrinsic radiosensitivity , especially in a clinical setting .", "output": "Genomic instability and mutation" }, { "input": "The radioprotective effects of dimethyl sulfoxide ( DMSO ) have been known for many years , and the suppression of hydroxyl ( OH ) radicals induced by ionizing radiation has been thought to be the main cause of this effect .\n\nHowever , the DMSO concentration used was very high , and might be toxic , in earlier studies .\n\nIn the present study , we administered a lower , non-toxic concentration ( 0.5% , i.e. , 64 mM ) of DMSO before irradiation and examined its radioprotective effects .\n\nColony formation assay and micronucleus assay showed significant radioprotective effects in CHO , but not in xrs5 , which is defective in the repair function of DNA double-strand breaks .\n\nThe levels of phosphorylated H2AX and the formation of 53BP1 foci 15 minutes after irradiation , which might reflect initial DNA double-strand breaks , in DMSO-treated CHO cells were similar to those in non-treated cells , suggesting that the radioprotective effects were not attributable to the suppression of general indirect action in the lower concentration of DMSO .\n\nOn the other hand , 2 hours after irradiation , the average number of 53BP1 foci , which might reflect residual DNA double-strand breaks , was significantly decreased in DMSO-treated CHO cells compared to non-treated cells .\n\nThe results indicated that low concentration of DMSO exerts radioprotective effects through the facilitation of DNA double-strand break repair rather than through the suppression of indirect action .", "output": "Genomic instability and mutation" }, { "input": "The aim of this study was to determine whether caffeine enhanced radiosensitivity of normal liver tissue in a rat radiation-induced liver disease model .\n\nBuffalo rat McA-RH7777 hepatocellular cancer cells and BRL3A normal liver cells were irradiated , and cell cycle distribution and apoptosis rates were analyzed .\n\nA rat model of radiation-induced liver disease was established , rats were randomized into four groups : control ; caffeine alone ; irradiation ( IR ) alone ; and caffeine plus IR ( Caff + IR ) group .\n\nApoptosis rates in normal rat liver tissue after IR were evaluated by TUNEL staining and caspase-3 Western blot .\n\nTransaminase activity was measured and histopathological examination was done after IR .\n\nCaffeine abrogated IR-induced G2 phase arrest ( Caff + IR vs. IR : 40.9 \u00b1 4.0 vs. 60.7 \u00b1 5.5% , at 12 h after IR ) and increased apoptosis rates ( Caff + IR vs. IR : 56.1 \u00b1 6.8 vs. 35.5 \u00b1 4.0% , at 72 h after IR ) in McA-RH7777 cells , but did not affect IR-induced G2 phase arrest and apoptosis rates at any time point after IR in BRL3A cells .\n\nCaffeine did not enhance apoptosis , transaminase activity , or histopathological injury of normal rat liver tissue at any time points after IR .\n\nThis study suggests that caffeine might not enhance radiosensitivity of normal liver tissue in vivo .\n\nIn an earlier study , we reported that caffeine enhanced radiosensitivity of human hepatocellular cancer in a nude mice model .\n\nTogether , these results offer feasibility of clinical application .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND IL-6 is a pro-inflammatory cytokine that signals via binding to a soluble or membrane bound receptor , while nitric oxide ( NO ) , an oxidative stress molecule , diffuses through the cell membrane without a receptor .\n\nBoth mediators signal through different mechanisms , yet they are dependent on NF\u03baB .\n\nWe proposed that both mediators are co-induced and co-regulated in inflamed mammary epithelial cells .\n\nMETHODS SCp2 mammary epithelial cells were treated with bacterial endotoxin ( ET ) for different time periods and analyzed for induction of IL-6 secretion and NO production by ELISA and Griess reaction , respectively .\n\nThe expression of IL-6 and induced NO synthase ( iNOS ) was assayed by real time PCR and/or western immunoblots , and the activation of NF\u03baB was assayed by immunobinding assay .\n\nTo investigate the role of mammary cell microenvironment ( cell-substratum or interaction of mammary epithelial cell types ; critical to mammary development , function , and disease ) in modulation of the inflammatory response , SCp2 cells were cultured with or without extracellular matrix ( EHS ) or in coculture with their myoepithelial counterpart ( SCg6 ) , and assayed for ET-induced IL-6 and NO .\n\nRESULTS Endotoxin induced NF\u03baB activation at 1 h after ET application .\n\nIL-6 secretion and NO production were induced , but with unexpected delay in expression of mRNA for iNOS compared to IL-6 .\n\nNF\u03baB/p65 activation was transient but NF\u03baB/p50 activation persisted longer .\n\nSelective inhibition of NF\u03baB activation by Wedelolactone reduced ET-induced expression of IL-6 mRNA and protein but not iNOS mRNA or NO production , suggesting differences in IL-6 and iNOS regulation via NF\u03baB .\n\nSCp2 cells in coculture with SCg6 but not in presence of EHS dramatically induced IL-6 secretion even in the absence of ET .\n\nET-induced NO production was blunted in SCp2/SCg6 cocultures compared to that in SCp2 alone .\n\nCONCLUSIONS The differential regulation of IL-6 and iNOS together with the differential activation of different NF\u03baB dimers suggest that IL-6 and iNOS are regulated by different NF\u03baB dimers , and differentially regulated by the microenvironment of epithelial cells .\n\nThe understanding of innate immune responses and inflammation in epithelia and linkage thereof is crucial for understanding the link between chronic inflammation and cancer in epithelial tissues such as the mammary gland .", "output": "Tumor promoting inflammation" }, { "input": "We recently showed that Nox4 NADPH oxidase is highly expressed in pancreatic ductal adenocarcinoma and that it is activated by growth factors and plays a pro-survival , anti-apoptotic role .\n\nHere we investigate the mechanisms through which insulin-like growth factor I and serum ( FBS ) activate NADPH oxidase in pancreatic cancer ( PaCa ) cells .\n\nWe show that in PaCa cells , NADPH oxidase is composed of Nox4 and p22(phox) catalytic subunits , which are both required for NADPH oxidase activity .\n\nInsulin-like growth factor I and FBS activate NADPH oxidase through transcriptional up-regulation of p22(phox) .\n\nThis involves activation of the transcription factor NF-\u03baB mediated by Akt kinase .\n\nUp-regulation of p22(phox) by the growth factors results in increased Nox4-p22(phox) complex formation and activation of NADPH oxidase .\n\nThis mechanism is different from that for receptor-induced activation of phagocytic NADPH oxidase , which is mediated by phosphorylation of its regulatory subunits .\n\nUp-regulation of p22(phox) represents a novel pro-survival mechanism through which growth factors and Akt inhibit apoptosis in PaCa cells .", "output": "Sustaining proliferative signaling" }, { "input": "The role of estrogen receptor beta ( ER\u03b2 ) in breast cancer is unclear .\n\nER\u03b2 is considered to have a protective role in breast cancer development based on findings demonstrating that ER\u03b2 expression inhibits ER\u03b1-mediated proliferation of breast cancer cells .\n\nWe previously demonstrated that ER\u03b2 causes a ligand independent G2 cell cycle arrest in MCF-7 cells .\n\nTo study the mechanisms of the ER\u03b2-mediated G2 cell cycle arrest , we investigated its effects on the regulatory pathways responsible for the G2/M phase transition .\n\nWe found that ER\u03b2 inhibits CDK1 activity , which is the critical determinant of the G2/M progression .\n\nCDK1 activity is modulated by both stimulatory and inhibitory factors .\n\nCyclin B1 is the major activator of CDK1 .\n\nER\u03b2 inhibited the cell cycle-dependent stimulation of cyclin B1 mRNA and protein .\n\nGADD45A and BTG2 are two major inhibitors of CDK1 , which have been implicated in breast tumor formation .\n\nBased on these findings , we explored if the expression pattern of GADD45A and BTG2 is affected by ER\u03b2 .\n\nWe found that ER\u03b2 stimulates GADD45A and BTG2 mRNA levels .\n\nThe induction of these two genes is caused by ER\u03b2 binding directly to these genes and recruiting c-jun and NCOA2 .\n\nOur findings demonstrated that unliganded ER\u03b2 causes a G2 cell cycle arrest by inactivating CDK1 through the repression of cyclin B1 and stimulation of GADD45A and BTG2 expression .\n\nThese results provide evidence that drugs that stimulate the production of unliganded ER\u03b2 may be effective new therapies to prevent breast cancer .", "output": "Sustaining proliferative signaling" }, { "input": "We investigated the priming effect and mechanism of granulocyte colony-stimulating factor ( G-CSF ) in chemotherapy with low-dose Ara-C and VP-16 for acute myeloid leukemia .\n\nWe analyzed cell proliferation , apoptosis , and cell cycle in vitro using leukemia cell lines 32Dcl3 , U937 , HL-60 , and Ba/F3 .\n\nCell proliferation assays were performed using the Trypan Blue dye exclusion method .\n\nFor detection of apoptosis , the Annexin V-binding capacity of treated cells was examined by flow cytometry .\n\nTo evaluate the cell cycle , we used an FITC BrdU Flow kit and conducted analysis by flow cytometry .\n\nThe combination of Ara-C and VP-16 significantly enhanced the observed effects compared with those of Ara-C or VP-16 alone .\n\nConcurrent administration of G-CSF further reduced the cell number and viability of 32Dcl3 and U937 cells , but not of HL-60 and Ba/F3 cells .\n\nApoptotic cells were significantly increased in number by the addition of G-CSF to 32Dcl3 and U937 cells , while G-CSF had no significant effect on HL-60 and Ba/F3 cell lines .\n\nThe addition of G-CSF significantly decreased the percentage of G0/G1-phase cells and significantly increased that of S-phase cells among 32Dcl3 and U937 cells .\n\nNo significant effect was observed for HL-60 and Ba/F3 cells .\n\nAn enhancement was confirmed for the combination of Ara-C , VP-16 , and G-CSF for 32Dcl3 and U937 cells but not for HL-60 and Ba/F3 cells .\n\nIt was thought that this difference was a result of different responses to G-CSF .\n\nG-CSF potentiates Ara-C- and VP-16-induced cytotoxicities through apoptosis induction by mobilizing resting G0-G1-phase cells into S phase .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "INTRODUCTION Clinical and experimental studies indicate oxygen free radicals and their reactive derivatives participation in formation of chronic inflammation states , which facilitate gastrointestinal tract tumors development .\n\nDuring malignant changes formation in epithelium of gastrointestinal tract increased oxygen radicals generation initiates lipid peroxidation and DNA and proteins oxidation processes .\n\nFinal lipid peroxidation products ( saturated and unsaturated aldehydes ) are highly reactive and characterized by greatly longer half life time than reactive oxygen species and capability to diffuse from places of their formation to distant cell areas .\n\nIn cells they react with important biological macromolecules such as DNA and proteins causing their structural and functional damages .\n\nThe effects of changes in cell membranes structure are increase in their permeability , depolarization , decrease of hydrophobicity and inhibition of enzymes , membrane channels and transporters .\n\nThese changes lead to the loss of cell integrity .\n\nSTUDY AIM Determination of lipid peroxidation level in blood serum patients with gastrointestinal tract tumors .\n\nMATERIALS AND METHODS Materials for studies were obtained from 170 patients with gastrointestinal tract tumors : 10 with stomach cancer , 20 with pancreatic cancer , 30 with primary liver cancer , 60 with primary colorectal cancer and 50 with metachronic colorectal cancer liver metastases .\n\nBlood was taken from patients 1 day before and 6 days after surgery .\n\nControl blood was obtained from 53 healthy blood donors .\n\nLipid peroxidation level was determined spectrophotometrically as a concentration of final lipid peroxidation products , which in reaction with tiobarbituric acid ( TBA ) form colour complex ( thiobarbituric acid-reactive substances , TBARS ) .\n\nRESULTS Higher lipid peroxidation level was observed in pre- and postoperative blood sera patients with gastrointestinal tumors in comparison to serum from healthy blood donors .\n\nCONCLUSIONS Increased lipid peroxidation level in peripheral blood of patients with gastrointestinal tract tumors is evidence of intensive oxidative stress and might indicate impairment of antioxidant defense mechanisms in organism .", "output": "Tumor promoting inflammation" }, { "input": "Tobacco-induced oxidative stress leads to chronic inflammation and is implicated in the development of many human epithelial cancers , including head and neck cancer .\n\nCigarette smoke exposure was shown to induce the expression of the \u0394Np63\u03b1 and nitric oxide synthase ( NOS)-2 in head and neck squamous cell carcinoma cells and immortalized oral keratinocytes .\n\nThe NOS2 promoter was found to contain various cognate sequences for several transcription factors including interferon regulatory factor ( IRF)-6 and p63 , which were shown in vivo binding to the NOS2 promoter in response to smoke exposure .\n\nSmall interfering ( si)-RNAs against both \u0394Np63\u03b1 and IRF6 decreased the induction of NOS2 promoter-driven reporter luciferase activity and were shown to inhibit NOS2 activity .\n\nFurthermore , both mainstream ( MSE ) and sidestream ( SSE ) smoking extracts induced changes in expression of autophagic marker , LC3B , while siRNA against \u0394Np63\u03b1 , IRF6 and NOS2 modulated these autophagic changes .\n\nOverall , these data support the notion that \u0394Np63\u03b1/IRF6 interplay regulates NOS2 transcription , thereby underlying the autophagic-related cancer cell response to tobacco exposure .", "output": "Resisting cell death" }, { "input": "Heat shock protein 90 ( Hsp90 ) is a molecular chaperone involved in folding and stabilizing multiple intracellular proteins that have roles in cell activation and proliferation .\n\nMany Hsp90 client proteins in tumor cells are mutated or overexpressed oncogenic proteins driving cancer cell growth , leading to the acceptance of Hsp90 as a potential therapeutic target for cancer .\n\nBecause several signal transduction molecules that are dependent on Hsp90 function are also involved in activation of innate and adaptive cells of the immune system , we investigated the mechanism by which inhibiting Hsp90 leads to therapeutic efficacy in rodent models of inflammation and autoimmunity .\n\nEC144 , a synthetic Hsp90 inhibitor , blocked LPS-induced TLR4 signaling in RAW 264.7 cells by inhibiting activation of ERK1/2 , MEK1/2 , JNK , and p38 MAPK but not NF-\u03baB .\n\nEx vivo LPS-stimulated CD11b(+) peritoneal exudate cells from EC144-treated mice were blocked from phosphorylating tumor progression locus 2 , MEK1/2 , and ERK1/2 .\n\nConsequently , EC144-treated mice were resistant to LPS administration and had suppressed systemic TNF-\u03b1 release .\n\nInhibiting Hsp90 also blocked in vitro CD4(+) T cell proliferation in mouse and human MLRs .\n\nIn vivo , semitherapeutic administration of EC144 blocked disease development in rat collagen-induced arthritis by suppressing the inflammatory response .\n\nIn a mouse collagen-induced arthritis model , EC144 also suppressed disease development , which correlated with a suppressed Ag-specific Ab response and a block in activation of Ag-specific CD4(+) T cells .\n\nOur results describe mechanisms by which blocking Hsp90 function may be applicable to treatment of autoimmune diseases involving inflammation and activation of the adaptive immune response .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Monoclonal antibodies ( mAbs ) to HER2 are currently used to treat breast cancer , but low clinical efficacy , along with primary and acquired resistance to therapy , commonly limit clinical applications .\n\nWe previously reported that combinations of antibodies directed at non-overlapping epitopes of HER2 are endowed with enhanced antitumor effects , probably due to accelerated receptor degradation .\n\nHere , we extend these observations to three-dimensional mammary cell models , and compare the effects of single mAbs with the effects of antibody combinations .\n\nCollectively , our in vitro assays and computational image analyses indicate that combining mAbs against different epitopes of HER2 better inhibits invasive growth .\n\nImportantly , while growth factors are able to reduce intraluminal apoptosis and induce an invasive phenotype , combinations of mAbs better than single mAbs can reverse the growth factor-induced phenotypes of HER2-overexpressing spheroids .\n\nIn conclusion , our studies propose that mAb combinations negate the biological effects of growth factors on invasive growth of HER2-overexpressing cells .\n\nHence , combining mAbs offers a therapeutic strategy , potentially able to enhance clinical efficacy of existing antireceptor immunotherapeutics .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Construction industry workers are exposed to many known carcinogens in their complex occupational environment .\n\nSince there are no past studies on genotoxicity among this group in the Indian subcontinent , workers engaged in different construction sites at Coimbatore , Tamil Nadu , India , were assessed here .\n\nWe enrolled 96 workers and 68 control subjects with similar mean age , smoking , tobacco chewing prevalence and alcohol consumption , for analysis of DNA damage in blood leucocytes by micronucleus ( MN ) and comet assays .\n\nDNA repair inhibition was also analyzed by assessing the XPD gene .\n\nConstruction workers showed a significant increase in MN and comet tail length compared to controls with adjustment for smoking habits , tobacco chewing , alcohol consumption and years of exposure ( P<0.05 ) .\n\nThe results indicated that chronic occupational exposure to cement during construction work could lead to increased levels of DNA damage and repair inhibition .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Chronic lymphocytic leukemia cells show prolonged survival in vivo , but rapidly die by spontaneous apoptosis in vitro , unless they are co-cultured with stromal cells or non-malignant leukocytes .\n\nThe objective of this study was to characterize the survival-inducing cross-talk of chronic lymphocytic leukemia cells with their microenvironment to identify novel therapeutic targets .\n\nDESIGN AND METHODS We analyzed and compared microarray-based expression profiles of chronic lymphocytic leukemia cells before and after three different survival-inducing culture conditions : ( i ) stromal cell co-culture , ( ii ) stromal cell conditioned medium and ( iii ) high cell density cultures of unsorted peripheral blood mononuclear cells .\n\nCytokine antibody arrays were applied to study the composition of soluble factors present in these cultures .\n\nRESULTS The different survival-supportive culture conditions induced distinct gene expression changes , the majority of which were common to all three conditions .\n\nPathway analyses identified - in addition to known signaling networks in chronic lymphocytic leukemia - novel pathways , of which Toll-like receptor signaling , nuclear respiratory factor-2 ( NRF2)-mediated oxidative stress response , and signaling via triggering receptor expressed on myeloid cells-1 ( TREM1 ) were the most relevant .\n\nA high proportion of up-regulated genes were inflammatory cytokines , of which chemokine ( C-C motif ) ligand 2 ( CCL2 ) was shown to be induced in monocytes by the presence of chronic lymphocytic leukemia cells in vitro .\n\nIn addition , increased serum levels of this chemokine were detected in patients with chronic lymphocytic leukemia .\n\nCONCLUSIONS Our data provide several lines of evidence that an inflammatory microenvironment is induced in survival-supportive cultures of chronic lymphocytic leukemia cells which might be directly or indirectly involved in the prolonged survival of the malignant cells .", "output": "Tumor promoting inflammation" }, { "input": "PURPOSE BMP-6 , which belongs to the TGF-\u03b2 superfamily , is a multifunctional molecule with distinct abilities in embryogenesis and organogenesis .\n\nOur recent research has implied that BMP-6 may suppress breast cancer metastasis .\n\nIn the present study , we extended to elucidate the molecular mechanism by which BMP-6 exerts its anti-tumorigenic effect .\n\nMETHODS The Boyden chamber assay was used to examine the ability of BMP-6 and HO-1 in MCF-7 malignant progress .\n\nRT-PCR , western blot , luciferase assay , and quantitative CHIP were used to determine the potential mechanism and signaling pathways by which BMP-6 and HO-1 function as anti-metastatic factors in MCF-7 cells .\n\nRESULTS The Boyden chamber assay showed that BMP-6 inhibited the migration and invasion of MCF-7 cells , which effect was significantly deprived by knockdown of HO-1 .\n\nWe further demonstrated that BMP-6 treatment resulted in an activation of HO-1 transcription through the recruitment of Smad1/5 to the Smad-responsive element on its promoter .\n\nIn addition , BMP-6-induced up-regulation of HO-1 exhibited an inhibitory effect on MMP-9 secretion in a paracrine action in MCF-7 cells .\n\nOverexpression of BMP-6 and HO-1 synergistically suppressed MMP-9 transcription , which effect was specifically mediated via the MAPK/p38/AP-1 signaling .\n\nHowever , blockade of HO-1 using ZnPPIX totally abolished BMP-6-regulated MMP-9 activation in MCF-7 cells .\n\nCONCLUSIONS These observations suggest a novel role of BMP-6/HO-1 cascade to relieve breast cancer metastasis by regulating the secretion of growth factors in tumor microenvironment .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "PURPOSE Germline mutations in BRCA1 result in a strong predisposition to breast cancer , with frequent loss of heterozygosity of the remaining wild-type allele .\n\nThe development of BRCA1 tumors is likely to depend on additional genetic alterations and gene expression changes which follow growth and DNA repair defects associated with BRCA1 deficiency .\n\nThe identification of these modifications offers an opportunity to find surrogate markers of BRCA1 tumors .\n\nHere , we sought to identify differentially expressed proteins related to BRCA1 depletion .\n\nEXPERIMENTAL DESIGN We used isogenic HeLa cells either stably knocked-down or not for BRCA1 ( BRCA1(KD) ) and compared protein profiles of these cells by DIGE .\n\nRESULTS We detected increased levels of Replication protein A2 ( RPA2 ) in BRCA1(KD) cells as compared to control cells .\n\nRPA2 is an essential protein required for DNA replication and repair .\n\nWe further demonstrated that depletion of RPA2 subunit delays growth of BRCA1(KD) respect to isogenic control cells .\n\nStrikingly , elevated levels of RPA2 were more frequently observed in BRCA1 tumors when triple-negative tumors from BRCA1 mutation carriers ( n=13 ) and non-carriers ( n=36 ) were stained in situ for RPA2 .\n\nCONCLUSIONS AND CLINICAL RELEVANCE RPA2 up-regulation may thus be involved in the growth and/or survival of BRCA1 tumor cells and useful in immunohistochemical discrimination of triple-negative BRCA1 tumors .", "output": "Genomic instability and mutation" }, { "input": "Deregulation of insulin-like growth factor-1 receptor ( IGF-1R ) is closely associated with malignant transformation and tumor cell survival in various cancers .\n\nWe found that IGF-1R expression level in leukemia cells positively correlated with the percentage of blast in bone marrow from de novo acute myeloid leukemia ( AML ) patients .\n\nMoreover , we showed that NVP-ADW742 , a novel small weight molecular inhibitor of IGF-IR , could induce apoptosis in both HL-60 cell line and primary AML blasts .\n\nHowever , no significant alteration of cell cycle was observed in HL-60 cells .\n\nFurther studies revealed that NVP-ADW742 induced Akt dephosphorylation , which might subsequently induce p38 phosphorylation and decrease antiapoptotic protein Bcl-2 expression in HL-60 cells .\n\nFinally , we demonstrated that NVP-ADW742 could synergize with Ara-C to induce the kill in a subset of drug-resistant AML specimens .\n\nWe suggested that IGF-lR targeting might be therapeutically beneficial for some AML patients .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND Epidemiologic data have shown that obesity independently increases colorectal cancer ( CRC ) risk , but the mechanisms are poorly understood .\n\nObesity is an inflammatory state , and chronic colonic inflammation induces CRC .\n\nOBJECTIVE We conducted this proof-of-principle study to seek evidence of obesity-associated colorectal inflammation and to evaluate effects of diet-induced weight loss .\n\nDESIGN We measured inflammatory cytokines , gene arrays , and macrophage infiltration in rectosigmoid mucosal biopsies of 10 obese premenopausal women [ mean \u00b1 SD body mass index ( in kg/m(2) ) : 35 \u00b1 3.5 ] before and after weight loss induced by a very-low-calorie diet .\n\nRESULTS Subjects lost a mean ( \u00b1SD ) of 10.1 \u00b1 1% of their initial weight .\n\nWeight loss significantly reduced fasting blood glucose , total cholesterol , triglycerides , LDL , tumor necrosis factor-\u03b1 ( TNF-\u03b1 ) , and interleukin ( IL)-8 concentrations ( P < 0.05 ) .\n\nAfter weight loss , rectosigmoid biopsies showed a 25-57% reduction in TNF-\u03b1 , IL-1\u03b2 , IL-8 , and monocyte chemotactic protein 1 concentrations ( P < 0.05 ) .\n\nT cell and macrophage counts decreased by 28% and 42% , respectively ( P < 0.05 ) .\n\nGene arrays showed dramatic down-regulation of proinflammatory cytokine and chemokine pathways , prostaglandin metabolism , and the transcription factors STAT3 ( signal transducer and activator of transcription 3 ) and nuclear transcription factor \u03baB .\n\nWeight loss reduced expression of FOS and JUN genes and down-regulated oxidative stress pathways and the transcription factors ATF ( activating transcription factor ) and CREB ( cyclic AMP response element-binding ) .\n\nCONCLUSIONS Our data show that diet-induced weight loss in obese individuals reduces colorectal inflammation and greatly modulates inflammatory and cancer-related gene pathways .\n\nThese data imply that obesity is accompanied by inflammation in the colorectal mucosa and that diet-induced weight loss reduces this inflammatory state and may thereby lower CRC risk .", "output": "Tumor promoting inflammation" }, { "input": "Recently , we identified the \" apoptotic ring, \" containing phosphorylated histone H2AX ( \u03b3-H2AX ) , as an early chromatin modification during apoptosis .\n\nBecause \u03b3-H2AX initiates the DNA damage response ( DDR ) , we tested whether the apoptotic H2AX response leads to the full recruitment of the DDR factors that normally coordinate DNA repair and cell-cycle checkpoints .\n\nWe show that the apoptotic H2AX response does not recruit the DDR factors because MDC1 ( mediator of DNA damage checkpoint protein 1 ) , which normally binds to \u03b3-H2AX in response to DNA damage and amplifies the DDR , is cleaved by caspase-3 .\n\nThis cleavage separates the BRCT and FHA domains of MDC1 and constitutes a novel mechanism for the inactivation of DNA repair in apoptotic cells .\n\nAlso , we show that downregulation of MDC1 increases the apoptotic response to TRAIL .\n\nTogether , these results implicate MDC1 in the cellular apoptotic response .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "SWAP-70 , a phosphatidylinositol trisphosphate ( PtdIns(3,4,5)P(3) ) binding protein , has been suggested to be involved in transformation of mouse embryo fibroblasts ( MEFs ) as well as membrane ruffling after growth factor stimulation of the cells .\n\nA mutant , SWAP-70-374 , was found to be able to bind to F-actin in vitro , whereas wild-type SWAP-70 failed to do so .\n\nThis mutant was present at the plasma membrane without any stimulation while the wild-type protein was present only in the cytosol unless cells were stimulated with EGF .\n\nExpression of this mutant in MEFs resulted in morphologic transformation , fast growth , and loss of contact inhibition , suggesting that SWAP-70 with this mutation can transform the cells .\n\nERK1/2 was activated in SWAP-70-374-transformed cells .\n\nUse of MEK inhibitors revealed that the ERK1/2 pathway does not affect the cell growth of MEFs but is responsible for loss of contact inhibition .\n\nTo investigate the function of SWAP-70 further , drugs that can inhibit SWAP-70-dependent cell responses were screened .\n\nAmong various drugs , sanguinarine was found to inhibit transformation of MEFs by SWAP-70-374 .\n\nThis drug was able to inhibit SWAP-70-mediated membrane ruffling as well , suggesting that its effect was closely related to the SWAP-70 signaling pathway .\n\nThese results suggest that SWAP-70-374 can activate some signaling pathways , including the ERK1/2 pathway , to transform MEFs .", "output": "Evading growth suppressors" }, { "input": "Lewis y ( LeY ) antigen is a difucosylated oligosaccharide carried by glycoconjugates on the cell surface .\n\nOverexpression of LeY is frequently observed in epithelial-derived cancers and has been correlated to the pathological staging and prognosis .\n\nHowever , the effects of LeY on ovarian cancer are not yet clear .\n\nPreviously , we transfected the ovarian cancer cell line RMG-I with the \u03b11,2-fucosyltransferase gene to obtain stable transfectants , RMG-I-H , that highly express LeY .\n\nIn the present study , we examined the proliferation , tumorigenesis , adhesion and invasion of the cell lines with treatment of LeY monoclonal antibody ( mAb ) .\n\nAdditionally , we examined the expression of TGF-\u03b21 , VEGF and b-FGF in xenograft tumors .\n\nThe results showed that the proliferation and adhesion in vitro were significantly inhibited by treatment of RMG-I-H cells with LeY mAb .\n\nWhen subcutaneously inoculated in nude mice , RMG-I-H cells produced large tumors , while mock-transfected cells RMG-I-C and the parental cells RMG-I produced small tumors .\n\nMoreover , the tumor formation by RMG-I-H cells was inhibited by preincubating the cells with LeY mAb .\n\nNotably , the expression of TGF-\u03b21 , VEGF and b-FGF all increased in RMG-I-H cells .\n\nIn conclusion , LeY plays an important role in promoting cell proliferation , tumorigenecity and adhesion , and these effects may be related to increased levels of growth factors .\n\nThe LeY antibody shows potential application in the treatment of LeY-positive tumors .", "output": "Sustaining proliferative signaling" }, { "input": "Maintenance of genomic integrity is essential for adult tissue homeostasis and defects in the DNA-damage response ( DDR ) machinery are linked to numerous pathologies including cancer .\n\nHere , we present evidence that the DDR exerts tumor suppressor activity in gliomas .\n\nWe show that genes encoding components of the DDR pathway are frequently altered in human gliomas and that loss of elements of the ATM/Chk2/p53 cascade accelerates tumor formation in a glioma mouse model .\n\nWe demonstrate that Chk2 is required for glioma response to ionizing radiation in vivo and is necessary for DNA-damage checkpoints in the neuronal stem cell compartment .\n\nFinally , we observed that the DDR is constitutively activated in a subset of human GBMs , and such activation correlates with regions of hypoxia .", "output": "Evading growth suppressors" }, { "input": "Melanoma progression is associated with the expression of different growth factors , cytokines , and chemokines .\n\nBecause TGF\u03b21 is a pleiotropic cytokine involved not only in physiologic processes but also in cancer development , we analyzed in A375 human melanoma cells , the effect of TGF\u03b21 on monocyte chemoattractant protein-1 ( MCP-1 ) and interleukin-10 ( IL-10 ) expression , two known factors responsible for melanoma progression .\n\nTGF\u03b21 increased the expression of MCP-1 and IL-10 in A375 cells , an effect mediated by the cross-talk between Smad , PI3K ( phosphoinositide 3-kinase)/AKT , and BRAF-MAPK ( mitogen activated protein kinase ) signaling pathways .\n\nSupernatants from TGF\u03b21-treated A375 cells enhanced MCP-1-dependent migration of monocytes , which , in turn , expressed high levels of TGF,\u03b21 , bFGF , and VEGF mRNA .\n\nMoreover , these supernatants also inhibited functional properties of dendritic cells through IL-10-dependent mechanisms .\n\nWhen using in vitro , the TGF\u03b21-blocking peptide P144 , TGF\u03b21-dependent Smad3 phosphorylation , and expression of MCP-1 and IL-10 were inhibited .\n\nIn vivo , treatment of A375 tumor-bearing athymic mice with P144 significantly reduced tumor growth , associated with a lower macrophage infiltrate and decreased intratumor MCP-1 and VEGF levels , as well as angiogenesis .\n\nFinally , in C57BL/6 mice with B16-OVA melanoma tumors , when administered with immunotherapy , P144 decreased tumor growth and intratumor IL-10 levels , linked to enhanced activation of dendritic cells and natural killer cells , as well as anti-OVA T-cell responses .\n\nThese results show new effects of TGF\u03b21 on melanoma cells , which promote tumor progression and immunosuppression , strongly reinforcing the relevance of this cytokine as a molecular target in melanoma .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Radiotherapy is a useful component of treatment strategies for esophageal cancer .\n\nThe role of autophagy in response to ionizing radiation was investigated in human esophageal squamous carcinoma cells .\n\nCell viability and clonogenic survival assay were used to evaluate the radiosensitivity of autophagy inhibitor ( 3-MA ) on esophageal squamous carcinoma cells .\n\nThe percentage of apoptotic cells and cell cycle analysis were assessed by flow cytometry ; DAPI staining was used to detect apoptotic cells .\n\nThe expression of beclin-1 and LC3 was measured using a Western blot .\n\nThe ultrastructural analysis was under the electron microscope. 6\u2003Gy irradiation induced a massive accumulation of autophagosomes accompanied by strong upregulation of beclin-1 and LC3-II expression in TE-1 cells .\n\nCompared with radiation alone , 3-MA combined with radiation significantly decreased cell viability , as well as autophagic ratio , beclin-1 , and LC3-II protein level .\n\nInhibition of autophagy increased radiation-induced apoptosis and the percentage of G2/M-phase cells .\n\nBlockade of autophagy with 3-MA enhanced cytotoxicity of radiotherapy in human esophageal squamous carcinoma cells .\n\nIt suggests that inhibition of autophagy could be used as adjuvant therapy to treat esophageal squamous cell carcinoma .", "output": "Resisting cell death" }, { "input": "Currently there is no good hepatocyte model for studying growth hormone ( GH ) function that reflects its normal physiological roles .\n\nHere we report the establishment of a functional hepatocyte cell line , SDRL-1 , from the liver of young male spontaneous dwarf rats ( SDR ) , with isolated GH deficiency .\n\nThis line has been maintained in Dulbecco's Modified Eagle Medium ( DMEM)/F12 medium supplemented with 10% fetal bovine serum ( FBS ) with retention of a near diploid karyotype for extended periods of time .\n\nWhen grown as a monolayer sheet , it displayed a pavement-like appearance and contact inhibition .\n\nThese cells have a poorly developed rough endoplasmic reticulum ( r-ER ) , few mitochondria and glycogen granules , and produce a small amount of albumin and \u03b1-fetoprotein , that is enhanced when grown on a collagen gel sponge .\n\nHuman recombinant GH stimulated JAK2 and STAT5b tyrosine phosphorylation and IGF-I production in a concentration-dependent manner .\n\nWhen the cells were cultured with GH-supplemented medium , the number of mitochondria and glycogen granules increased together with the r-ER and Golgi apparatus .\n\nA number of microvilli were observed on the surface of the cultured cells , further suggesting that this cell line is composed of normally functioning hepatocytes .\n\nIn summary , we established a novel hepatocyte cell line ( SDRL-1 ) , that appears to display normal function , which we propose can serve as a good in vitro model for studying GH-target organ interactions .", "output": "Evading growth suppressors" }, { "input": "The polycomb group family protein BMI-1 is overexpressed by and functions as an oncogene in many different human cancers .\n\nWe have previously shown that BMI-1 promotes the tumorigenicity of Ewing sarcoma family tumors ( ESFTs ) and that this is mediated independently of CDKN2A repression .\n\nIn this study , we have discovered that high levels of BMI-1 confer resistance to contact inhibition in ESFT cells .\n\nUsing stable retroviral transduction , we evaluated the consequences of BMI-1 knockdown on the growth of CDKN2A wild-type and mutant ESFT cells in subconfluent and confluent conditions .\n\nAlthough knockdown of BMI-1 had no effect on proliferation in low-density cultures , at high cell densities it resulted in cell cycle arrest and death .\n\nThe normal cell contact inhibition response is mediated , in large part , by the recently described Hippo pathway which functions to inhibit cell proliferation and promote cell death by inactivating the Yes-Associated Protein ( YAP ) .\n\nSignificantly , we found that YAP levels , activity and expression did not diminish in confluent ESFT cells that expressed high levels of BMI-1 .\n\nIn contrast , YAP expression and nuclear localization were reduced in confluent BMI-1 knockdown cells suggesting that silencing of BMI-1 restored contact inhibition by restoring normal activation of the Hippo-YAP growth-suppressor pathway .\n\nImportantly , knockdown of YAP in ESFT cells resulted in profound inhibition of cell proliferation and anchorage-independent colony formation suggesting that stabilization and continued expression of YAP is critical for ESFT growth and tumorigenicity .\n\nTogether , these studies reveal a previously unrecognized link between BMI-1 , contact inhibition and the Hippo-YAP pathway and suggest that resistance to contact inhibition in BMI-1 overexpressing cancer cells may be in part a result of Hippo inhibition and aberrant stabilization of YAP .", "output": "Evading growth suppressors" }, { "input": "Extensive biochemical and structural analyses have been performed on the putative DNA repair proteins of hyperthermophilic archaea , in contrast to the few genetic analyses of the genes encoding these proteins .\n\nAccordingly , little is known about the repair pathways used by archaeal cells at high temperature .\n\nHere , we attempted to disrupt the genes encoding the potential repair proteins in the genome of the hyperthermophilic archaeon Thermococcus kodakaraensis .\n\nWe succeeded in isolating null mutants of the hjc , hef , hjm , xpb , and xpd genes , but not the radA , rad50 , mre11 , herA , nurA , and xpg/fen1 genes .\n\nPhenotypic analyses of the gene-disrupted strains showed that the xpb and xpd null mutants are only slightly sensitive to ultraviolet ( UV ) irradiation , methyl methanesulfonate ( MMS ) and mitomycin C ( MMC ) , as compared with the wild-type strain .\n\nThe hjm null mutant showed sensitivity specifically to mitomycin C. On the other hand , the null mutants of the hjc gene lacked increasing sensitivity to any type of DNA damage .\n\nThe Hef protein is particularly important for maintaining genome homeostasis , by functioning in the repair of a wide variety of DNA damage in T. kodakaraensis cells .\n\nDeletion of the entire hef gene or of the segments encoding either its nuclease or helicase domain produced similar phenotypes .\n\nThe high sensitivity of the \u0394hef mutants to MMC suggests that Hef performs a critical function in the repair process of DNA interstrand cross-links .\n\nThese damage-sensitivity profiles suggest that the archaeal DNA repair system has processes depending on repair-related proteins different from those of eukaryotic and bacterial DNA repair systems using homologous repair proteins analyzed here .", "output": "Genomic instability and mutation" }, { "input": "Carbon nanotubes have a wide range of applications in various industries and their use is likely to rise in the future .\n\nCurrently , a major concern is that with the increasing use and production of these materials , there may be increased health risks to exposed workers .\n\nLong ( > 15 microm ) straight nanotubes may undergo frustrated phagocytosis which is likely to result in reduced clearance .\n\nWe examine here the effects of multiwalled carbon nanotubes of different sizes on monocytic THP-1 cells , with regard to their ability to stimulate increased expression of the HO-1 and GST genes and their ability to produce nuclear translocation of the transcription factor , Nrf2 , as well as the release of several pro-inflammatory cytokines and mediators of inflammation .\n\nOur results suggest that long ( 50 microm ) carbon nanotubes ( 62.5 microg/ml for 4 hours ) produce increased nuclear translocation of Nrf2 and increased HO-1 gene expression compared with shorter entangled nanotubes .\n\nThere was no increased gene expression for GST .\n\nThe long nanotubes ( NT1 ) caused increased release of the proinflammatory cytokine IL-1beta , an effect which was diminished by the antioxidant trolox , suggesting a role of oxidative stress in the upregulation of this cytokine .\n\nTentatively , our study suggests that long carbon nanotubes may exert their effect in THP-1 cells in part via an oxidative stress mechanism .", "output": "Tumor promoting inflammation" }, { "input": "RKIP-1 is a metastasis suppressor that is frequently downregulated in aggressive cancers .\n\nHowever , the consequences of RKIP loss in primary or immortalized cells have not yet been explored .\n\nUsing HEK-293 RKIP depleted ( termed HEK-499 ) and Flp-In T-Rex-293 RKIP inducible cell lines combined with whole transcriptome analysis , we show that RKIP-1 silencing accelerates DNA synthesis and G1/S transition entry by inducing the expression of cdc6 , MCM 2 , 4 , 6 , 7 , cdc45L , cyclin D2 , cyclin E2 , cyclin D1 , SKP2 and the downregulation of p21(cip1) .\n\nMoreover , RKIP depletion accelerates the time from nuclear envelop breakdown ( NEB ) to anaphase markedly , while the upregulation of RKIP shortened the NEB to anaphase time .\n\nWe show that RKIP depletion induces the expression of NEK6 , a molecule known to enhance G2/M transition , and down-regulates G2/M checkpoint molecules like Aurora B , cyclin G1 and sertuin that slow the G2/M transition time .\n\nThese subtle changes in the kinetics of the cell cycle culminate in a higher proliferation rate of HEK-499 compared to control cells .\n\nFinally , we show that RKIP depletion enhances cellular motility by inducing the expression/stabilization of \u03b2-catenin , vimentin , MET and PAK1 .\n\nOverall , our data suggest that modulation of the cell cycle checkpoints and motility by RKIP may be fundamental to its metastasis suppressive function in cancer and that RKIP role in a cell is more intricate and diverse than previously thought .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "DNA damage checkpoints in the cell cycle may be important barriers against cancer progression in human cells .\n\nFanconi anemia ( FA ) is an inherited DNA instability disorder that is associated with bone marrow failure and a strong predisposition to cancer .\n\nAlthough FA cells experience constitutive chromosomal breaks , cell cycle arrest at the G2 DNA damage checkpoint , and an excess of cell death , some patients do become clinically stable , and the mechanisms underlying this , other than spontaneous reversion of the disease-causing mutation , are not well understood .\n\nHere we have defined a clonal phenotype , termed attenuation , in which FA patients acquire an abrogation of the G2 checkpoint arrest .\n\nAttenuated cells expressed lower levels of CHK1 ( also known as CHEK1 ) and p53 .\n\nThe attenuation could be recapitulated by modulating the ATR/CHK1 pathway , and CHK1 inhibition protected FA cells from cell death .\n\nFA patients who expressed the attenuated phenotype had mild bone marrow deficiency and reached adulthood , but several of them eventually developed myelodysplasia or leukemia .\n\nBetter understanding of attenuation might help predict a patient's clinical course and guide choice of treatment .\n\nOur results also highlight the importance of evaluating the cellular DNA damage checkpoint and repair pathways in cancer therapies in general .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "Bone is the second most common metastatic site in patients with renal cell carcinoma presenting with metastases ( mRCC ) at diagnosis .\n\nComplications of metastatic bone disease , including bone pain , fractures , spinal cord compression , and hypercalcaemia , are the primary cause of decline in the quality of life of patients with mRCC .\n\nCurrently , treatment for mRCC bone metastases is generally palliative .\n\nBisphosphonates are also used ; however , the efficacy of bisphosphonates in conjunction with targeted agents is currently unknown .\n\nAs growth factors play a critical role in the development of bone metastases , there is a biological rationale for the use of targeted agents to treat them .\n\nWe report here the case of two patients with mRCC with surgically unresectable sacral bone metastases treated with sunitinib , who are still alive with long-term stabilization of metastases of 48 and 31 months .\n\nResults suggest targeted agents such as sunitinib may be an effective treatment for bone metastases .", "output": "Activating invasion and metastasis" }, { "input": "Cyclin-dependent kinases are most extensively studied targets for cancer chemotherapy since the tumor cells exhibit false checkpoints and can proliferate even if the genome is compromised .\n\nCyclin-dependent kinases ensure the tight regulation of the cell cycle execution by mediating phosphorylation of cellular proteins .\n\nDeregulation of the cyclin-dependent kinase 2 activity by cellular and external factors leads to many diseases like cancers .\n\nDifferent methods like radiolabeled , fluorescence and luminescence are available for screening of library of compounds against kinases .\n\nHowever , bioluminescent methods offer several advantages like low background and no effect of fluorescent compound interference .\n\nPresent study is focused on development , optimization and validation of cyclin-dependent kinase 2 assay which is suitable for identification potent and selective , ATP competitive and non-competitive inhibitors of cyclin-dependent kinase 2 .\n\nThe aim of present investigation was to optimize the assay for cyclin-dependent kinase 2/cylin A and cyclin-dependent kinase 2/cyclin E with use of bioluminescence based biochemical reaction .\n\nBoth cyclin-dependent kinase 2 which are cyclin-dependent kinase 2/cyclin A and cyclin-dependent kinase 2/cyclin E complexes , have different affinity for ATP .\n\nTherefore , both isoform analogs of cyclin-dependent kinase 2 were optimized separately .\n\nOptimum cyclin-dependent kinase 2/cyclin A and cyclin-dependent kinase 2/cyclin E concentration were found to be 250 ng/well and 200 ng/well , respectively .\n\nOptimum substrate ( histone H1 ) concentration was found to be 2.5 mg/ml for both cyclin-dependent kinase 2 analogs .\n\nOptimum reaction time was found to be 20 min for both cyclin-dependent kinase 2/cyclin complexes .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "BACKGROUND Induction of osteolytic bone lesions in multiple myeloma is caused by an uncoupling of osteoclastic bone resorption and osteoblastic bone formation .\n\nCurrent management of myeloma bone disease is limited to the use of antiresorptive agents such as bisphosphonates .\n\nMETHODOLOGY/PRINCIPAL FINDINGS We tested the effects of daily administered parathyroid hormone ( PTH ) on bone disease and myeloma growth , and we investigated molecular mechanisms by analyzing gene expression profiles of unique myeloma cell lines and primary myeloma cells engrafted in SCID-rab and SCID-hu mouse models .\n\nPTH resulted in increased bone mineral density of myelomatous bones and reduced tumor burden , which reflected the dependence of primary myeloma cells on the bone marrow microenvironment .\n\nTreatment with PTH also increased bone mineral density of uninvolved murine bones in myelomatous hosts and bone mineral density of implanted human bones in nonmyelomatous hosts .\n\nIn myelomatous bone , PTH markedly increased the number of osteoblasts and bone-formation parameters , and the number of osteoclasts was unaffected or moderately reduced .\n\nPretreatment with PTH before injecting myeloma cells increased bone mineral density of the implanted bone and delayed tumor progression .\n\nHuman global gene expression profiling of myelomatous bones from SCID-hu mice treated with PTH or saline revealed activation of multiple distinct pathways involved in bone formation and coupling ; involvement of Wnt signaling was prominent .\n\nTreatment with PTH also downregulated markers typically expressed by osteoclasts and myeloma cells , and altered expression of genes that control oxidative stress and inflammation .\n\nPTH receptors were not expressed by myeloma cells , and PTH had no effect on myeloma cell growth in vitro .\n\nCONCLUSIONS/SIGNIFICANCE We conclude that PTH-induced bone formation in myelomatous bones is mediated by activation of multiple signaling pathways involved in osteoblastogenesis and attenuated bone resorption and myeloma growth ; mechanisms involve increased osteoblast production of anti-myeloma factors and minimized myeloma induction of inflammatory conditions .", "output": "Tumor promoting inflammation" }, { "input": "Our purpose in this randomized , double blind , placebo controlled study was to find out the possible effect of a polyphenolic pine bark extract , Pycnogenol\u00ae ( Pyc ) on the level of 8-oxo-7,8-dihydroguanine ( 8-oxoG ) as representative of oxidative damage to DNA and on the DNA repair ability of elderly people .\n\nAccording to our results , three months of Pyc administration had no effect on the level of oxidative damage to DNA or on repair ability , but we found a relationship between the level of 8-oxoG and repair ability of DNA in this group .\n\nTo conclude , even if the positive effect of Pyc was not confirmed in the case of elderly people it is important to highlight the necessity of further investigations about the mechanisms of Pyc acting on different age groups .", "output": "Genomic instability and mutation" }, { "input": "Dentin matrix protein 1 ( DMP1 ) is a member of the small integrin-binding ligand N-linked glycoprotein ( SIBLING ) family , a group of proteins initially described as mineralized extracellular matrices components .\n\nMore recently , SIBLINGs have been implicated in several key steps of cancer progression , including angiogenesis .\n\nAlthough proangiogenic activities have been demonstrated for 2 SIBLINGs , the role of DMP1 in angiogenesis has not yet been addressed .\n\nWe demonstrate that this extracellular matrix protein induced the expression of vascular endothelial cadherin ( VE-cadherin ) , a key regulator of intercellular junctions and contact inhibition of growth of endothelial cells that is also known to modulate vascular endothelial growth factor receptor 2 ( VEGFR-2 ) activity , the major high-affinity receptor for VEGF .\n\nDMP1 induced VE-cadherin and p27(Kip1) expression followed by cell-cycle arrest in human umbilical vein endothelial cells ( HUVECs ) in a CD44-dependent manner .\n\nVEGF-induced proliferation , migration , and tubulogenesis responses were specifically blocked on DMP1 pretreatment of HUVECs .\n\nIndeed , after VE-cadherin induction , DMP1 inhibited VEGFR-2 phosphorylation and Src-mediated signaling .\n\nHowever , DMP1 did not interfere with basic fibroblast growth factor-induced angiogenesis .\n\nIn vivo , DMP1 significantly reduced laser-induced choroidal neovascularization lesions and tumor-associated angiogenesis .\n\nThese data enable us to put DMP1 on the angiogenic chessboard for the first time and to identify this protein as a new specific inhibitor of VEGF-induced angiogenesis .", "output": "Inducing angiogenesis, Evading growth suppressors" }, { "input": "Despite the deployment of multimodal therapies involving neurosurgical resection , radio- and polychemotherapy , the prognosis for glioblastoma patients remains poor .\n\nThese tumors are pathologically characterized by their associated angiogenesis and diffuse brain invasion , processes that are probably closely linked to the unfavorable prognosis of this disease .\n\nAccordingly , pharmacological inhibition of glioblastoma invasion and approaches that impede angiogenesis are considered to be promising therapeutic strategies to combat these tumors .\n\nNevertheless , the anti-angiogenic therapies for glioblastoma currently available are transient and palliative at best .\n\nBlocking the effects of fibroblast growth factor ( FGF ) may represent a novel mean of inhibiting the angiogenesis associated with glioblastoma , as it mediates the angiogenesis induced by other factors and it is an angiogenic factor by itself .\n\nIn addition , the survival of glioma cells and their resistance to chemotherapeutic agents are highly FGF-dependent .\n\nWe show here that a recently described inhibitor of FGF , 2,5-dihydroxyphenyl-sulfonate ( 2,5DHPS , dobesilate ) , stimulates the apoptosis of tumor cells , inhibits glioblastoma invasion and suppresses its associated angiogenesis .\n\nMoreover , this agent augments the efficiency of chemotherapeutic agents in a rat model of orthotopic brain tumor .\n\nThese results suggest that 2,5DHPS treatment may represent a promising therapy for malignant glioma .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Modeling the behavior of mammalian arachnoid cells is critical to understand hydrocephalus and other brain disorders involving abnormal flow of cerebrospinal fluid , yet relatively little is known about the physiology of arachnoid cells due to lack of a robust three-dimensional model system .\n\nExplanted primary cultures have been the only option to study transport across arachnoid cell membranes , but practical limitations of primary culture include slow growth , early senescence , and poor reproducibility .\n\nThe purpose of this study was to create immortalized rat arachnoid cell lines to permit in vitro study of arachnoid granulations and properties of cerebrospinal fluid ( CSF ) flow .\n\nWe established and partially characterized two immortalized cell lines generated from primary rat arachnoid cells , using retroviral gene transfer of SV40 large T antigen ( SV40 LTAg ) either with or without human telomerase ( hTERT ) .\n\nThe established cell lines stably express either SV40 LTAg alone , or SV40 LTAg and hTERT , and demonstrate high proliferative rate , contact inhibition at confluence , and stable expression of protein markers characteristic of native arachnoid cells over more than 160 passages .", "output": "Enabling replicative immortality, Evading growth suppressors" }, { "input": "Melanoma cells express and interact with laminins ( LMs ) and other basement membrane components during invasion and metastasis .\n\nIn the present study we have investigated the production and migration-promoting activity of laminin isoforms in melanoma .\n\nImmunohistochemistry of melanoma specimens and immunoprecipitation/western blotting of melanoma cell lines indicated expression of laminin-111/121 , laminin-211 , laminin-411/421 , and laminin-511/521 .\n\nLaminin-332 was not detected .\n\nIn functional assays , laminin-111 , laminin-332 , and laminin-511 , but not laminin-211 and laminin-411 , strongly promoted haptotactic cell migration either constitutively or following stimulation with insulin-like growth factors .\n\nBoth placenta and recombinant laminin-511 preparations were highly active , and the isolated recombinant IVa domain of LM\u03b15 also promoted cell migration .\n\nFunction-blocking antibodies in cell migration assays revealed \u03b16\u03b21 integrin as the major receptor for laminin-111 , and both \u03b13\u03b21 and \u03b16\u03b21 integrins for laminin-332 and laminin-511 .\n\nIn contrast , isolated LM\u03b15 IVa domain-promoted melanoma cell migration was largely mediated via \u03b1V\u03b23 integrin and inhibited by RGD peptides .\n\nGiven the ubiquitous expression of \u03b15 laminins in melanoma cells and in melanoma-target tissues/anatomical structures , as well as the strong migration-promoting activity of these laminin isoforms , the \u03b15 laminins emerge as putative primary extracellular matrix mediators of melanoma invasion and metastasis via \u03b13\u03b21 and other integrin receptors .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Isolinderanolide B ( IOB ) , a butanolide extracted from the stems of Cinnamomum subavenium , was investigated for its antiproliferative activity in T24 human bladder cancer cells .\n\nTo identity the anticancer mechanism of IOB , its effect on apoptosis , cell cycle distribution , and levels of p53 , p21 Waf1/Cip1 , Fas/APO-1 receptor , and Fas ligand was assayed .\n\nEnzyme-linked immunosorbent assay showed that the G0/G1 phase arrest is because of increase in the expression of p21 Waf1/Cip1 .\n\nAn enhancement in Fas/APO-1 and membrane-bound Fas ligand ( mFasL ) might be responsible for the apoptotic effect induced by IOB .\n\nThis study reports the novel finding that the induction of p21 Waf1/Cip1 and activity of the Fas/mFas ligand apoptotic system may participate in the antiproliferative activity of IOB in T24 cells .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "Pegylated Interferon-\u03b12b ( pIFN-\u03b1 ) is an integral part of the drug regimen currently employed against melanoma .\n\nInterferon Regulatory Factor-1 ( IRF-1 ) plays an important role in the transcriptional regulation of the IFN response , cell cycle and apoptosis .\n\nWe have studied pIFN-\u03b1 induced responses when combined with the chemotherapy agent , vinblastine in tumor and endothelial cell lines and the connection to IRF-1 signaling .\n\nLevels of IRF-1/IRF-2 protein expression were found to be decreased in tumor versus normal tissues. pIFN-\u03b1 induced IRF-1 signaling in human melanoma ( M14 ) and endothelial ( EA.hy926 ) cells and enhanced cell death when combined with vinblastine .\n\nUpon combined IFN-\u03b1 and vinblastine treatment , p21 expression , PARP cleavage and activated Bak levels were increased in M14 cells .\n\nAn increase in p21 and cyclin D1 expression occurred in EA.hy926 cells after 6 h of treatment with pIFN-\u03b1 which dissipated by 24 h .\n\nThis biphasic response , characteristic of cellular senescence , was more pronounced upon combined treatment .\n\nExposure of the EA.hy926 cells to pIFN-\u03b1 was associated with an enlarged , multinucleated , \u03b2-galactosidase-positive senescent phenotype .\n\nThe overall therapeutic mechanism of IFN-\u03b1 combined with chemotherapy may be due to both direct tumor cell death via IRF-1 signaling and by premature senescence of endothelial cells and subsequent effects on angiogenesis in the tumor microenvironment .", "output": "Inducing angiogenesis, Enabling replicative immortality, Evading growth suppressors, Resisting cell death" }, { "input": "DNA repair enzymes play an important role in the development of various kinds of cancer .\n\nWe here analyzed associations of XPD Lys751Gln , APEX1 Asp148Glu , XRCC1 Arg399Gln , and XRCC3 Thr241Met gene polymorphisms in DNA repair pathways in relation to the risk of lung cancer using PCR-RFLP .\n\nThe study involved 104 lung cancer patients and 120 non-cancer controls divided into non-smokers and smokers .\n\nWe found a statistically significant interaction between APEX1 Asp148Glu and the risk for lung cancer ( adjusted OR 2.78 , 95% CI 1.58-4.90 , p=0.0004 ) , of both adenocarcinoma ( adjusted OR 2.24 , 95%CI 1.18-4.25 , p=0.014 ) and squamous cell carcinoma ( adjusted OR 4.75 , 95%CI 1.79-12.6 , p=0.002 ) types .\n\nXRCC1 Arg399Gln showed a borderline significant association with adenocarcinoma ( adjusted OR 1.89 , 95%CI 1.00-3.57 , p=0.051 ) .\n\nThe combined effect of smoking and presence of the APEX1 Asp148Glu demonstrated a significant association with risk of lung cancer ( adjusted OR 3.61 , 95% CI 1.74-7.50 , p=0.001 ) .\n\nThe XPD Lys751Gln and XRCC3 Thr241Met genotypes displayed no statistically significant risk .\n\nOur findings suggest that the APEX1 Asp148Glu is associated with increased risk for primary lung cancer in Japanese individuals partaking in smoking .", "output": "Genomic instability and mutation" }, { "input": "Chemoprevention is one feasible approach to decreasing morbidity and mortality of non-small cell lung cancer ( NSCLC ) .\n\nThe present study aimed to explore the mechanisms of chemoprevention of NSCLC by Prunella vulgaris L .\n\n( PV ) using a PV extract of 60% ethanol ( P-60 ) .\n\nIn an A/J mouse model benzo[a]pyrene induction of lung tumors was significantly reduced difference by P-60 group .\n\nIn addition , P-60 was found to have the ability to regulate cell cycle and induce apoptosis in SPC-A-1 cells .\n\nTherefore , we propose that P-60 has potential as a lung cancer chemopreventive agent .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND Nucleolin is one of the major proteins of the nucleolus , but it is also expressed on the cell surface where is serves as a binding protein for variety of ligands implicated in tumorigenesis and angiogenesis .\n\nEmerging evidence suggests that the cell-surface expressed nucleolin is a strategic target for an effective and nontoxic cancer therapy .\n\nMETHODOLOGY/PRINCIPAL FINDINGS By monitoring the expression of nucleolin mRNA , and by measuring the level of nucleolin protein recovered from the surface and nucleus of cells , here we show that the presence of nucleolin at the cell surface is dependent on the constant induction of nucleolin mRNA .\n\nIndeed , inhibitors of RNA transcription or translation block expression of surface nucleolin while no apparent effect is observed on the level of nucleolin in the nucleus .\n\nThe estimated half-life of surface nucleolin is less than one hour , whereas that of nuclear nucleolin is more than 8 hours .\n\nNucleolin mRNA induction is reduced markedly in normal fibroblasts that reach confluence , while it occurs continuously even in post-confluent epithelial tumor cells consistent with their capacity to proliferate without contact inhibition .\n\nInterestingly , cold and heat shock induce nucleolin mRNA concomitantly to enhanced mRNA expression of the heat shock protein 70 , thus suggesting that surface nucleolin induction also occurs in response to an environmental insult .\n\nAt the cell surface , one of the main functions of nucleolin is to shuttle specific extracellular ligands by an active transport mechanism , which we show here to be calcium dependent .\n\nCONCLUSION/SIGNIFICANCE Our results demonstrate that the expression of surface nucleolin is an early metabolic event coupled with tumor cell proliferation and stress response .\n\nThe fact that surface nucleolin is constantly and abundantly expressed on the surface of tumor cells , makes them a preferential target for the inhibitory action of anticancer agents that target surface nucleolin .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "BACKGROUND NF\u03baB signaling is of paramount importance in the regulation of apoptosis , proliferation , and inflammatory responses during human development and homeostasis , as well as in many human cancers .\n\nReceptor Tyrosine Kinases ( RTKs ) , including the Fibroblast Growth Factor Receptors ( FGFRs ) are also important in development and disease .\n\nHowever , a direct relationship between growth factor signaling pathways and NF\u03baB activation has not been previously described , although FGFs have been known to antagonize TNF\u03b1-induced apoptosis .\n\nMETHODOLOGY/PRINCIPAL FINDINGS Here , we demonstrate an interaction between FGFR4 and IKK\u03b2 ( Inhibitor of NF\u03baB Kinase \u03b2 subunit ) , an essential component in the NF\u03baB pathway .\n\nThis novel interaction was identified utilizing a yeast two-hybrid screen [ 1 ] and confirmed by coimmunoprecipitation and mass spectrometry analysis .\n\nWe demonstrate tyrosine phosphorylation of IKK\u03b2 in the presence of activated FGFR4 , but not kinase-dead FGFR4 .\n\nFollowing stimulation by TNF\u03b1 ( Tumor Necrosis Factor \u03b1 ) to activate NF\u03baB pathways , FGFR4 activation results in significant inhibition of NF\u03baB signaling as measured by decreased nuclear NF\u03baB localization , by reduced NF\u03baB transcriptional activation in electophoretic mobility shift assays , and by inhibition of IKK\u03b2 kinase activity towards the substrate GST-I\u03baB\u03b1 in in vitro assays .\n\nFGF19 stimulation of endogenous FGFR4 in TNF\u03b1-treated DU145 prostate cancer cells also leads to a decrease in IKK\u03b2 activity , concomitant reduction in NF\u03baB nuclear localization , and reduced apoptosis .\n\nMicroarray analysis demonstrates that FGF19 + TNF\u03b1 treatment of DU145 cells , in comparison with TNF\u03b1 alone , favors proliferative genes while downregulating genes involved in apoptotic responses and NF\u03baB signaling .\n\nCONCLUSIONS/SIGNIFICANCE These results identify a compelling link between FGFR4 signaling and the NF\u03baB pathway , and reveal that FGFR4 activation leads to a negative effect on NF\u03baB signaling including an inhibitory effect on proapoptotic signaling .\n\nWe anticipate that this interaction between an RTK and a component of NF\u03baB signaling will not be limited to FGFR4 alone .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND/AIMS Mesenchymal stem cells ( MSCs ) have been implicated in antitumor therapy for hematopoietic and non-hematopoietic tumors .\n\nCell-contact and soluble factors are demonstrated to play a role in the growth inhibition of tumor cells mediated by MSCs in vitro , while there is little clue about signaling pathways involved in the process .\n\nP38 MAPK has been implicated as a suppressor of cell proliferation and tumorigenesis .\n\nWe here investigate whether p38 MAPK is involved in MSC-induced growth inhibition of leukemic tumor cells .\n\nMethods : We characterized the effect of human umbilical cord mesenchymal stem cells ( UC-MSCs ) on proliferation , cell cycle and phosphorylation pattern of p38 MAPK in HL60 and K562 cells .\n\nSB203580 , a specific inhibitor of p38 MAPK , or p38 MAPK-small interfering RNA ( siRNA ) , were used to identify the role of p38 in growth suppression by UC-MSCs .\n\nWe also investigated the expression of cell cycle regulators .\n\nRESULTS Treatment with UC-MSCs led to potent proliferation-inhibition of HL60 and K562 cells without inducing apoptosis .\n\nGrowth inhibition by UC-MSCs was due to G0/G1 arrest .\n\nUC-MSCs increased phosphorylation of p38 MAPK in HL60 and K562 cells .\n\nPharmacological inhibition or genetic silencing ( through siRNA ) of p38 MAPK partially abrogated the proliferation-suppression and cell cycle arrest caused by UC-MSCs .\n\nUC-MSCs also modulated the expression of cell cycle regulatory proteins in HL60 and K562 cells while SB203580 reversed the effect .\n\nCONCLUSION Taken together , our findings indicate that p38 MAPK is critical for the growth inhibitory effect of UC-MSCs on leukemic tumor cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "By using the rat azoxymethane ( AOM)-induced colon carcinogenesis model , which mirrors many clinical features of human colorectal cancer , we examined whether genetic changes occurring early in colonic mucosa are predictive of treatment efficacy .\n\nIn the present study the administration of the chemopreventive agent lupulone over the course of 7 weeks postinitiation reduced the number of preneoplastic lesions in the colonic mucosa by 50% .\n\nAt the molecular level we observed the downregulation of genes involved in the inflammatory response , including IL-1\u03b2 and TNF-\u03b1 , and of matrix metalloproteinase-7 gene and protein expression .\n\nWe also observed a substantial upregulation of components of the innate immune system , \u03b1-defensin-5 and lipocalin 2 .\n\nLupulone induced the expression of apoptosis-related genes and caused a reversal of the B-cell lymphoma/leukemia 2 ( Bcl-2 ; antiapoptotic ) to Bcl-2 associated X protein ( Bax ; proapoptotic ) transcript and protein ratios ( Bcl-2/Bax > 1 in AOM controls and Bcl-2/Bax < 1 in lupulone-treated AOM rats ) .\n\nHere , we identify several target genes that could be considered early biomarkers of colon carcinogenesis and indicative of drug efficacy .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "UNLABELLED Oxygen free radicals and their reactive derivatives participate in formation of chronic inflammation states , which facilitate development of gastrointestinal tract tumors .\n\nOxidative stress is one of the main causes of damage to cell membranes in result of exacerbated lipid peroxidation process .\n\nEnd products of lipid peroxidation ( aldehydes , organic peroxides ) react with important biological macromolecules such as DNA and proteins , cause changes in cell membrane structure and properties leading to loss of its integrity .\n\nIntensification of the lipid peroxidation process is a factor which may also lead to a malfunction in the antioxidant barrier , which further weakens the defense of cells against oxygen free radicals and promotes the onset and development of cancer .\n\nThe aim of the study was the determination of lipid peroxidation level in gastrointestinal tract tumors ( stomach , liver , colon , and colorectal cancer to liver metastases ) .\n\nMATERIAL AND METHODS Materials for studies were obtained from 150 patients with gastrointestinal tract tumors : 10 with stomach cancer , 30 with malignant and benign liver cancers , 60 with primary colorectal cancer , and 50 with metachronous colorectal cancer liver metastases .\n\nWe also investigated 25 patients with liver cirrhosis , which was treated as a pre-cancerous condition .\n\nIn total , 175 patients were examined .\n\nTumor specimens , and normal adjacent tissues ( 6-7 cm from the edge of the tumor ) , which served as control tissue in studies , were collected from patients ( with their consent ) during surgery .\n\nAdditionally , liver specimens were collected from patients with liver cirrhosis .\n\nLipid peroxidation level was determined spectrophotometrically as a concentration of final lipid peroxidation products , which in reaction with tiobarbituric acid ( TBA ) form colour complex ( thiobarbituric acid-reactive substances - TBARS ) .\n\nRESULTS The study showed the highest concentration of TBARS in benign , and the lowest in malignant liver tumors .\n\nOther types of gastrointestinal tumors studied , were characterized by similar levels of lipid peroxidation .\n\nTBARS concentration in these tumors was approximately 2-fold higher than in malignant liver tumors and much lower than in benign tumors .\n\nIn all cancers of the digestive tract with the exception of malignant liver tumors increased level of TBARS was found , comparing with control tissue .\n\nThe concentration of TBARS in cirrhotic liver was lower than in control .\n\nThe level of lipid peroxidation in liver cirrhosis and malignant liver tumors was similar .\n\nThere were no significant differences in TBARS concentration in the tumors of particular sections of the intestine and normal colon .\n\nThe highest concentration of TBARS was found in G1 grade of colorectal cancer .\n\nIn subsequent grades of cells differentiation ( G2 and G3 ) its concentration was lower .\n\nThe highest level of lipid peroxidation , expressed as the concentration of TBARS was found in the I stage of colorectal cancer clinical advancement .\n\nThe significantly lowest concentration of TBARS was shown for stage II ( UICC ) .\n\nCONCLUSIONS The level of lipid peroxidation in cancerous cells of gastrointestinal tract indicates increased oxidative stress .\n\nThe changes of lipid peroxidation level--a marker of oxidative stress in gastrointestinal tumors appear to be closely associated with their development stages ( liver cirrhosis/malignant liver cancer ; colorectal cancer/colorectal cancer liver metastases ) and are likely to create such conditions , in which cancerous cells may proliferate , undergo gradual dedifferentiation and malignancy , and generate metastases .", "output": "Tumor promoting inflammation" }, { "input": "Cigarette smoke ( CS ) is a rich source of radicals , predisposing the cell to oxidative stress resulting in inflammation .\n\nChronic inflammation is a recognized risk factor for carcinogenesis .\n\nCyclooxygenase-2 ( COX-2 ) is a mediator of inflammatory pathway and may , therefore , contribute to carcinogenesis .\n\nThere are several reports that suggest the association between CS and COX-2 associated risk to cancer .\n\nIn the present study , we examined the role of celecoxib ( a selective COX-2 inhibitor ) in modulating the oxidative stress caused by CS inhalation in mice .\n\nCS exposure for a period of 10 weeks caused oxidative stress in the pulmonary and hepatic tissues , as evident from the increase in lipid peroxidation levels ( LPO ) and decrease in reduced glutathione ( GSH ) levels .\n\nCelecoxib ( 125 mg/kg body weight for 8 weeks ) administration to CS inhaling mice reduced the oxidative stress by decreasing the LPO levels and enhancing the GSH levels in comparison to the CS-exposed group .\n\nCS exposure repressed the enzymatic antioxidant defense system , as evident from the decrease in catalase ( CAT ) and superoxide dismutase ( SOD ) activities .\n\nCo-adminstration of celecoxib considerably reversed the changes in the enzymatic antioxidant defense system .\n\nHistopathological studies of lungs showed that CS exposure induced alveolar wall destruction and air space enlargement .\n\nIn co-treated group , the alveolar septa were thicker than normal with apparent infiltration of inflammatory cells .\n\nIn CS-exposed group , hepatic tissue exhibited vacuolization and macrophage infiltration .\n\nCo-treatment with celecoxib restored the normal histoarchitechture in hepatic tissues of CS inhaling mice .\n\nThus , the present study demonstrated that celecoxib adminstration reduced the oxidative stress-mediated risk to carcinogenesis , due to its ability to boost the antioxidant defense system .", "output": "Tumor promoting inflammation" }, { "input": "Background : Several polymorphisms in the DNA repair gene have been extensively studied in the association with various human cancers such as breast cancer .\n\nMaterial and methods : We investigated the association of polymorphisms in the DNA repair genes XRCC1-Arg399Gln , XRCC2-Arg188His and RAD51-135G/C with the breast cancer risk .\n\nGenotypes were determined by PCR-RFLP assays in 220 patients with breast cancer and 220 age-matched healthy controls .\n\nResults : Our results demonstrated a significant positive association between the XRCC1 399Gln/Gln homozygous genotype and breast carcinoma , with an adjusted odds ratio ( OR ) of 2.08 [ 1.08-3.98 ] .\n\nThe 399Gln allele variant was also associated with type I breast cancer ( OR = 1.41 [ 0.98-2.01 ] , p = 0.034 ) .\n\nThe distributions of genotypes and alleles of the genes XRCC2 and RAD51 polymorphism were not significantly associated with the different stages of breast carcinoma ( p > 0.05 ) .\n\nConclusion : These results suggest that 399Gln allele of XRCC1 Arg399Gln may be a risk factor for breast cancer in the Polish population .", "output": "Genomic instability and mutation" }, { "input": "Thrombopoietin ( TPO ) receptor agonists represent a new approach for the treatment of thrombocytopenia , which may develop as a consequence of immune thrombocytopenia , chemotherapy treatment , chronic hepatitis C infection , or myelodysplastic syndromes .\n\nThere are concerns that use of certain growth factors can hasten disease progression in some types of hematologic malignancies and solid tumors .\n\nIn this study , expression of MPL ( TPO-R ) mRNA was examined in tumor cell lines , patient tumor samples ( renal cell carcinoma , prostatic carcinoma , soft tissue and bony/cartilage sarcoma , colon cancer , and lymphoma ) , and normal tissues using microarray analysis and qRT-PCR .\n\nMPL mRNA is expressed at very low or undetectable levels compared with erythropoietin receptor ( EPOR ) , human epidermal growth factor ( ERBB2 ; HER2 ) , and insulin-like growth factor-1 receptor ( IGF1R ) in these patient samples .\n\nThese data suggest TPO-R agonists will likely preferentially stimulate proliferation and differentiation of cells of megakaryocytic lineage , potentially demonstrating their utility for correcting thrombocytopenia in clinical settings .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Polymorphisms in DNA repair genes are associated with ability to remove DNA lesions , and therefore may contribute to an individual's susceptibility to different types of cancer .\n\nBase excision repair ( BER ) , and nucleotide excision repair ( NER ) are the main DNA repair pathways .\n\nThe present study was conducted to determine the frequency distribution of single nucleotide polymorphisms ( SNPs ) selected for genes in these two pathways i.e .\n\nOGG1 Exon 7 ( C1245G ) , XPC Intron 9 ( PAT ) , and Exon 15 ( A33512C ) in a North Indian population in comparison with global populations .\n\nMETHODS Genotyping was achieved by PCR-based analysis in 224 normal healthy , unrelated individuals of similar ethnicity .\n\nRESULTS Allelic frequencies in wild type of OGG1 Exon 7 C>G were 73% ( C ) ; XPC PAT D>I 75% ( D ) ; and XPC Exon 15 A>C 60.71.9% A. On the other hand , the variant allele frequency were 27% ( G ) in OGG1 Exon 7 C>G ; 25% ( I ) in XPC PAT ; and 28.1% ( C ) in XPC Exon 15 A>C .\n\nMajor differences from other ethnic populations were observed .\n\nCONCLUSIONS Our results suggest that frequency distribution in these DNA repair genes exhibited a distinctive pattern in our population which could be attributed to ethnic variation .\n\nThis could assist in high-risk screening of humans exposed to environmental carcinogens and cancer predisposition in different ethnic groups .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVES Epidermal growth factor ( EGF ) stimulates cell proliferation by binding to its receptor ( epidermal growth factor receptor ) , and the overexpression of this receptor is associated with poorer prognosis .\n\nThe EGF gene presents a polymorphism at position 61 ( A/G ) , associated with higher EGF production .\n\nWe examined the association between this polymorphism and cervical cancer through a case-control study .\n\nMETHODS This study used the PCR-restriction fragment length polymorphism method on a sample of 384 women with cervical lesions and 500 controls of white ethnicity .\n\nRESULTS In cases of cervical cancer , we found an increased risk of progression to advanced disease ( The International Federation of Gynecology and Obstetrics stage IIb/IV ) ( odds ratios=2.05 ; 95% confidence intervals=1.11 to 3.79 ; P=0.021 ) , and this risk was particularly evident in G carriers for younger women ( odds ratios=2.96 ; 95% confidence intervals=1.41-6.20 , P=0.003 ) .\n\nCONCLUSIONS We hypothesize the onset of an advanced disease-driven selective pressure due to the effect of oncogenic human papillomavirus types in a favorable genetic background observed in G carrier women .\n\nThese results suggest that EGF functional polymorphism may influence cervical cancer prognosis through an EGF/epidermal growth factor receptor pathway .", "output": "Sustaining proliferative signaling" }, { "input": "AIMS Angiogenesis from thyroid cancer cell plays the important roles in post-surgical persistent , recurrent , and metastatic papillary thyroid cancer ( PTC ) .\n\nThis study is to investigate the expression of angiopoietin-1 ( Ang-1 ) , angiopoietin-2 ( Ang-2 ) , Tek/Tie-2 receptor , and vascular endothelial growth factors ( VEGF ) in normal , benign thyroid tissues and different stage of PTC .\n\nWe expect angiogenetic factors are important in the presentation of local-regional neck or distant metastases in PTC .\n\nMATERIALS AND RESULTS A total of 101 tissues from the subjects underwent thyroidectomy were enrolled in the study .\n\nThere were 22 control and 79 thyroid cancer patients in different TNM stagings were collected .\n\nAng-1 illustrated highest mean immunostaining score in metastatic group .\n\nComparing with normal and benign thyroid tissues , thyroid cancer tissues illustrated significantly high expression of three angiogenetic factors and Tie-2 receptor .\n\nOf the PTC , significantly high expression of three angiogenetic factors and Tie-2 receptor were illustrated in recurrent cases .\n\nVEGF showed statistical difference in disease-free cancer mortality , and recurrent groups .\n\nCONCLUSIONS Immunochemical staining illustrated VEGF , Ang-1 , Ang-2 expression in PTC tissues related to clinical staging ; however , we need more information concerning these factors with long-term follow-up results .", "output": "Inducing angiogenesis" }, { "input": "OBJECTIVES Despite the well-defined histological types of non-small cell lung cancer ( NSCLC ) , a given stage is often associated with wide-ranging survival rates and treatment outcomes .\n\nThis disparity has led to an increased demand for the discovery and identification of new informative biomarkers .\n\nMETHODS In the current study , we screened 81 NSCLC samples using Illumina whole-genome gene expression microarrays in an effort to identify differentially expressed genes and new NSCLC biomarkers .\n\nRESULTS We identified novel genes whose expression was upregulated in NSCLC , including SPAG5 , POLH , KIF23 , and RAD54L , which are associated with mitotic spindle formation , DNA repair , chromosome segregation , and dsDNA break repair , respectively .\n\nWe also identified several novel genes whose expression was downregulated in NSCLC , including SGCG , NLRC4 , MMRN1 , and SFTPD , which are involved in extracellular matrix formation , apoptosis , blood vessel leakage , and inflammation , respectively .\n\nWe found a significant correlation between RNA degradation and survival in adenocarcinoma cases .\n\nCONCLUSIONS Even though the follow-up time was too limited to draw final conclusions , we were able to show better prediction p values in a group selection based on molecular profiles compared to histology .\n\nThe current study also uncovered new candidate biomarker genes that are likely to be involved in diverse processes associated with NSCLC development .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Hesperidin , a flavanone present in citrus fruits , has been identified as a potent anticancer agent because of its proapoptotic and antiproliferative characteristics in some tumor cells .\n\nHowever , the precise mechanisms of action are not entirely understood .\n\nAIM The main purpose of this study is to investigate the involvement of peroxisome proliferator-activated receptor-gamma ( PPAR\u03b3 ) in hesperidin's anticancer actions in human pre-B NALM-6 cells , which expresses wild-type p53 .\n\nMETHODS The effects of hesperidin on cell-cycle distribution , proliferation , and caspase-mediated apoptosis were examined in NALM-6 cells in the presence or absence of GW9662 .\n\nThe expression of peroxisome proliferator-activated receptor-gamma ( PPAR\u03b3 ) , p53 , phospho-I\u03baB , Bcl-2 , Bax , and XIAP proteins were focused on using the immunoblotting assay .\n\nThe transcriptional activities of PPAR\u03b3 and nuclear factor-kappaB ( NF-\u03baB ) were analyzed by the transcription factor assay kits .\n\nThe expression of PPAR\u03b3 and p53 was analyzed using the RT-PCR method .\n\nRESULTS Hesperidin induced the expression and transcriptional activity of PPAR\u03b3 and promoted p53 accumulation and downregulated constitutive NF-\u03baB activity in a PPAR\u03b3-dependent and PPAR\u03b3-independent manner .\n\nThe growth-inhibitory effect of hesperidin was partially reduced when the cells preincubated with PPAR\u03b3 antagonist prior to the exposure to hesperidin .\n\nCONCLUSIONS The findings of this study clearly demonstrate that hesperidin-mediated proapoptotic and antiproliferative actions are regulated via both PPAR\u03b3-dependent and PPAR\u03b3-independent pathways in NALM-6 cells .\n\nThese data provide the first evidence that hesperidin could be developed as an agent against hematopoietic malignancies .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "SCOPE In this study , we evaluated the efficacy of lycopene against the growth of prostate cancer in vivo .\n\nMETHODS AND RESULTS Athymic nude mice were implanted subcutaneously with human androgen-independent prostate carcinoma PC-3 cells .\n\nThey were supplemented with a low or a high dose of lycopene ( 4 and 16\u2009mg/kg ) and a single dose of \u03b2-carotene ( 16\u2009mg/kg ) twice a week for 7 wk .\n\nAt the end of the experiment , both lycopene and \u03b2-carotene strongly inhibited the tumor growth , as evidenced by the decrease in tumor volume and tumor weight .\n\nHigh-dosage lycopene and \u03b2-carotene significantly decreased the expression of proliferating cell nuclear antigen in tumor tissues and increased the levels of insulin-like growth factor-binding protein-3 in plasma .\n\nIn addition , high-dosage lycopene supplementation significantly decreased the vascular endothelial growth factor ( VEGF ) levels in plasma .\n\nIn contrast , \u03b2-carotene supplementation significantly increased the VEGF levels , as compared with tumor control group .\n\nCONCLUSION Lycopene and \u03b2-carotene supplementation suppressed the growth of prostate tumor cells , and the effects are likely associated with reduction of proliferation ( attenuation of proliferating cell nuclear antigen expression ) and with interference of the insulin-like growth factor 1 signaling ( increased plasma insulin-like growth factor-binding protein-3 levels ) .\n\nFurthermore , the inhibition of VEGF by lycopene suggests that the antitumor mechanisms of lycopene also involve anti-angiogenesis .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Radiotherapy for head and neck tumors often results in persistent loss of function in salivary glands .\n\nPatients suffering from impaired salivary function frequently terminate treatment prematurely because of reduced quality of life caused by malnutrition and other debilitating side-effects .\n\nIt has been previously shown in mice expressing a constitutively active form of Akt ( myr-Akt1 ) , or in mice pretreated with IGF1 , apoptosis is suppressed , which correlates with maintained salivary gland function measured by stimulated salivary flow .\n\nInduction of cell cycle arrest may be important for this protection by allowing cells time for DNA repair .\n\nWe have observed increased accumulation of cells in G2/M at acute time-points after irradiation in parotid glands of mice receiving pretreatment with IGF1 .\n\nAs p21 , a transcriptional target of the p53 family , is necessary for maintaining G2/M arrest , we analyzed the roles of p53 and p63 in modulating IGF1-stimulated p21 expression .\n\nPretreatment with IGF1 reduces binding of \u0394Np63 to the p21 promoter after irradiation , which coincides with increased p53 binding and sustained p21 transcription .\n\nOur data indicate a role for \u0394Np63 in modulating p53-dependent gene expression and influencing whether a cell death or cell cycle arrest program is initiated .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "BACKGROUND Prostate cancer ( PCa ) progression is often associated with transactivation of the androgen receptor ( AR ) by endogenous hormones/growth factors .\n\nOne such factor affecting growth , proliferation , and apoptostis ( pro-/anti- ) in various cancers is the adipokine leptin .\n\nThis research studied leptin-induced signaling and apoptosis in androgen sensitive ( LNCaP , PC3/AR ) and insensitive ( PC3 , DU145 ) PCa cell lines .\n\nMETHODS Signaling was studied by immunoblotting in cells overexpressing leptin receptors ( LRb ) , Janus kinase 2 ( JAK2 ) , and kinase negative-HER2-YFP cDNAs .\n\nApoptosis was measured by immunoblotting of apoptotic proteins and by Hoechst staining of condensed DNA .\n\nRESULTS Leptin rapidly induced activation of JAK2 , STAT3 , and MAPK ( ERK1/2 ) signaling cascades ; it may also induce HER2 transactivation via leptin-induced phospho-JAK2 .\n\nLeptin was then shown to exert clear pro-apoptotic effects , increasing levels of caspase 3 , cleavage of its substrate , poly ( ADP-ribose ) polymerase ( PARP ) to cleaved PARP(89) , levels of CK 18 , a cytoskeletal protein formed during apoptosis , and DNA condensation .\n\nKinase inhibitors indicated that leptin-induced apoptosis is probably mediated by balanced activation of JAK2/STAT3 , p38 MAPK , and PKC pathways in PCa cells .\n\nA human leptin mutein LRb antagonist , L39A/D40A/F41A , fully inhibited leptin-induced phosphorylation of JAK2 , ERK1/2 , and Akt/PKB , and partially abrogated effects on apoptotic proteins .\n\nIn LNCaP and PC3/AR cells , leptin increased AR protein levels in correlation with raised apoptotic markers .\n\nThus , AR may mediate , at least partly , the leptin-induced apoptotic response .\n\nCONCLUSIONS Leptin can clearly induce apoptosis in human PCa cell lines .\n\nThese findings could lead to development of new leptin agonists with enhanced pro-apoptotic effects and targeted for use in human PCa .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "E6-associated protein ( E6-AP ) is a dual function protein .\n\nIt acts as an E3 ubiquitin-protein ligase enzyme and coactivator of steroid hormone receptors such as estrogen ( ER\u03b1 ) and progesterone ( PR ) receptors .\n\nIt promotes the degradation of ER\u03b1 and PR through the ubiquitin-proteasome pathway .\n\nFurthermore , it has been shown that the levels of E6-AP are inversely associated with that of ER\u03b1 in human breast tumors .\n\nBut the role of wild-type human E6-AP and its ubiquitin-protein ligase activity in mammary tumorigenesis is still unknown .\n\nTo investigate this role , the authors utilized transgenic mice lines that specifically overexpress either the wild-type human E6-AP ( E6-AP(WT) ) or the ubiquitin-protein ligase defective E6-AP that contains C833S mutation ( E6-AP(C833S) ) in the mammary gland .\n\nTo further substantiate the role of E6-AP in the development of breast tumorigenesis , it was also examined the expression of E6-AP in a large cohort of human breast cancer samples .\n\nThe transgenic mice that overexpress wild-type E6-AP ( E6-AP(WT) ) fail to develop mammary tumors .\n\nUnlike the E6-AP(WT) mice , the E6-AP(C833S) mice that overexpress ubiquitin-protein ligase defective E6-AP protein develop mammary hyperplasia with a median latency of 18months .\n\nThese observations suggest that the inactivation of the ubiquitin-protein ligase function of E6-AP is sufficient to initiate the process of mammary tumor development .\n\nFurthermore , the data also suggests that E6-AP exerts its effects on target cells by modulating the protein levels and functions of ER\u03b1 and PR .\n\nIn addition , it was found in human breast cancer patients that the level of E6-AP is decreased in invasive breast tumors compared to normal breast tissue .\n\nMoreover , the authors also show that the survival patterns for E6-AP negative patients were worse compared to E6-AP positive patients .\n\nTaken together , these data suggests that E6-AP may act as a tumor suppressor in breast .", "output": "Activating invasion and metastasis" }, { "input": "Ultraviolet ( UV ) of sunlight is a complete carcinogen that can burn skin , enhance inflammation , and drive skin carcinogenesis .\n\nPreviously , we have shown that sulforaphane ( SFN ) inhibited chemically induced skin carcinogenesis via nuclear factor ( erythroid-derived 2)-like 2 ( Nrf2 ) and others have shown that broccoli sprout extracts containing high SFN protected against UV-induced skin carcinogenesis in SKH-1 hairless mice .\n\nA recent study showed that there was no difference between Nrf2 knockout ( Nrf2 KO ) and Nrf2 wild-type ( WT ) BALB/C mice after exposing to high dose of UVB .\n\nSince Nrf2 plays critical roles in the anti-oxidative stress/anti-inflammatory responses , it is relevant to assess the role of Nrf2 for photoprotection against UV .\n\nIn this context , the role of Nrf2 in UVB-induced skin inflammation in Nrf2 WT and Nrf2 KO C57BL/6 mice was studied .\n\nA single dose of UVB ( 300\u2009mJ/cm(2) ) resulted in skin inflammation in both WT and Nrf2 KO ( -/- ) mice ( KO mice ) at 8\u2009h and 8\u2009d following UVB irradiation .\n\nIn the WT mice inflammation returned to the basal level to a greater extent when compared to the KO mice .\n\nSFN treatment of Nrf2 WT but not Nrf2 KO mice restored the number of sunburn cells back to their basal level by 8\u2009d after UVB irradiation .\n\nAdditionally , UVB-induced short-term inflammatory biomarkers ( interleukin-1\u03b2 and interleukin-6 ) were increased in the KO mice and UVB-induced apoptotic cells in the KO mice were significantly higher as compared to that in the WT .\n\nTaken together , our results show that functional Nrf2 confers a protective effect against UVB-induced inflammation , sunburn reaction , and SFN-mediated photoprotective effects in the skin .", "output": "Tumor promoting inflammation" }, { "input": "The purpose of this study was to determine the impact of the non-steroidal anti-inflammatory drug tepoxalin on canine tumour cell growth and describe the changes associated with tepoxalin treatment .\n\nIn vitro experiments were performed to assess tepoxalin-associated alterations in tumour cell growth .\n\nClinically achievable tepoxalin concentrations did not significantly alter tumour cell growth in vitro .\n\nVascular endothelial growth factor ( VEGF ) production and hypoxia-inducible factor-1\u03b1 dose-dependently increased in vitro in the presence of tepoxalin .\n\nA canine osteosarcoma xenograft was used to determine in vivo effects of tepoxalin on tumour growth and angiogenesis .\n\nDespite increased VEGF in vitro , there was a significant growth delay associated with tepoxalin treatment .\n\nNormal dogs were administered tepoxalin to assess effects on systemic VEGF production , but not found to have significantly increased VEGF .\n\nThese data suggest that tepoxalin may moderately inhibit tumour growth and may be administered as an analgesic to tumour-bearing dogs .", "output": "Inducing angiogenesis" }, { "input": "NK1 is a tachykinin receptor highly relevant to tumorigenesis and metastasis development in breast cancer and other carcinomas .\n\nDespite the substantial efforts done to develop potent NK1 receptor antagonists , none of these antagonists had shown good antitumor activity in clinical trials .\n\nNow , we have tested the effect of inhibition of the neuropeptide Substance P ( SP ) , a NK1 ligand , as a potential therapeutic approach in cancer .\n\nWe found that the inhibition of SP with antibodies strongly inhibit cell growth and induce apoptosis in breast , colon , and prostate cancer cell lines .\n\nThese effects were accompained by a decrease in the mitogen-activated kinase singaling pathway .\n\nInterestingly , in some cell lines SP abrogation decreased the steady state of Her2 and EGFR , suggesting that SP-mediated signaling is important for the basal activity of these ErbB receptors .\n\nIn consequence , we observed a blockade of the cell cycle progression and the inhibition of several cell cycle-related proteins including mTOR .\n\nSP inhibition also induced cell death in cell lines resistant to Lapatinib and Trastuzumab that have increased levels of active Her2 , suggesting that this therapeutic approach could be also effective for those cancers resistant to current anti-ErbB therapies .\n\nThus , we propose a new therapeutic strategy for those cancers that express NK1 receptor and/or other tachykinin receptors , based in the immuno-blockade of the neuropeptide SP .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Expression of epidermal growth factor receptor ( EGFR ) , a potent regulator of cellular homeostasis , is associated with aggressive tumor behavior .\n\nThe mechanism by which EGFR inhibition functions is unclear , with controversial results demonstrating an effect on the tumor cells , endothelial cells , or pericytes .\n\nEGFR activation has been linked to the expression of vascular endothelial growth factor ( VEGF ) , a known mitogen of angiogenesis , but the relationship between these factors and their effect on tumor vessel development is vague .\n\nWe hypothesized that using an EGFR inhibitor on a human Ewing's sarcoma model would inhibit tumor growth by suppressing vessel proliferation .\n\nMETHODS A cell proliferation assay was performed on the Ewing's sarcoma ( SK-NEP-1 ) cell line .\n\nTumor cells were implanted intrarenally in athymic mice .\n\nAnimals received daily gavage with vehicle or gefitinib 1 wk following implantation .\n\nMice ( n = 12/cohort ) were euthanized 6 wk following implantation .\n\nRemaining mice were maintained without treatment for 2 wk .\n\nVascular changes were assessed by angiography and immunohistochemically .\n\nEGFR and vascular endothelial growth factor ( VEGF ) expression were quantified using quantitative polymerase chain reaction ( qPCR ) .\n\nRESULTS Gefitinib suppressed in vitro cell growth with an IC(50) = 1.36 \u03bcM .\n\nMinimal tumor growth suppression was noted at 6 wk ( 6.01 \ufffd 1.2 g in control versus 4.61 \ufffd 0.9 g treated , P = 0.36 ) .\n\nAfter cessation of gefitinib , tumor growth was increased in both groups ( 7.37 \ufffd 1.62 g versus 6.77 \ufffd 1.53 g , P = 0.79 ) .\n\nMicrovessel density was unchanged despite EGFR inhibition ( 161,000 \ufffd 16,000 pixels versus 135,000 \ufffd 18,000 pixels , P = 0.31 ) .\n\nAt 6 wk , the vascular maturity index was similar in both groups ( 3.63 \ufffd 1.12 versus 4.09 \ufffd 1.71 , P = 0.83 ) .\n\nA downward trend in EGFR expression ( 49% of control ) and an upward trend in VEGF levels ( 50% of control ) occurred in the treated group .\n\nCONCLUSIONS EGFR expression was suppressed in cultured cells and xenograft tumors .\n\nDespite a cytotoxic effect on cell lines , gefitinib had little effect on tumor growth .\n\nNo effects on the tumor vasculature were noted in the setting of EGFR suppression , suggesting that angiogenesis induced by SK-NEP-1 cells is refractory to EGFR inhibition .\n\nInterestingly , the resulting increase in VEGF expression following EGFR blockade , provides an alternative pro-angiogenic pathway promoting tumor survival .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "BACKGROUND Androgens and the androgen receptor ( AR ) play important roles in the development of male urogenital organs .\n\nWe previously found that mice with total AR knockout ( ARKO ) and epithelial ARKO failed to develop normal prostate with loss of differentiation .\n\nWe have recently knocked out AR gene in smooth muscle cells and found the reduced luminal infolding and IGF-1 production in the mouse prostate .\n\nHowever , AR roles of stromal fibroblasts in prostate development remain unclear .\n\nMETHODS To further probe the stromal fibroblast AR roles in prostate development , we generated tissue-selective knockout mice with the AR gene deleted in stromal fibroblasts ( FSP-ARKO ) .\n\nWe also used primary culture stromal cells to confirm the in vivo data and investigate mechanisms related to prostate development .\n\nRESULTS The results showed cellular alterations in the FSP-ARKO mouse prostate with decreased epithelial proliferation , increased apoptosis , and decreased collagen composition .\n\nFurther mechanistic studies demonstrated that FSP-ARKO mice have defects in the expression of prostate stromal growth factors .\n\nTo further confirm these in vivo findings , we prepared primary cultured mouse prostate stromal cells and found knocking down the stromal AR could result in growth retardation of prostate stromal cells and co-cultured prostate epithelial cells , as well as decrease of some stromal growth factors .\n\nCONCLUSIONS Our FSP-ARKO mice not only provide the first in vivo evidence in Cre-loxP knockout system for the requirement of stromal fibroblast AR to maintain the normal development of the prostate , but may also suggest the selective knockdown of stromal AR might become a potential therapeutic approach to battle prostate hyperplasia and cancer .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Identification of the proteins that are associated with estrogen receptor ( ER ) status is a first step towards better understanding of the hormone-dependent nature of breast carcinogenesis .\n\nAlthough a number of gene expression analyses have been conducted , protein complement has not been systematically investigated to date .\n\nBecause proteins are primary targets of therapeutic drugs , in this study , we have attempted to identify proteomic signatures that demarcate ER-positive and -negative breast cancers .\n\nUsing highly enriched breast tumor cells , replicate analyses from 3 ER\u03b1+ and 3 ER\u03b1- human breast tumors resulted in the identification of 2,995 unique proteins with \u22652 peptides .\n\nAmong these , a number of receptor tyrosine kinases and intracellular kinases that are abundantly expressed in ER\u03b1+ and ER\u03b1- breast cancer tissues were identified .\n\nFurther , label-free quantitative proteome analysis revealed that 236 proteins were differentially expressed in ER\u03b1+ and ER\u03b1- breast tumors .\n\nAmong these , 141 proteins were selectively up-regulated in ER\u03b1+ , and 95 proteins were selectively up-regulated in ER\u03b1- breast tumors .\n\nComparison of differentially expressed proteins with a breast cancer database revealed 98 among these have been previously reported to be involved in breast cancer .\n\nBy Gene Ontology molecular function , dehydrogenase , reductase , cytoskeletal proteins , extracellular matrix , hydrolase , and lyase categories were significantly enriched in ER\u03b1+ , whereas selected calcium-binding protein , membrane traffic protein , and cytoskeletal protein were enriched in ER\u03b1- breast tumors .\n\nBiological process and pathway analysis revealed that up-regulated proteins of ER\u03b1+ were overrepresented by proteins involved in amino acid metabolism , proteasome , and fatty acid metabolism , while up-regulated proteins of ER\u03b1- were overrepresented by proteins involved in glycolysis pathway .\n\nThe presence and relative abundance of 4 selected differentially abundant proteins ( liprin-\u03b11 , fascin , DAP5 , and \u03b2-arrestin-1 ) were quantified and validated by immunohistochemistry .\n\nIn conclusion , unlike in vitro cell culture models , the in vivo signaling proteins and pathways that we have identified directly from human breast cancer tissues may serve as relevant therapeutic targets for the pharmacological intervention of breast cancer .", "output": "Cellular energetics" }, { "input": "The androgen receptor ( AR ) plays a central role in prostate cancer progression to the castration-resistant ( CR ) lethal state .\n\nL-Dopa decarboxylase ( DDC ) is an AR coactivator that increases in expression with disease progression and is coexpressed with the receptor in prostate adenocarcinoma cells , where it may enhance AR activity .\n\nHere , we hypothesize that the DDC enzymatic inhibitor , carbidopa , can suppress DDC-coactivation of AR and retard prostate tumor growth .\n\nTreating LNCaP prostate cancer cells with carbidopa in transcriptional assays suppressed the enhanced AR transactivation seen with DDC overexpression and decreased prostate-specific antigen ( PSA ) mRNA levels .\n\nCarbidopa dose-dependently inhibited cell growth and decreased survival in LNCaP cell proliferation and apoptosis assays .\n\nThe inhibitory effect of carbidopa on DDC-coactivation of AR and cell growth/survival was also observed in PC3 prostate cancer cells ( stably expressing AR ) .\n\nIn vivo studies demonstrated that serum PSA velocity and tumor growth rates elevated \u223c2-fold in LNCaP xenografts , inducibly overexpressing DDC , were reverted to control levels with carbidopa administration .\n\nIn castrated mice , treating LNCaP tumors , expressing endogenous DDC , with carbidopa delayed progression to the CR state from 6 to 10 weeks , while serum PSA and tumor growth decreased 4.3-fold and 5.4-fold , respectively .\n\nOur study is a first time demonstration that carbidopa can abrogate DDC-coactivation of AR in prostate cancer cells and tumors , decrease serum PSA , reduce tumor growth and delay CR progression .\n\nSince carbidopa is clinically approved , it may be readily used as a novel therapeutic strategy to suppress aberrant AR activity and delay prostate cancer progression .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Ruthenium(II) methylimidazole complexes , with the general formula [ Ru(MeIm)(4)(N\u2322N)](2+) ( N\u2322N=tip ( RMC1 ) , iip ( RMC2 ) , dppz ( RMC3 ) , dpq ( RMC4 ) ; MeIm=1-methylimidazole , tip=2-(thiophene-2-yl)-1H-imidazo [ 4,5-f ] [ 1,10]phenanthroline , iip=2-(1H-imidazol-4-yl)-1H-imidazo [ 4,5-f ] [ 1,10]phenanthroline , dppz=dipyrido[3,2-a:2',3'-c]phenazine , dpq=pyrazino [ 2,3-f ] [ 1,10]phenanthroline ) , were synthesized and characterized .\n\nAs determined by MTT ( 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide ) assay , these complexes displayed potent anti-proliferation activity against various cancer cells .\n\nRMC1 inhibited the growth of A549 ( human lung adenocarcinoma ) lung cells through induction of apoptotic cell death , as evidenced by the accumulation of cell population in sub-G1 phase .\n\nRMC1 also induced the depletion of mitochondrial membrane potential in A549 cells by regulating the expression of pro-survival and pro-apoptotic Bcl-2 family members .\n\nAnother experiment showed that Bid protein was also activated by RMC1 , which implied that RMC1 could existed two pathways crosstalk , namely , have exogenous death receptor signaling pathway .\n\nThese results demonstrated that RMC1 induced cancer cell death by acting on both mitochondrial and death receptor apoptotic pathways , suggesting that RMC1 could be a candidate for further evaluation as a chemotherapeutic agent against human cancers .", "output": "Resisting cell death" }, { "input": "The type I insulin-like growth factor receptor ( IGF1R ) contributes to cancer cell biology .\n\nDisruption of IGF1R signaling alone or in combination with cytotoxic agents has emerged as a new therapeutic strategy .\n\nOur laboratory has shown that sequential treatment with doxorubicin ( DOX ) and anti-IGF1R antibodies significantly enhanced the response to chemotherapy .\n\nIn this study , we examined whether inhibition of the tyrosine kinase activity of this receptor family would also enhance chemotherapy response .\n\nCis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo[1,5-a]pyrazin-8-ylamine ( PQIP ) inhibited IGF1R and insulin receptor ( InsR ) kinase activity and downstream activation of ERK1/2 and Akt in MCF-7 and LCC6 cancer cells .\n\nPQIP inhibited both monolayer growth and anchorage-independent growth in a dose-dependent manner .\n\nPQIP did not induce apoptosis , but rather , PQIP treatment was associated with an increase in autophagy .\n\nWe examined whether sequential or combination therapy of PQIP with DOX could enhance growth inhibition .\n\nPQIP treatment together with DOX or DOX followed by PQIP significantly inhibited anchorage-independent growth in MCF-7 and LCC6 cells compared to single agent alone .\n\nIn contrast , pre-treatment with PQIP followed by DOX did not enhance the cytotoxicity of DOX in vitro .\n\nFurthermore , OSI-906 , a PQIP derivative , inhibited IGF-I signaling in LCC6 xenograft tumors in vivo .\n\nWhen given once a week , simultaneous administration of OSI-906 and DOX significantly enhanced the anti-tumor effect of DOX .\n\nIn summary , these results suggest that timing and duration of the IGF1R/InsR tyrosine kinase inhibitors with chemotherapeutic agents should be evaluated in clinical trials .\n\nLong-term disruption of IGF1R/InsR may not be necessary when combined with cytotoxic chemotherapy .", "output": "Resisting cell death" }, { "input": "Programmed cell death 6 ( PDCD6 ) was originally found as a pro-apoptotic protein , but its molecular mechanism is not well understood .\n\nIn this study , we have attempted to investigate the effects of PDCD6 on the inhibition of angiogenesis-mediated cell growth as a novel anti-angiogenic protein .\n\nPurified recombinant human PDCD6 inhibited cell migration in a concentration-time-dependent manner .\n\nWe also found that overexpressed PDCD6 suppressed vascular endothelial growth factor ( VEGF)-induced proliferation , invasion , and capillary-like structure tube formation in vitro .\n\nPDCD6 suppressed phosphorylation of signaling regulators downstream from PI3K , including Akt , mammalian target of rapamycin ( mTOR ) , glycogen synthase kinase-3\u03b2(GSK-3\u03b2) , ribosomal protein S6 kinase ( p70S6K ) , and also decreased cyclin D1 expression .\n\nWe found binding PDCD6 to VEGFR-2 , a key player in the PI3K/mTOR/P70S6K signaling pathway .\n\nTaken together , these data suggest that PDCD6 plays a significant role in modulating cellular angiogenesis .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "We recently isolated an exon-4-deleted epidermal growth factor receptor ( EGFR ) variant , termed de4 EGFR .\n\nBecause the extracellular domain alteration of receptors often influences the antitumor effect of therapeutic antibodies , it is essential to test the sensitivity of de4 EGFR(+) tumors to anti-EGFR antibodies .\n\nTherefore , in this study , the antitumor activities of mAb CH12 , an anti-EGFRvIII antibody developed in our laboratory , as well as a U.S. Food and Drug Administration-approved anti-EGFR antibody , cetuximab ( C225 ) , were characterized on de4 EGFR(+) models .\n\nThe results of FACS assays showed that CH12 bound to de4 EGFR with a higher avidity than did C225 .\n\nInterestingly , CH12 , but not C225 , significantly inhibited the metastasis and growth of U87MG-de4 EGFR xenografts , with a growth-inhibition ratio of 46.48% in vivo , and prolonged the survival of the tumor-bearing mice by 37.2% .\n\nTreatment with CH12 significantly suppressed tumor proliferation and angiogenesis with increased tumor apoptosis .\n\nMechanistically , de4 EGFR protein expression was virtually undetectable in the U87MG-de4 EGFR xenografts treated with CH12 .\n\nThis may account for the observed reduction of Akt and Erk phosphorylation , cyclin D1 , Bcl-2 , and Bcl-x(L) expression and the increase of p27 and E-cadherin expression .\n\nIntriguingly , LAMP-1 , a major component of the lysosome , was significantly up-regulated in the CH12-treated group but not in the C225-treated group , suggesting its contribution to the degradation of de4 EGFR .\n\nTaken together , our data demonstrated that mAb CH12 is a promising therapeutic agent for treating de4 EGFR(+) gliomas .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Glioma tumors are refractory to conventional treatment .\n\nGlioblastoma multiforme is the most aggressive type of primary brain tumors in humans .\n\nIn this study , we introduce oxidative stress-energy depletion ( OSED ) therapy as a new suggested treatment for glioblastoma .\n\nOSED utilizes D-amino acid oxidase ( DAO ) , which is a promising therapeutic protein that induces oxidative stress and apoptosis through generating hydrogen peroxide ( H2O2 ) .\n\nOSED combines DAO with 3-bromopyruvate ( 3BP ) , a hexokinase II ( HK II ) inhibitor that interferes with Warburg effect , a metabolic alteration of most tumor cells that is characterized by enhanced aerobic glycolysis .\n\nOur data revealed that 3BP induced depletion of energetic capabilities of glioma cells. 3BP induced H2O2 production as a novel mechanism of its action .\n\nC6 glioma transfected with DAO and treated with D-serine together with 3BP-sensitized glioma cells to 3BP and decreased markedly proliferation , clonogenic power and viability in a three-dimensional tumor model with lesser effect on normal astrocytes .\n\nDAO gene therapy using atelocollagen as an in vivo transfection agent proved effective in a glioma tumor model in Sprague-Dawley ( SD ) rats , especially after combination with 3BP .\n\nOSED treatment was safe and tolerable in SD rats .\n\nOSED therapy may be a promising therapeutic modality for glioma .", "output": "Cellular energetics" }, { "input": "BACKGROUND As a highly conserved system , the activation of the Notch pathway has been implicated in the tumorigenesis of various hematologic diseases , including leukemias , lymphomas , and multiple myeloma .\n\nThe Notch3 receptor is frequently expressed in T-cell acute lymphoblastic leukemia ( T-ALL ) .\n\nMETHODS To explore its possibility as a therapeutic target for T-ALL , we investigated the effect of Notch3 silencing on Jurkat and SupT1 cells using a novel tumor-specific short hairpin RNA ( shRNA ) driven by survivin promoters .\n\nRESULTS We found that downregulated expression of Notch3 correlated with significant apoptosis and inhibition of proliferation .\n\nCONCLUSION These facts suggest that downregulating expression of Notch3 could attenuate the Notch signaling activity in T-ALL .\n\nAll these results indicate that inhibition of Notch3 expression can result in potent antitumor activity in T-ALL .", "output": "Resisting cell death" }, { "input": "BACKGROUND Hepatocellular carcinoma ( HCC ) is the most common liver cancer .\n\nTherapeutic results are usually unsatisfactory because liver tumors recur often .\n\nImmunologic factors may be related to the recurrence of HCC ; however , this possibility is mentioned only rarely .\n\nMETHODS Thirty HCC patients undergoing hepatectomies were divided into 3 groups according to the diameters of their HCCs : group A ( n = 8 ) , diameter \u22643 cm ; group B ( n = 8 ) , diameter >3 cm and \u22645 cm ; and group C ( n = 14 ) , diameter >5 cm .\n\nT-lymphocytes from peripheral blood , nontumor liver tissue , and the HCC were analyzed .\n\nRESULTS The percentage of CD25+ in the CD4+ T cells did not differ between the peripheral blood and the nontumor liver tissue among the 3 groups .\n\nCD25+ cells were increased in the tumor tissue in group C patients ( range , 6-41% ; median , 22.9% ; P = .003 ) , compared to group A patients .\n\nThe percentage of CD25+ in the CD4+ T cells in tumor tissue was positively correlated with tumor sizes ( r = 0.556 ) .\n\nThese CD4+ CD25+ lymphocytes produced transforming growth factor-\u03b2 and interferon-\u03b3 but not interleukin-10 , and were anergic to plate-coated monoclonal antibodies ( anti-CD3/anti-CD28 ) .\n\nThe characteristics of these antibodies were comparable to those of regulatory T cells .\n\nWhen the infiltration lymphocytes including CD4+ CD25+ T cells were added to the mixed lymphocyte reaction activated by autologous tumor lysate-pulsed dendritic cells , the proliferation of lymphocytes was inhibited .\n\nCONCLUSION The increase of CD4+ CD25+ T cells in the tumor microenvironment correlates with tumor sizes .\n\nThese CD4+ CD25+ regulatory T cells appeared to suppress the immune response activated by dendritic cells .", "output": "Tumor promoting inflammation" }, { "input": "Thyroid hormone ( T(3) ) mediates cellular growth , development , and differentiation by binding to the nuclear thyroid hormone receptor ( TR ) .\n\nRecent studies suggest that long-term hypothyroidism is associated with human hepatocellular carcinoma ( HCC ) independent from other major HCC risk factors .\n\nDickkopf ( DKK ) 4 , a secreted protein , antagonizes the Wnt signal pathway .\n\nIn this study , we demonstrate that T(3) may play a suppressor role by inducing DKK4 expression in HCC cells at both the messenger RNA ( mRNA ) and protein levels .\n\nDKK4 was down-regulated in 67.5% of HCC cancerous tissues .\n\nThe decrease in DKK4 levels was accompanied by a concomitant decrease in TR protein levels in the matched cancerous tissues in 31% of tissues compared by immunoblotting with the adjacent noncancerous tissues .\n\nFurther , TR and DKK4 expression levels were positively correlated in both normal and cancerous specimens by tissue array analysis .\n\nIn function assays , stable DKK4 transfected into J7 or HepG2 cells decreased cell invasion in vitro .\n\nConversely , knocking down DKK4 restores cell invasiveness .\n\nDKK4-expressing J7 clones showed increased degradation of \u03b2-catenin , but down-regulation of CD44 , cyclin D1 , and c-Jun .\n\nTo investigate the effect of DKK4 and TR on tumor growth in vivo , we established a xenograft of J7 cells in nude mice .\n\nJ7-DKK4 and J7-TR\u03b11 overexpressing mice , which displayed growth arrest , lower lung colony formation index , and smaller tumor size than in control mice , supporting an inhibitory role of DKK4 in tumor progression .\n\nCONCLUSION : Taken together , these data suggest that the TR/DKK4/Wnt/\u03b2-catenin cascade influences the proliferation and migration of hepatoma cells during the metastasis process and support a tumor suppressor role of the TR .", "output": "Activating invasion and metastasis" }, { "input": "Uncontrolled estrogen exposure can induce an imbalance in BCL2/BAX expression in endometrial cells , leading to precancerous lesions and type I endometrial adenocarcinoma .\n\nThis study aimed to explore the mechanism underlying this phenomenon .\n\nWe show that the activated estrogen receptor can suppress the expression of BAX by upregulating a group of microRNAs including hsa-let-7 family members and hsa-miR-27a , thereby promoting an increased BCL2/BAX ratio as well as enhanced survival and proliferation in the affected cells .\n\nThese ER-regulated hsa-let-7 microRNAs can be detected in most hyperplastic endometria , suggesting their potential utility as indicators of estrogen over-exposure .", "output": "Sustaining proliferative signaling" }, { "input": "Nitric oxide ( NO ) shows tumoricidal activity .\n\nWe had previously reported that NO downregulates the phosphatidylinositol-3-kinase/Akt pathway , but upregulates the MEK/ERK pathway downstream of growth factor signaling .\n\nWe hypothesized that NO donor and MEK inhibitor in combination synergistically inhibit the viability of cancer cells compared to either NO donor or MEK inhibitor alone .\n\nWe determined the effects of S-nitrosoglutathione ( GSNO , NO-donor ) and U0126 ( MEK inhibitor ) on insulin-like growth factor-I ( IGF-I ) and epidermal growth factor ( EGF ) signaling , proliferation and invasion in cancer cell lines .\n\nGSNO inhibits phosphorylation of IGF-I receptor ( IGF-IR ) , EGF receptor ( EGFR ) and Akt , but upregulates ERK1/2 phosphorylation in MIAPaCa-2 and HCT-116 cells after stimulation by IGF-I and EGF .\n\nOn the other hand , U0126 inhibits phosphorylation of ERK1/2 , but upregulates phosphorylation of IGF-IR and EGFR in MIAPaCa-2 and HCT-116 cells .\n\nThe combination of GSNO and U0126 downregulates phosphorylation of IGF-IR , EGFR , Akt and ERK1/2 after stimulation by IGF-I and EGF .\n\nGSNO as well as U0126 , inhibits the proliferation of MIAPaCa-2 , HCT-116 , Panc-1 , MCF-7 , HT-29 and AGS cells in a dose-dependent manner .\n\nGSNO and U0126 in combination synergistically inhibit proliferation and invasion of cancer cells .\n\nThese results indicate that the combined treatment of NO donor and MEK inhibitor may be promising in cancer therapy .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Zinc dyshomeostasis can induce cell death .\n\nHowever , the mechanisms involved have not been fully elucidated in prostate cancer ( PCa ) cells , which differ dramatically from normal cells in their zinc handling ability .\n\nHere , we studied the effects of the ionophore Zn-pyrithione ( ZP ) and the chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine ( TPEN ) .\n\nBoth compounds induced cell death at micromolar concentrations when incubated with androgen-dependent ( LNCaP ) , androgen-independent ( PC3 , DU145 ) and androgen-sensitive ( C4-2 ) PCa cell-lines .\n\nCompared to PCa cells , RWPE1 prostate epithelial cells were less sensitive to ZP and more sensitive to TPEN , but total cellular zinc levels were changed similarly .\n\nZnSO4 enhanced the toxicity of ZP , but inhibited the effects of TPEN as expected .\n\nThe morphological/biochemical responses to ZP and TPEN differed .\n\nZP decreased ATP levels and stimulated ERK , AKT and PKC phosphorylation .\n\nDNA laddering was observed only at low doses of ZP but all doses of TPEN .\n\nTPEN activated caspase 3/7 and induced PARP-cleavage , DNA-fragmentation , ROS-formation and apoptotic bodies .\n\nPKC and ERK-pathway inhibitors , and antioxidants protected against ZP-induced but not TPEN-induced death .\n\nInhibitors of MPTP-opening protected both .\n\nCell death in response to TPEN ( but not ZP ) was diminished by a calpain inhibitor and largely prevented by a caspase 3 inhibitor .\n\nOverall , the results indicated primarily a necrotic cell death for ZP and an apoptotic cell death for TPEN .\n\nThe enhanced sensitivity of PCa cells to ZP and the apparent ability of ZP and TPEN to kill quiescent and rapidly dividing cells in a p53-independent manner suggest that ZP/TPEN might be used to develop adjunct treatments for PCa .", "output": "Resisting cell death" }, { "input": "Insulin-like growth factor ( IGF)-I receptor ( IGF-IR ) signaling is required for carcinogenicity and progression of several cancers but the function of this pathway and its utility as a therapeutic target have not been studied comprehensively in biliary tract carcinomas ( BTC ) .\n\nWe investigated the immunohistochemical expression of elements of the IGF axis , matrilysin , overexpression of p53 and the methylation status of the IGFBP-3 promoter in 80 surgically resected BTC .\n\nWe also assessed the effect of IGF-IR blockade on signal transduction , proliferation and survival in three BTC cell lines using a new tyrosine kinase inhibitor , BMS-536924 , and dominant negative IGF-IR ( IGF-IR/dn ) .\n\nThe effects of IGF-IR blockade was also studied in nude mouse xenograft models .\n\nIGF-I was expressed in 60% and IGF-II in 50% of tumors .\n\nHigh expression was associated with tumor size .\n\nIGF-IR was expressed in 69% of the cases and was associated with advanced stage and matrilysin expression .\n\nHypermethylation of the IGFBP-3 promoter was detected in 41% of BTC and was inversely correlated with p53 expression .\n\nBMS-536924 blocked autophosphorylation of IGF-IR and both Akt and ERK activation by both IGF-I and insulin .\n\nBMS-536924 suppressed proliferation and tumorigenicity in vitro in a dose-dependent fashion .\n\nThis inhibitor upregulated chemotherapy-induced apoptosis in a dose-dependent fashion .\n\nMoreover , IGF-IR blockade was effective against tumors in mice .\n\nIGF-IR might identify a subset of BTC with a particularly aggressive phenotype and is a candidate therapeutic target in this disease .\n\nBMS-536924 might have significant therapeutic utility .", "output": "Resisting cell death" }, { "input": "Breast cancer is the malignant neoplasia with the highest incidence in women worldwide .\n\nChronic oxidative stress and inflammation have been indicated as major mediators during carcinogenesis and cancer progression .\n\nHuman studies have not considered the complexity of tumor biology during the stages of cancer advance , limiting their clinical application .\n\nThe purpose of this study was to characterize systemic oxidative stress and immune response parameters in early ( ED ; TNM I and II ) and advanced disease ( AD ; TNM III and IV ) of patients diagnosed with infiltrative ductal carcinoma breast cancer .\n\nOxidative stress parameters were evaluated by plasmatic lipoperoxidation , carbonyl content , thiobarbituric reactive substances ( TBARS ) , nitric oxide levels ( NO ) , total radical antioxidant parameter ( TRAP ) , superoxide dismutase , and catalase activities and GSH levels .\n\nImmune evaluation was determined by TNF-\u03b1 , IL-1\u03b2 , IL-12 , and IL-10 levels and leukocytes oxidative burst evaluation by chemiluminescence .\n\nTissue damage analysis included heart ( total CK and CKMB ) , liver ( AST , ALT , GGT ) , and renal ( creatinine , urea , and uric acid ) plasmatic markers .\n\nC-reactive protein ( CRP ) and iron metabolism were also evaluated .\n\nAnalysis of the results verified different oxidative stress statuses occur at distinct cancer stages .\n\nED was characterized by reduction in catalase , 8-isoprostanes , and GSH levels , with enhanced lipid peroxidation and TBARS levels .\n\nAD exhibited more pronounced oxidative status , with reduction in catalase activity and TRAP , intense lipid peroxidation and high levels of NO , TBARs , and carbonyl content .\n\nED patients presented a Th2 immune pattern , while AD exhibited Th1 status .\n\nCRP levels and ferritin were increased in both stages of disease .\n\nLeukocytes burst impairment was observed in both the groups .\n\nPlasma iron levels were significantly elevated in AD .\n\nThe data obtained indicated that oxidative stress enhancement and immune response impairment may be necessary to ensure cancer progression to advanced stages and may result from both host and tumor inflammatory mediators .", "output": "Tumor promoting inflammation" }, { "input": "Peroxisome proliferator-activated receptor-\u03b3 ( PPAR\u03b3 ) is an anti-inflammatory molecule .\n\nTo study its biologic function in myeloid cells , dominant-negative PPAR\u03b3 ( dnPPAR\u03b3 ) was overexpressed in a myeloid-specific bitransgenic mouse model .\n\nIn this bitransgenic system , overexpression of the dnPPAR\u03b3-Flag fusion protein in myeloid-lineage cells abnormally elevated frequencies and total numbers of IL-7R\u03b1(-)Lin(-)c-Kit(+)Sca-1(-) , Lin(-)/Scal(+)/c-Kit(+) , common myeloid , and granulocyte-monocyte progenitor populations in the BM. dnPPAR\u03b3 overexpression led to up-regulation of IL-1\u03b2 , IL-6 , and TNF\u03b1 in the blood plasma .\n\nAs a result , CD11b(+)Ly6G(+) cells were systemically increased in association with activation of Stat3 , NF-\u03baB , Erk1/2 , and p38 molecules .\n\nMyeloid-derived suppressor cells ( MDSCs ) inhibited the proliferation and lymphokine production of wild-type CD4+ T cells in vitro .\n\nCD4+ T cells from doxycycline-treated bitransgenic mice displayed reduced proliferation and lymphokine release .\n\nBoth CD4+ and CD8+ T-cell populations were decreased in doxycycline-treated bitransgenic mice .\n\nMultiple forms of carcinoma and sarcoma in the lung , liver , spleen , and lymph nodes were observed in doxycycline-treated bitransgenic mice .\n\nBM transplantation revealed that a myeloid-autonomous defect was responsible for MDSC expansion , immunosuppression , and tumorigenesis in these mice .\n\nThese studies suggest that anti-inflammatory PPAR\u03b3 in myeloid-lineage cells plays a key role in controlling pro-inflammatory cytokine synthesis , MDSC expansion , immunosuppression , and the development of cancer .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Endothelin plays important roles in various physiological functions including vascular constriction .\n\nRecent studies reported that the endothelin receptors ETA and ETB are highly expressed in lung and skin tumor tissues .\n\nIn contrast , there are few reports on endothelin signalling in the proliferation of head and neck cancer .\n\nWe found that both ETA and ETB endothelin receptors were overexpressed in tumor cells of tongue cancer samples by immunohistochemistry .\n\nETA and ETB were expressed in cultured lingual and esophageal squamous cell carcinoma ( SCCs ) cell lines .\n\nWhen both cultured cell lines were treated with an ETA selective antagonist ( BQ123 ) or an ETB selective antagonist ( BQ788 ) , inhibition of cell growth was observed .\n\nSimilar results were observed when SCCs were treated with specific siRNA for the suppression of ETA or ETB .\n\nFurthermore , inhibition of the mitogen-activated protein ( MAP ) kinase pathway by the treatments with ET receptor antagonists and siRNA was also observed .\n\nThese results indicate that endothelin signalling may , in part , play important roles in cell growth in SCCs through the MAP kinase pathway .", "output": "Sustaining proliferative signaling" }, { "input": "Estrogen receptor ( ER ) and NF-\u03baB are transcription factors with profound effects on breast cancer cell proliferation and survival .\n\nWhile many studies demonstrate that ER and NF-\u03baB can repress each other , we previously identified a gene signature that is synergistically upregulated by these two factors in more aggressive luminal B breast tumors .\n\nHerein , we examine a novel mechanism of cross talk between ER and NF-\u03baB that results in the upregulation of the antiapoptotic gene BIRC3 ( also known as cIAP2 ) .\n\nWe demonstrate that NF-\u03baB , acting through two response elements , is required for ER recruitment to an adjacent estrogen response element ( ERE ) in the BIRC3 promoter .\n\nThis effect is accompanied by a major increase in NF-\u03baB-dependent histone acetylation around the ERE .\n\nInterestingly , CBP , a histone acetyltransferase previously implicated in repressive interactions between ER and NF-\u03baB , plays a permissive role by promoting histone acetylation and ER recruitment , as well as enhanced expression of BIRC3 .\n\nThese findings suggest a new gene regulatory mechanism by which inflammation and NF-\u03baB activation can influence ER recruitment to inherently inactive ER binding sites .\n\nThis fine-tuning mechanism may explain how two factors that generally repress each other's activity may work together on certain genes to promote breast cancer cell survival and tumor progression .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "We present a novel gain-of-function mutation of TGF-\u03b2 receptor II ( T\u03b2RII ) found in human oral squamous cell carcinoma ( OSCC ) .\n\nExpression of E221V/N238I mutant T\u03b2RII enhanced TGF-\u03b2 signaling .\n\nMutation of T\u03b2RII conferred cells higher migratory and invasive capabilities and MMP-2 activity .\n\nIn mouse tumor model , mutant tumors exhibited poor differentiation and E-cadherin relocalization to the cytosol .\n\nLipid-raft-dependent endocytosis of T\u03b2RII was attenuated in mutant T\u03b2RII , suggesting that enhancement of TGF-\u03b2 signaling by this mutation is due to delayed T\u03b2RII internalization .\n\nTaken together , our results show a novel gain-of-function T\u03b2RII mutation , which enhances TGF-\u03b2 signaling leading to more invasive phenotypic changes in human OSCC .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Androgen deprivation is currently a standard-of-care , first-line therapy for prostate cancer in the United States .\n\nAlthough this regimen effectively regresses androgen-dependent disease , relapse often occurs in an androgen-independent manner and is associated with poor prognosis .\n\nSuch castration-resistant prostate cancer represents a major clinical challenge , and the mechanisms underlying castration resistance are not fully understood .\n\nEpithelial-mesenchymal transition ( EMT ) is a key developmental process and has also been implicated in cancer metastasis and therapeutic resistance in recent years .\n\nHowever , the factors contributing to EMT in human cancers remain unclear .\n\nHere , we show that both normal mouse prostate tissue and human LuCaP35 prostate tumor explants display an EMT as well as increased stem cell-like features following androgen deprivation .\n\nImportantly , we observed similar changes in mesenchymal features in prostate tumors from patients treated with androgen-deprivation therapy .\n\nIn addition , we have delineated a feedback loop involving the androgen receptor and the Zeb1 transcription factor that seems to mediate this transition .\n\nIn summary , we show for the first time that androgen deprivation induces EMT in both normal prostate and prostate cancer , revealing a potentially important consequence of a standard-of-care treatment for prostate cancer .\n\nThis finding could have significant implications for second-line treatment strategies in this clinical setting .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "P53 has an important role in the processing of starvation signals .\n\nP53-dependent molecular mediators of the Warburg effect reduce glucose consumption and promote mitochondrial function .\n\nWe therefore hypothesized that the retention of wild-type p53 characteristic of primary glioblastomas limits metabolic demands induced by deregulated signal transduction in the presence of hypoxia and nutrient depletion .\n\nHere we report that short hairpin RNA-mediated gene suppression of wild-type p53 or ectopic expression of mutant temperature-sensitive dominant-negative p53(V135A) increased glucose consumption and lactate production , decreased oxygen consumption and enhanced hypoxia-induced cell death in p53 wild-type human glioblastoma cells .\n\nSimilarly , genetic knockout of p53 in HCT116 colon carcinoma cells resulted in reduced respiration and hypersensitivity towards hypoxia-induced cell death .\n\nFurther , wild-type p53 gene silencing reduced the expression of synthesis of cytochrome c oxidase 2 ( SCO2 ) , an effector necessary for respiratory chain function .\n\nAn SCO2 transgene reverted the metabolic phenotype and restored resistance towards hypoxia in p53-depleted and p53 mutant glioma cells in a rotenone-sensitive manner , demonstrating that this effect was dependent on intact oxidative phosphorylation .\n\nSupplementation with methyl-pyruvate , a mitochondrial substrate , rescued p53 wild-type but not p53 mutant cells from hypoxic cell death , demonstrating a p53-mediated selective aptitude to metabolize mitochondrial substrates .\n\nFurther , SCO2 gene silencing in p53 wild-type glioma cells sensitized these cells towards hypoxia .\n\nFinally , lentiviral gene suppression of SCO2 significantly enhanced tumor necrosis in a subcutaneous HCT116 xenograft tumor model , compatible with impaired energy metabolism in these cells .\n\nThese findings demonstrate that glioma and colon cancer cells with p53 wild-type status can skew the Warburg effect and thereby reduce their vulnerability towards tumor hypoxia in an SCO2-dependent manner .\n\nTargeting SCO2 may therefore represent a valuable strategy to enhance sensitivity towards hypoxia and may complement strategies targeting glucose metabolism .", "output": "Cellular energetics" }, { "input": "The aryl hydrocarbon receptor ( AhR ) is a ligand-activated transcription factor .\n\nRecent studies have reported the anti-tumor effects of the AhR in breast cancer .\n\nIn this study , we investigated the anti-tumor effect of AhR activation based on the cancer stem cell hypothesis .\n\nWe show that AhR activation suppressed mammosphere formation of MCF-7 cells and decreased the proportion of cells with high ALDH-1 ( aldehyde dehydrogenase 1 ) activity .\n\nIn addition , we also demonstrate that AhR activation regulates self-renewal signaling by down-regulating Wnt/\u03b2-catenin and Notch .", "output": "Sustaining proliferative signaling" }, { "input": "In cancer cells , the aberrant conversion of pyruvate into lactate instead of acetyl-CoA in the presence of oxygen is known as the Warburg effect .\n\nThe consequences and mechanisms of this metabolic peculiarity are incompletely understood .\n\nHere we report that p53 status is a key determinant of the Warburg effect .\n\nWild-type p53 expression decreased levels of pyruvate dehydrogenase kinase-2 ( Pdk2 ) and the product of its activity , the inactive form of the pyruvate dehydrogenase complex ( P-Pdc ) , both of which are key regulators of pyruvate metabolism .\n\nDecreased levels of Pdk2 and P-Pdc in turn promoted conversion of pyruvate into acetyl-CoA instead of lactate .\n\nThus , wild-type p53 limited lactate production in cancer cells unless Pdk2 could be elevated .\n\nTogether , our results established that wild-type p53 prevents manifestation of the Warburg effect by controlling Pdk2 .\n\nThese findings elucidate a new mechanism by which p53 suppresses tumorigenesis acting at the level of cancer cell metabolism .", "output": "Cellular energetics" }, { "input": "Histone deacetylase inhibitors ( HDACi ) are promising epigenetic cancer chemotherapeutics rapidly approaching clinical use .\n\nIn this study , we tested using in vitro and in vivo models the differential biological effects of a novel HDAC inhibitor [ belinostat ( PXD101) ] , in a wide panel of androgen-sensitive and androgen-independent tumor cells .\n\nBelinostat significantly increased acetylation of histones H3 and H4 .\n\nBelinostat potently inhibited the growth of prostate cancer cell lines ( IC50 range from 0.5 to 2.5 \ufffdM ) with cytotoxic activity preferentially against tumor cells .\n\nThis agent induced G2/M arrest and increased significantly the percentage of apoptosis mainly in androgen-sensitive tumor cells confirming its growth-inhibitory effects .\n\nThe cell death mechanisms were studied in three different prostate cancer cell lines with different androgen dependence and expression of androgen receptor ; LAPC-4 and 22rv1 ( androgen-dependent and expressing androgen receptor ) and PC3 ( androgen-independent not expressing androgen receptor ) .\n\nBelinostat induced the expression of p21 and p27 , acetylation of p53 and G2/M arrest associated with Bcl2 and Bcl-Xl downmodulation and significant reduction of survivin , IAPs and Akt/pAkt and increased caspase-8 and -9 expression/activity .\n\nBelinostat effectiveness was dependent on the androgen receptor ( AR ) , since the stable transfection of AR greatly increased the efficacy of this HDAC inhibitor .\n\nThese observations were correlated using in vivo models .\n\nWe demonstrated that belinostat preferentially resulted in antitumor effect in androgen-dependent tumor cells expressing AR .\n\nOur findings provide evidence that belinostat may be a promising anticancer drug for prostate cancer expressing AR , supporting its clinical role in prostate cancer .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Pancreatic cancer remains the fourth most common cause of cancer-related death in the United States .\n\nPotent therapeutic strategies are urgently needed for pancreatic cancer .\n\nCucurmosin is a novel type1 ribosome-inactivating protein ( RIP ) isolated from the sarcocarp of Cucurbita moschata ( pumpkin ) .\n\nDue to its cytotoxicity , cucurmosin can inhibit tumor cell proliferation through induction of apoptosis on tumor cells , but the specific mechanism is still unclear .\n\nWe explored the function of cucurmosin in BxPC-3 pancreatic cancer cells using multiple cellular and molecular approaches such as 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay , flow cytometry , reverse transcription polymerase chain reaction ( RT-PCR ) , Western blotting and transmission electron microscopy for observing typical changes and formation of apoptotic bodies .\n\nWe found that cucurmosin inhibited the proliferation of BxPC-3 cells in a time- and dose-dependent manner , and increased the cell population in the G0-G1 phase .\n\nWith increasing concentration of cucurmosin , the expression of EGFR , p-PI3K , Akt , p-Akt , mTOR , p-mTOR , P70S6K-\u03b1 , p-P70S6K-\u03b1 , 4E-BP1 and p-4E-BP1 at the protein level was decreased , whereas the expression of p-Bad and caspase-9 was elevated .\n\nHowever , the mRNA expression of EGFR did not change .\n\nThese findings suggest that cucurmosin can down-regulate the expression of EGFR by targeting .\n\nCucurmosin induces the apoptosis of BxPC-3 pancreatic cancer cells via the PI3K/Akt/mTOR signaling pathway .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Previous studies have demonstrated that mesenchymal stromal cells ( MSCs ) enhance cell survival through upregulation and secretion of stanniocalcin-1 ( STC1 ) .\n\nThis study shows that MSC-derived STC1 promotes survival of lung cancer cells by uncoupling oxidative phosphorylation , reducing intracellular reactive oxygen species ( ROS ) , and shifting metabolism towards a more glycolytic metabolic profile .\n\nMSC-derived STC1 upregulated uncoupling protein 2 ( UCP2 ) in injured A549 cells in an STC1-dependent manner .\n\nKnockdown of UCP2 reduced the ability of MSCs and recombinant STC1 ( rSTC1 ) to reduce cell death in the A549 population. rSTC1-treated A549 cells displayed decreased levels of ROS , mitochondrial membrane potential ( MMP ) , and increased lactate production , all of which were dependent on the upregulation of UCP2 .\n\nOur data suggest that MSCs can promote cell survival by regulating mitochondrial respiration via STC1 .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "OBJECTIVE The HER2 receptor is involved in pathways essential for cell proliferation , and is an important predictive and prognostic factor in breast cancer .\n\nHER2 probably plays a critical role in many types of cancer , including urothelial carcinoma of the bladder ( UCB ) .\n\nStage T1 UCB exhibits heterogeneous clinical behaviour , and the frequency of HER2 expression in such disease has not been thoroughly examined .\n\nThe aim of this study was to use an immunohistochemical technique to evaluate the frequency of HER2 expression in a defined population-based cohort of patients registered as having primary stage T1 UCB .\n\nMATERIAL AND METHODS The initial study population comprised 285 patients registered as having primary stage T1 UCB .\n\nThe original histological specimens were re-evaluated with regard to T stage and World Health Organization grade .\n\nHospital records provided information on tumour size , multiplicity , possible presence of histologically proven recurrence and progression .\n\nThe patients were followed for at least 5 years or until death .\n\nIn tumours still considered stage T1 after re-evaluation , HER2 was investigated by immunohistochemistry of paraffin-embedded material and scored according to the guidelines used in breast cancer .\n\nRESULTS After histopathological re-evaluation , 201 patients were still T1 UCB and could be investigated regarding HER2 expression .\n\nHER2 overexpression was observed in 25 of those patients ( 12.4% ) .\n\nHER2 status was not significantly associated with recurrence or progression .\n\nCONCLUSIONS HER2 was overexpressed in 12.4% of the present cohort of patients with primary stage T1 UCB .\n\nThere was no significant association between tumour HER2 status and prognosis .", "output": "Sustaining proliferative signaling" }, { "input": "The aim of the present study was to investigate the apoptosis of human ovarian cancer cell lines , A2780 and CP70 , induced by a novel curcumin analogue , B19 .\n\nThe proliferation of cells was detected with methyl thiazolyl tetrazolium ( MTT ) assay and apoptosis was examined by flow cytometry .\n\nReactive oxygen species ( ROS ) were assessed by the fluorescent indicator DCF-DA .\n\nThe protein expression of the endoplasmic reticulum ( ER ) stress pathways , GRP78 , XBP-1 , ATF-4 and CHOP , was examined with Western blotting .\n\nA growth inhibitory effect was observed after treatment with B19 in a dose-dependent manner and with more potential than curcumin .\n\nAt 20 \ufffdM , B19 induced significant apoptosis in CP70 cells .\n\nFurthermore , B19 induced the ER stress response , while curcumin had no effect on ER stress .\n\nThese results suggest that B19 has more effective antitumor properties than curcumin , and is associated with the activation of ER stress and ROS in ovarian cancer cells .", "output": "Resisting cell death" }, { "input": "Human carcinomas are defined by recurrent chromosomal aneuploidies , which result in a tissue-specific distribution of genomic imbalances .\n\nIn order to develop models for these genome mutations and to determine their role in tumorigenesis , we generated 45 spontaneously transformed murine cell lines from normal epithelial cells derived from bladder , cervix , colon , kidney , lung , and mammary gland .\n\nPhenotypic changes , chromosomal aberrations , centrosome number , and telomerase activity were assayed in control uncultured cells and in three subsequent stages of transformation .\n\nSupernumerary centrosomes , binucleate cells , and tetraploidy were observed as early as 48 hr after explantation .\n\nIn addition , telomerase activity increased throughout progression .\n\nLive-cell imaging revealed that failure of cytokinesis , not cell fusion , promoted genome duplication .\n\nSpectral karyotyping demonstrated that aneuploidy preceded immortalization , consisting predominantly of whole chromosome losses ( 4 , 9 , 12 , 13 , 16 , and Y ) and gains ( 1 , 10 , 15 , and 19 ) .\n\nAfter transformation , focal amplifications of the oncogenes Myc and Mdm2 were frequently detected .\n\nFifty percent of the transformed lines resulted in tumors on injection into immunocompromised mice .\n\nThe phenotypic and genomic alterations observed in spontaneously transformed murine epithelial cells recapitulated the aberration pattern observed during human carcinogenesis .\n\nThe dominant aberration of these cell lines was the presence of specific chromosomal aneuploidies .\n\nWe propose that our newly derived cancer models will be useful tools to dissect the sequential steps of genome mutations during malignant transformation , and also to identify cancer-specific genes , signaling pathways , and the role of chromosomal instability in this process .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Epidermal growth factor receptor-tyrosine kinase inhibitors ( EGFR-TKIs ) show dramatic antitumor activity in a subset of patients with non-small cell lung cancer who have an active mutation in the epidermal growth factor receptor ( EGFR ) gene .\n\nOn the other hand , some lung cancer patients with wild type EGFR also respond to EGFR-TKIs , suggesting that EGFR-TKIs have an effect on host cells as well as tumor cells .\n\nHowever , the effect of EGFR-TKIs on host microenvironments is largely unknown .\n\nA multiple organ metastasis model was previously established in natural killer cell-depleted severe combined immunodeficient mice using human lung cancer cells .\n\nThis model was used to investigate the therapeutic efficacy of erlotinib , an EGFR-TKI , on multiple organ metastases induced by human small cell lung cancer cells ( SBC-5 cells ) that did not express EGFR .\n\nAlthough erlotinib did not have any effect on the proliferation of SBC-5 cells in vitro , it significantly suppressed bone and lung metastases in vivo , but not liver metastases .\n\nAn immunohistochemical analysis revealed that , erlotinib significantly suppressed the number of osteoclasts in bone metastases , whereas no difference was seen in microvessel density .\n\nMoreover , erlotinib inhibited EGF-induced receptor activator of nuclear factor kappa-B expression in an osteoblastic cell line ( MC3T3-E1 cells ) .\n\nThese results strongly suggested that erlotinib prevented bone metastases by affecting host microenvironments irrespective of its direct effect on tumor cells .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Heat shock protein 27 ( Hsp27 ) is emerging as a promising therapeutic target for treatment of various cancers .\n\nAlthough the role of Hsp27 in protection from stress-induced intrinsic cell death has been relatively well studied , its role in Fas ( death domain containing member of the tumor necrosis factor receptor superfamily)-induced apoptosis and cell proliferation remains underappreciated .\n\nHere , we show that Hsp27 silencing induces dual coordinated effects , resulting in inhibition of cell proliferation and sensitization of cells to Fas-induced apoptosis through regulation of PEA-15 ( 15-kDa phospho-enriched protein in astrocytes ) .\n\nWe demonstrate that Hsp27 silencing suppresses proliferation by causing PEA-15 to bind and sequester extracellular signal-regulated kinase ( ERK ) , resulting in reduced translocation of ERK to the nucleus .\n\nConcurrently , Hsp27 silencing promotes Fas-induced apoptosis by inducing PEA-15 to release Fas-associating protein with a novel death domain ( FADD ) , thus allowing FADD to participate in death receptor signaling .\n\nConversely , Hsp27 overexpression promotes cell proliferation and suppresses Fas-induced apoptosis .\n\nFurthermore , we show that Hsp27 regulation of PEA-15 activity occurs in an Akt-dependent manner .\n\nSignificantly , Hsp27 silencing in a panel of phosphatase and tensin homolog on chromosome 10 ( PTEN ) wild-type or null cell lines , and in LNCaP cells that inducibly express PTEN , resulted in selective growth inhibition of PTEN-deficient cancer cells .\n\nThese data identify a dual coordinated role of Hsp27 in cell proliferation and Fas-induced apoptosis via Akt and PEA-15 , and indicate that improved clinical responses to Hsp27-targeted therapy may be achieved by stratifying patient populations based on tumor PTEN expression .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "A distinct group of breast cancers , called \" basal \" or \" triple-negative \" ( TN ) cancers express both basal cytokeratins and the epidermal growth factor receptor , but fail to express estrogen receptors , progesterone receptors or HER2 and have stem-like or mesenchymal features .\n\nThey are particularly aggressive , are frequently chemo-resistant , with p53 mutation , up-regulation of IL-6 and Stat3 .\n\nBecause TN cells are particularly sensitive to the anti-diabetic agent metformin , we hypothesized that it may target JAK2/Stat3 signaling .\n\nThe effects of metformin upon Stat3 expression and activation were examined in four human TN cell lines .\n\nMetformin's effects were also studied in sublines with forced over-expression of constitutively active ( CA ) Stat3 , as well as lines with stable knockdown of Stat3 .\n\nMetformin inhibited Stat3 activation ( P-Stat3 ) at Tyr705 and Ser727 and downstream signaling in each of the four parental cell lines .\n\nCA-Stat3 transfection attenuated , whereas Stat3 knockdown enhanced , the effects of metformin upon growth inhibition and apoptosis induction .\n\nA Stat3 specific inhibitor acted synergistically with metformin in reducing cell growth and inducing apoptosis .\n\nAn mTOR inhibitor showed no significant interaction with metformin .\n\nIn summary , Stat3 is a critical regulator of metformin action in TN cancer cells , providing the potential for enhancing metformin's efficacy in the clinical setting .", "output": "Resisting cell death" }, { "input": "Recent evidences suggest that the activity of glycogen synthase kinase-3 ( GSK3 ) contributes to the tumorigenic potential of pancreatic cancer cells through modulation of cell proliferation and survival .\n\nHowever , further investigations are needed to identify GSK3-dependent mechanisms involved in the control of pancreatic cancer cell proliferation and survival .\n\nThis study was undertaken to provide further support for a role of GSK3 in pancreatic cancer cell growth as well as to identify new cellular and molecular mechanisms involved .\n\nHerein , we demonstrate that prolonged inhibition of GSK3 triggers an apoptotic response only in human pancreatic cancer cells but not in human non-transformed pancreatic epithelial cells .\n\nWe show that prolonged inhibition of GSK3 activity increases Bim messenger RNA and protein expressions .\n\nMoreover , we provide evidence that activation of the c-jun N-terminal kinase ( JNK ) pathway is necessary for the GSK3 inhibition-mediated increase in Bim expression and apoptotic response .\n\nFinally , we demonstrate that concomitant inhibition of GSK3 potentiates the death ligand-induced apoptotic response in pancreatic cancer cells but not in non-transformed pancreatic epithelial cells and that this effect also requires JNK activity .\n\nConsidering that different approaches leading to stimulation of death receptor signaling are under clinical trials for treatment of unresectable or metastatic pancreatic cancer , inhibition of GSK3 could represent an attractive new avenue to improve their effectiveness .", "output": "Resisting cell death" }, { "input": "The innate immune response involves a variety of inflammatory reactions that can result in inflammatory disease and cancer if they are not resolved and instead are allowed to persist .\n\nThe effective activation and resolution of innate immune responses relies on the production and posttranscriptional regulation of mRNAs encoding inflammatory effector proteins .\n\nThe RNA-binding protein HuR binds to and regulates such mRNAs , but its exact role in inflammation remains unclear .\n\nHere we show that HuR maintains inflammatory homeostasis by controlling macrophage plasticity and migration .\n\nMice lacking HuR in myeloid-lineage cells , which include many of the cells of the innate immune system , displayed enhanced sensitivity to endotoxemia , rapid progression of chemical-induced colitis , and severe susceptibility to colitis-associated cancer .\n\nThe myeloid cell-specific HuR-deficient mice had an exacerbated inflammatory cytokine profile and showed enhanced CCR2-mediated macrophage chemotaxis .\n\nAt the molecular level , activated macrophages from these mice showed enhancements in the use of inflammatory mRNAs ( including Tnf , Tgfb , Il10 , Ccr2 , and Ccl2 ) due to a lack of inhibitory effects on their inducible translation and/or stability .\n\nConversely , myeloid overexpression of HuR induced posttranscriptional silencing , reduced inflammatory profiles , and protected mice from colitis and cancer .\n\nOur results highlight the role of HuR as a homeostatic coordinator of mRNAs that encode molecules that guide innate inflammatory effects and demonstrate the potential of harnessing the effects of HuR for clinical benefit against pathologic inflammation and cancer .", "output": "Tumor promoting inflammation" }, { "input": "Hormone-dependent estrogen receptor positive ( ER+ ) breast cancers generally respond well to anti-estrogen therapy .\n\nUnfortunately , hormone-independent estrogen receptor negative ( ER- ) breast cancers are aggressive , respond poorly to current treatments and have a poor prognosis .\n\nNew approaches and targets are needed for the prevention and treatment of ER- breast cancer .\n\nThe NF-\u03baB signaling pathway is strongly implicated in ER- tumor genesis , constituting a possible target for treatment .\n\nHydrogen sulfide-releasing aspirin ( HS-ASA ) , a novel and safer derivative of aspirin , has shown promise as an anti-cancer agent .\n\nWe examined the growth inhibitory effect of HS-ASA via alterations in cell proliferation , cell cycle phase transitions , and apoptosis , using MDA-MB-231 cells as a model of triple negative breast cancer .\n\nTumor xenografts in mice , representing human ER- breast cancer , were evaluated for reduction in tumor size , followed by immunohistochemical analysis for proliferation , apoptosis and expression of NF-\u03baB .\n\nHS-ASA suppressed the growth of MDA-MB-231 cells by induction of G(0)/G(1) arrest and apoptosis , down-regulation of NF-\u03baB , reduction of thioredoxin reductase activity , and increased levels reactive oxygen species .\n\nTumor xenografts in mice , were significantly reduced in volume and mass by HS-ASA treatment .\n\nThe decrease in tumor mass was associated with inhibition of cell proliferation , induction of apoptosis and decrease in NF-\u03baB levels in vivo .\n\nHS-ASA has anti-cancer potential against ER- breast cancer and merits further study .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Resisting cell death" }, { "input": "Transcriptional coactivator amplified in breast cancer 1 ( AIB1 ) plays important roles in the progression of several cancers such as prostate cancer , breast cancer , and hepatocellular carcinoma .\n\nHowever , its role in cholangiocarcinoma ( CCA ) , a chemoresistant bile duct carcinoma with a poor prognosis , remains unclear .\n\nIn this study we found that AIB1 protein was frequently overexpressed in human CCA specimens and CCA cell lines .\n\nDown-regulation of AIB1 induced the G2/M arrest and decreased the expression of mitosis-promoting factors including Cyclin A , Cyclin B , and Cdk1 through suppressing the Akt pathway , which resulted in inhibiting CCA cell proliferation .\n\nIn addition , AIB1 enhanced the chemoresistance of CCA cells at least in part through up-regulating the expression of antiapoptotic protein Bcl-2 .\n\nAIB1 regulated the expression of Bcl-2 in CCA cells through activating the Akt pathway as well as suppressing intracellular reactive oxygen species ( ROS ) .\n\nAIB1 suppressed ROS by up-regulating antioxidants such as glutathione synthetase and glutathione peroxidase , which are targets of the NF-E2-related factor 2 ( Nrf2 ) , a critical transcription factor that regulates antioxidants , detoxification enzymes , and drug efflux proteins .\n\nAIB1 also increased the expression of another two Nrf2 targets , ABCC2 and ABCG2 , to enhance drug efflux .\n\nAIB1 served as an essential coactivator for Nrf2 activation by physically interacting with Nrf2 to enhance its transcriptional activity .\n\nConclusion : AIB1 plays an important role in proliferation and chemoresistance of CCA through simultaneous activation of Akt and Nrf2 pathways , suggesting that AIB1 is a potential molecular target for CCA treatment .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND The receptor tyrosine kinase family includes many transmembrane proteins with diverse physiological and pathophysiological functions .\n\nThe involvement of tyrosine kinase signaling in promoting a more aggressive tumor phenotype within the context of chemotherapeutic evasion is gaining recognition .\n\nThe Ron receptor is a tyrosine kinase receptor that has been implicated in the progression of breast cancer and evasion of tamoxifen therapy .\n\nRESULTS Here , we report that Ron expression is correlated with in situ , estrogen receptor alpha ( ER\u03b1)-positive tumors , and is higher in breast tumors following neoadjuvant tamoxifen therapy .\n\nWe also demonstrate that the majority of mammary tumors isolated from transgenic mice with mammary specific-Ron overexpression ( MMTV-Ron mice ) , exhibit appreciable ER expression .\n\nMoreover , genetic-ablation of ER\u03b1 , in the context of Ron overexpression , leads to delayed mammary tumor initiation and growth , but also results in an increased metastasis .\n\nCONCLUSIONS Ron receptor overexpression is associated with ER\u03b1-positive human and murine breast tumors .\n\nIn addition , loss of ER\u03b1 on a Ron overexpressing background in mice leads to the development of breast tumors which grow slower but which exhibit more metastasis and suggests that targeting of ER\u03b1 , as in the case of tamoxifen therapy , may reduce the growth of Ron overexpressing breast cancers but may cause these tumors to be more metastatic .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Chronic myelogenous leukemia ( CML ) results from a chromosomal translocation in hematopoietic stem or early progenitor cells that gives rise to the oncogenic BCR/ABL fusion protein .\n\nClinically , CML has a chronic phase that eventually evolves into an accelerated stage and blast crisis .\n\nA CML-specific immune response is thought to contribute to the control of disease .\n\nWhether the immune system can also promote disease progression is not known .\n\nIn the present study , we investigated the possibility that the TNF receptor family member CD27 is present on leukemia stem cells ( LSCs ) and mediates effects of the immune system on CML .\n\nIn a mouse model of CML , BCR/ABL+ LSCs and leukemia progenitor cells were found to express CD27 .\n\nBinding of CD27 by its ligand , CD70 , increased expression of Wnt target genes in LSCs by enhancing nuclear localization of active \u03b2-catenin and TRAF2- and NCK-interacting kinase ( TNIK ) .\n\nThis resulted in increased proliferation and differentiation of LSCs .\n\nBlocking CD27 signaling in LSCs delayed disease progression and prolonged survival .\n\nFurthermore , CD27 was expressed on CML stem/progenitor cells in the bone marrow of CML patients , and CD27 signaling promoted growth of BCR/ABL+ human leukemia cells by activating the Wnt pathway .\n\nSince expression of CD70 is limited to activated lymphocytes and dendritic cells , our results reveal a mechanism by which adaptive immunity contributes to leukemia progression .\n\nIn addition , targeting CD27 on LSCs may represent an attractive therapeutic approach to blocking the Wnt/\u03b2-catenin pathway in CML .", "output": "Tumor promoting inflammation" }, { "input": "The DNA damage response ( DDR ) cascade and ROS ( reactive oxygen species ) signaling are both involved in the induction of cell death after DNA damage , but a mechanistic link between these two pathways has not been clearly elucidated .\n\nThis study demonstrates that ROS induction after treatment of cells with neocarzinostatin ( NCS ) , an ionizing radiation mimetic , is at least partly mediated by increasing histone H2AX .\n\nIncreased levels of ROS and cell death induced by H2AX overexpression alone or DNA damage leading to H2AX accumulation are reduced by treating cells with the antioxidant N-Acetyl-L-Cysteine ( NAC ) , the NADP(H) oxidase ( Nox ) inhibitor DPI , expression of Rac1N17 , and knockdown of Nox1 , but not Nox4 , indicating that induction of ROS by H2AX is mediated through Nox1 and Rac1 GTPase .\n\nH2AX increases Nox1 activity partly by reducing the interaction between a Nox1 activator NOXA1 and its inhibitor 14-3-3zeta .\n\nThese results point to a novel role of histone H2AX that regulates Nox1-mediated ROS generation after DNA damage .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Hexachlorobenzene ( HCB ) is an organochlorine pesticide that acts as an endocrine disruptor in humans and rodents .\n\nThe development of breast cancer strongly depends on endocrine conditions modulated by environmental factors .\n\nWe have demonstrated that HCB is a tumor co-carcinogen in rats and an inducer of proliferation in MCF-7 cells , in an estrogen receptor \u03b1 ( ER\u03b1)-dependent manner , and of migration in MDA-MB-231 breast cancer cell line .\n\nIn the present study , we examined HCB effect on c-Src/human epidermal growth factor receptor ( HER1 ) and ER\u03b1 signaling pathways in mammary glands and in N-nitroso-N-methylurea ( NMU)-induced mammary tumors in rats .\n\nFurthermore , we evaluated histopathological changes and serum hormone levels .\n\nRats were separated into four groups : control , HCB ( 100 mg/kg b.w. ) , NMU ( 50 mg/kg b.w. ) and NMU-HCB .\n\nOur data show that HCB increases c-Src and HER1 activation , c-Src/HER1 association , and Y699-STAT5b and ERK1/2 phosphorylation in mammary glands .\n\nHCB also enhances Y537-ER\u03b1 phosphorylation and ER\u03b1/c-Src physical interaction .\n\nIn tumors , HCB also induces c-Src and HER1 activation , c-Src/HER1 association , as well as T308-Akt and Y699-STAT5b phosphorylation .\n\nIn addition , the pesticide increases ER\u03b1 protein content and decreases p-Y537-ER\u03b1 levels and ER\u03b1/c-Src association in tumors .\n\nHCB increases serum 17-beta estradiol and prolactin contents and decreases progesterone , FSH and LH levels in rats without tumors , while the opposite effect was observed in rats with tumors .\n\nTaken together , our results indicate that HCB induces an estrogenic effect in mammary gland , increasing c-Src/HER1 and ER\u03b1 signaling pathways .\n\nHCB stimulates c-Src/HER1 pathway , but decreases ER\u03b1 activity in tumors , appearing to shift them towards a higher malignancy phenotype .", "output": "Sustaining proliferative signaling" }, { "input": "DNA repair is essential in maintaining genome integrity and defects in different steps of the process have been linked to cancer and aging .\n\nIt is a long lasting question how DNA repair is spatially and temporarily organized in the highly compartmentalized nucleus and whether the diverse nuclear compartments regulate differently the efficiency of repair .\n\nIncreasing evidence suggest the involvement of nuclear pore complexes in repair of double-strand breaks ( DSBs ) in yeast .\n\nHere , we show that the human nucleoporin 153 ( NUP153 ) has a role in repair of DSBs and in the activation of DNA damage checkpoints .\n\nWe explore the mechanism of action of NUP153 and we propose its potential as a novel therapeutic target in cancers .", "output": "Genomic instability and mutation" }, { "input": "Imiquimod is a synthetic compound with antitumor properties ; a 5% cream formulation is successfully used to treat skin tumors .\n\nThe antitumor effect of imiquimod is multifactorial , although its ability to modulate immune responses by triggering TLR7/8 is thought to be key .\n\nAmong the immune cells suggested to be involved are plasmacytoid DCs ( pDCs ) .\n\nHowever , a direct contribution of pDCs to tumor killing in vivo and the mechanism of their recruitment to imiquimod-treated sites have never been demonstrated .\n\nUsing a mouse model of melanoma , we have now demonstrated that pDCs can directly clear tumors without the need for the adaptive immune system .\n\nTopical imiquimod treatment led to TLR7-dependent and IFN-\u03b1/\u03b2 receptor 1-dependent ( IFNAR1-dependent ) upregulation of expression of the chemokine CCL2 in mast cells .\n\nThis was essential to induce skin inflammation and for the recruitment of pDCs to the skin .\n\nThe recruited pDCs were CD8\u03b1+ and induced tumor regression in a TLR7/MyD88- and IFNAR1-dependent manner .\n\nLack of TLR7 and IFNAR1 or depletion of pDCs or CD8\u03b1+ cells from tumor-bearing mice completely abolished the effect of imiquimod .\n\nTLR7 was essential for imiquimod-stimulated pDCs to produce IFN-\u03b1/\u03b2 , which led to TRAIL and granzyme B secretion by pDCs via IFNAR1 signaling .\n\nBlocking these cytolytic molecules impaired pDC-mediated tumor killing .\n\nOur results demonstrate that imiquimod treatment leads to CCL2-dependent recruitment of pDCs and their transformation into a subset of killer DCs able to directly eliminate tumor cells .", "output": "Tumor promoting inflammation" }, { "input": "Breast cancer is the most common malignancy in women , and many breast cancer patients fail conventional treatment strategies of chemotherapy , radiation , and antiestrogen therapy .\n\nResearch into the molecular pathways and biomarkers involved in the development of breast cancer should yield information that will guide therapeutic decisions .\n\nEpidermal growth factor receptor ( EGFR ) and cyclooxygenase-2 ( COX-2 ) are involved in the carcinogenesis of breast cancer and exist tight crosstalk with estrogen receptor ( ER ) pathway .\n\nCombination of EGFR and COX-2 inhibitors , therefore , could be an effective strategy for reducing cell growth in estrogen-dependent breast cancer .\n\nIn order to verify the effects of EGFR and COX-2 inhibitors , breast cancer cells MCF-7 and SKBR-3 were characterized for receptors status and then treated with respective inhibitors ( nimotuzumab and celecoxib ) alone and in combination .\n\nBoth cell lines were sensitive to celecoxib , but not to nimotuzumab .\n\nHowever , combination of two drugs demonstrated synergistic effects on cell killing .\n\nMoreover , association of two drugs resulted in SKBR-3 cells , a further G0/G1 phase arrest than one drug alone .\n\nDownregulation of p-EGFR , p-Akt , p-mTOR , and amplified in breast cancer 1 ( AIB1 ) were observed in both cell lines , and upregulation of E-cadherin was only found in MCF-7 , after treatment with single agent or in combination .\n\nThese studies suggest that nimotuzumab and celecoxib exert synergistic antiproliferation effects in breast cancer , which partly correlates with ER status .\n\nDue to Akt/mTOR , EMT and AIB1 pathways participate in this process , therefore , E-cadherin and AIB1 may be considered as possible biomarkers to predict response in ER-positive breast cancer cells treated with EGFR and COX-2 inhibitors .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "OBJECTIVE : Emerging evidences implicate long noncoding RNAs ( lncRNAs ) are deregulated in cancer development .\n\nThe purpose of the current study is to investigate the role of new lncRNA , named PlncRNA-1 , in prostate cancer ( CaP ) pathogenesis .\n\nMATERIALS AND METHODS : In this study , real-time q-PCR was used to demonstrate the expression of PlncRNA-1 in 16 pairs CaP tissues and matched normal tissues , 14 pairs CaP tissues and BPH tissues , 4 CaP cell lines , including LNCaP , LNCaP-AI , PC3 , and C4-2 , and 2 normal prostate epithelial cell lines RWPE-1 and PWR-1E .\n\nAfter PlncRNA-1 was suppressed by siRNA in LNCaP and LNCaP-AI cell lines , cell proliferation and apoptosis were assessed using CCK-8 and terminal deoxynucleotidyl transferase dUTP nick end labeling ( TUNEL ) .\n\nAfter PlncRNA-1 and AR was suppressed by siRNA in LNCaP and LNCaP-AI cell lines , real-time q-PCR and Western blotting were used to measure reciprocal regulation of PlncRNA-1 and AR .\n\nRESULTS : We showed that expression PlncRNA-1 , was significantly higher in CaP cells relative to normal prostate epithelial cells , as well as higher in human CaPs compared with normal tissues and benign prostatic hyperplasia ( BPH ) .\n\nSilencing of PlncRNA-1 significantly reduced cell proliferation and induced apoptosis in CaP cell lines LNCaP and LNCaP-AI .\n\nMechanistically , PlncRNA-1 suppression by siRNA resulted in a decrease of androgen receptor ( AR ) mRNA , protein and AR downstream target .\n\nOf note , blockade of AR signaling with siRNA also resulted in a suppression of PlncRNA-1 expression in CaP cell lines .\n\nCONCLUSIONS : Our study suggests reciprocal regulation of PlncRNA-1 and androgen receptor contribute to CaP pathogenesis and that PlncRNA-1 is a potential therapy target .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "The ATM kinase and p53 are key tumor suppressor factors that control the genotoxic stress response pathway .\n\nThe ATM substrate Mdm2 controls p53 activity by either targeting p53 for degradation or promoting its synthesis by binding the p53 mRNA .\n\nThe physiological role and regulation of Mdm2's dual function toward p53 is not known .\n\nHere we show that ATM-dependent phosphorylation of Mdm2 at Ser395 is required for the p53 mRNA-Mdm2 interaction .\n\nThis event also promotes SUMO-conjugation of Mdm2 and its nucleoli accumulation .\n\nInterfering with the p53 mRNA-Mdm2 interaction prevents p53 stabilization and activation following DNA damage .\n\nThese results demonstrate how ATM activity switches Mdm2 from a negative to a positive regulator of p53 via the p53 mRNA .", "output": "Genomic instability and mutation" }, { "input": "The Ras association domain family 1 isoform A ( RASSF1A ) is a tumor suppressor whose inactivation is implicated in the development of many human cancers , including breast carcinomas .\n\nLittle is known about the tumor-suppressive function of RASSF1A in breast tissue and whether its inactivation is mechanistically involved in the initiation and progression of breast tumors .\n\nHere , we show that RASSF1A inhibits breast cancer growth in vivo , and suppresses estrogen receptor ( ER\u03b1 ) expression and function .\n\nReconstitution of RASSF1A in MCF7 cells led to decreased ER\u03b1 levels and reduced sensitivity to estrogen ( E2 ) .\n\nConcomitantly , we observed decreased expression of Id1 as well as the E2-responsive genes Bcl-2 and c-Myc that cooperatively contribute to the immortalization and transformation of breast epithelial cells .\n\nThis downregulation was associated with induction of cell-cycle arrest and senescence that constitute early barriers to cancer initiation and progression .\n\nKnockdown of ER\u03b1 showed that downregulation of ER\u03b1 suffices to increase senescence and inhibit expression of Bcl-2 , c-Myc and Id1 .\n\nHowever , enforced expression of ER\u03b1 only partially rescued RASSF1A-mediated growth inhibition and senescence , suggesting that suppression of ER\u03b1 expression and activity is not the only mechanism by which RASSF1A inhibits growth and survival of breast cancer cells .\n\nEctopic expression of Bcl-2 , c-Myc and Id1 had little or no effect on RASSF1A-mediated growth arrest , indicating that RASSF1A acts dominantly over these oncogenes .\n\nMechanistically , RASSF1A was found to suppress ER\u03b1 expression through Akt1 .\n\nIt also transiently inhibited ER\u03b1-induced Ras-MAPK activity after exposure of cells to E2 .\n\nTogether , our data show that RASSF1A acts as a tumor suppressor in ER\u03b1+ mammary epithelial cells , in part through inhibiting ER\u03b1 expression and activity .\n\nThese findings suggest that RASSF1A has a key role in suppressing the transformation of human breast epithelial cells and ER\u03b1+ breast cancer initiation .", "output": "Sustaining proliferative signaling, Enabling replicative immortality" }, { "input": "BACKGROUND Luteolin is a 3',4',5,7-tetrahydroxyflavone found in various fruits and vegetables .\n\nWe have shown previously that luteolin reduces HT-29 cell growth by inducing apoptosis and cell cycle arrest .\n\nThe objective of this study was to examine whether luteolin downregulates the insulin-like growth factor-I receptor ( IGF-IR ) signaling pathway in HT-29 cells .\n\nMETHODS In order to assess the effects of luteolin and/or IGF-I on the IGF-IR signaling pathway , cells were cultured with or without 60 \u03bcmol/L luteolin and/or 10 nmol/L IGF-I .\n\nCell proliferation , DNA synthesis , and IGF-IR mRNA levels were evaluated by a cell viability assay , [ 3H]thymidine incorporation assays , and real-time polymerase chain reaction , respectively .\n\nWestern blot analyses , immunoprecipitation , and in vitro kinase assays were conducted to evaluate the secretion of IGF-II , the protein expression and activation of IGF-IR , and the association of the p85 subunit of phophatidylinositol-3 kinase ( PI3K ) with IGF-IR , the phosphorylation of Akt and extracellular signal-regulated kinase ( ERK)1/2 , and cell division cycle 25c ( CDC25c ) , and PI3K activity .\n\nRESULTS Luteolin ( 0 - 60 \u03bcmol/L ) dose-dependently reduced the IGF-II secretion of HT-29 cells .\n\nIGF-I stimulated HT-29 cell growth but did not abrogate luteolin-induced growth inhibition .\n\nLuteolin reduced the levels of the IGF-IR precursor protein and IGF-IR transcripts .\n\nLuteolin reduced the IGF-I-induced tyrosine phosphorylation of IGF-IR and the association of p85 with IGF-IR .\n\nAdditionally , luteolin inhibited the activity of PI3K activity as well as the phosphorylation of Akt , ERK1/2 , and CDC25c in the presence and absence of IGF-I stimulation .\n\nCONCLUSIONS The present results demonstrate that luteolin downregulates the activation of the PI3K/Akt and ERK1/2 pathways via a reduction in IGF-IR signaling in HT-29 cells ; this may be one of the mechanisms responsible for the observed luteolin-induced apoptosis and cell cycle arrest .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Curcumin ( CUR ; diferuloylmethane ) , a rhizome extract of Curcuma Longa L. is commonly used as a food coloring and flavoring agent .\n\nAlthough oriental and Ayurvedic medicines have traditionally used CUR in the treatment of diseases , conventional medicine has just begun to recognize its potential therapeutic value .\n\nNumerous recent studies have demonstrated the ability of CUR to halt or prevent certain types of cancer , decrease inflammation , and improve cardiovascular health .\n\nHowever , very few studies have examined its ability to protect against drug-induced organ injury .\n\nThis study explored whether CUR pre-exposure has the potential to prevent acetaminophen ( APAP)-induced : ( i ) hepatotoxicity , ( ii ) genomic injury , ( iii ) oxidative stress in the liver , and ( iv ) apoptotic and necrotic cell deaths in the liver in vivo .\n\nAdditional goals were to investigate the interplay of pro- and anti-apoptotic genes and their ultimate impact on various forms of cell death .\n\nIn order to study the CUR-APAP interaction , male B6C3F1 mice were gavaged with CUR ( 17 mg/kg/day , p.o. ) for 12 days followed by a single APAP exposure ( 400 mg/kg , ip ) .\n\nFour groups of animals ( control , CUR , APAP , CUR+APAP ) were sacrificed 24 h after APAP exposure .\n\nThe results indicated that APAP-induced liver injury associated events as serum ALT ( 80-fold ) , lipid peroxidation ( 357% ) and DNA fragmentation ( 469% ) were markedly reduced to 3-fold , 134% and 162% , respectively , in the CUR+APAP group .\n\nThe APAP-induced increase in expression of pro-apoptotic genes ( Bax , caspase-3 ) decreased while expression of anti-apoptotic genes ( Bcl-XL ) increased in CUR preexposed mouse livers , and these changes were mirrored in the pattern of apoptotic and necrotic cell deaths .\n\nLevels of DNA damage sensor P\u2075\u00b3 and its counterpart Mdm2 were also analyzed during this interaction .\n\nBased on the available literature , and these results , it seems likely that CUR may impart global protection in vivo against drug-induced liver injury by opposing several crucial events instrumental to both apoptosis and necrosis .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Cancer cells universally increase glucose and glutamine consumption , leading to the altered metabolic state known as the Warburg effect ; one metabolic pathway , highly dependent on glucose and glutamine , is the hexosamine biosynthetic pathway .\n\nIncreased flux through the hexosamine biosynthetic pathway leads to increases in the post-translational addition of O-linked \u03b2-N-acetylglucosamine ( O-GlcNAc ) to various nuclear and cytosolic proteins .\n\nA number of these target proteins are implicated in cancer , and recently , O-GlcNAcylation was shown to play a role in breast cancer ; however , O-GlcNAcylation in other cancers remains poorly defined .\n\nHere , we show that O-GlcNAc transferase ( OGT ) is overexpressed in prostate cancer compared with normal prostate epithelium and that OGT protein and O-GlcNAc levels are elevated in prostate carcinoma cell lines .\n\nReducing O-GlcNAcylation in PC3-ML cells was associated with reduced expression of matrix metalloproteinase ( MMP)-2 , MMP-9 , and VEGF , resulting in inhibition of invasion and angiogenesis .\n\nOGT-mediated regulation of invasion and angiogenesis was dependent upon regulation of the oncogenic transcription factor FoxM1 , a key regulator of invasion and angiogenesis , as reducing OGT expression led to increased FoxM1 protein degradation .\n\nConversely , overexpression of a degradation-resistant FoxM1 mutant abrogated OGT RNAi-mediated effects on invasion , MMP levels , angiogenesis , and VEGF expression .\n\nUsing a mouse model of metastasis , we found that reduction of OGT expression blocked bone metastasis .\n\nAltogether , these data suggest that as prostate cancer cells alter glucose and glutamine levels , O-GlcNAc modifications and OGT levels become elevated and are required for regulation of malignant properties , implicating OGT as a novel therapeutic target in the treatment of cancer .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "It is prevailingly thought that the antiestrogens tamoxifen and ICI 182 , 780 are competitive antagonists of the estrogen-binding site of the estrogen receptor-alpha ( ER-\u03b1 ) .\n\nHowever , a plethora of evidence demonstrated both antiestrogens exhibit agonist activities in different systems such as activation of the membrane-initiated signaling pathways .\n\nThe mechanisms by which antiestrogens mediate estrogen-like activities have not been fully established .\n\nPreviously , a variant of ER-\u03b1 , EP-\u03b136 , has been cloned and showed to mediate membrane-initiated estrogen and antiestrogen signaling in cells only expressing ER-\u03b136 .\n\nHere , we investigated the molecular mechanisms underlying the antiestrogen signaling in ER-negative breast cancer MDA-MB-231 and MDA-MB-436 cells that express high levels of endogenous ER-\u03b136 .\n\nWe found that the effects of both 4-hydoxytamoxifen ( 4-OHT ) and ICI 182 , 780 ( ICI ) exhibited a non-monotonic , or biphasic dose response curve ; antiestrogens at low concentrations , elicited a mitogenic signaling pathway to stimulate cell proliferation while at high concentrations , antiestrogens inhibited cell growth .\n\nAntiestrogens at l nM induced the phosphorylation of the Src-Y416 residue , an event to activate Src , while at 5 \ufffdM induced Src-Y527 phosphorylation that inactivates Src .\n\nAntiestrogens at 1 nM also induced phosphorylation of the MAPK/ERK and activated the Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathways but not at 5 \ufffdM .\n\nKnock-down of ER-\u03b136 abrogated the biphasic antiestrogen signaling in these cells .\n\nOur results thus indicated that ER-\u03b136 mediates biphasic antiestrogen signaling in the ER-negative breast cancer cells and Src functions as a switch of antiestrogen signaling dependent on concentrations of antiestrogens through the EGFR/STAT5 pathway .", "output": "Sustaining proliferative signaling" }, { "input": "The published results on nanoparticles cytotoxicity and genotoxicity such as titanium dioxide nanoparticles ( TiO(2) NPs ) are inconsistent , and often conflicting and insufficient .\n\nSince different parameters may have impact on the toxicity results , there is need to lay stress on detailed characterization of NPs and the use of different testing conditions for assessment of NPs toxicity .\n\nIn order to investigate whether dispersion procedures influence NP cytotoxicity and genotoxicity , we compared two protocols giving TiO(2) NP dispersions with different stability and agglomeration states .\n\nDetailed primary and secondary characteristics of both TiO(2) NP dispersions in culture media were carried out before toxicological testing ; TK6 human lymphoblast cells , EUE human embryonic epithelial cells and Cos-1 monkey kidney fibroblasts were used to assess cytotoxicity ( by trypan blue exclusion , proliferation activity and plating efficiency assays ) and genotoxicity ( by the comet assay ) .\n\nDNA strand breaks were detected by the alkaline comet assay .\n\nDNA oxidation lesions ( especially 8-oxo-7,8-dihydroguanine , 8-oxoG ) were measured with a modified comet assay including incubation with specific repair enzyme formamidopyrimidine DNA glycosylase ( FPG ) .\n\nThe TiO(2) NPs dispersion with large agglomerates ( 3 min sonication and no serum in stock solution ) induced DNA damage in all three cell lines , while the TiO(2) NPs dispersed with agglomerates less than 200 nm ( foetal serum in stock solution and sonication 15 min ) had no effect on genotoxicity .\n\nAn increased level of DNA oxidation lesions detected in Cos-1 and TK6 cells indicates that the leading mechanism by which TiO(2) NPs trigger genotoxicity is most likely oxidative stress .\n\nOur results show that the dispersion method used can influence the results of toxicity studies .\n\nTherefore at least two different dispersion procedures should be incorporated into assessment of cyto- and genotoxic effects of NPs .\n\nIt is important , when assessing the hazard associated with NPs , to establish standard testing procedures and thorough strategies to consider the diverse conditions relevant to possible exposures .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "The NF-kB family of transcription factors regulates important biological functions including cell growth , survival and the immune response .\n\nWe found that Human Papillomavirus type 16 ( HPV-16 ) E7 and E6/E7 proteins inhibited basal and TNF-alpha-inducible NF-kB activity in human epithelial cells cultured from the cervical transformation zone , the anatomic region where most cervical cancers develop .\n\nIn contrast , HPV-16 E6 regulated NF-kB in a cell type- and cell growth-dependent manner .\n\nNF-kB influenced immortalization of cervical cells by HPV16 .\n\nInhibition of NF-kB by an IkB alpha repressor mutant increased colony formation and immortalization by HPV-16 .\n\nIn contrast , activation of NF-kB by constitutive expression of p65 inhibited proliferation and immortalization .\n\nOur results suggest that inhibition of NF-kB by HPV-16 E6/E7 contributes to immortalization of cells from the cervical transformation zone .", "output": "Enabling replicative immortality" }, { "input": "High-grade gliomas ( HGG ) , are the most common aggressive brain tumours in adults .\n\nInhibitors targeting growth factor signalling pathways in glioma have shown a low clinical response rate .\n\nTo accurately evaluate response to targeted therapies further in vitro studies are necessary .\n\nGrowth factor pathway expression using epidermal growth factor receptor ( EGFR ) , mutant EGFR ( EGFRvIII ) , platelet derived growth factor receptor ( PDGFR ) , C-Kit and C-Abl together with phosphatase and tensin homolog ( PTEN ) expression and downstream activation of AKT and phosphorylated ribosomal protein S6 ( P70S6K ) was analysed in 26 primary glioma cultures treated with the tyrosine kinase inhibitors ( TKIs ) erlotinib , gefitinib and imatinib .\n\nResponse to TKIs was assessed using 50% inhibitory concentrations ( IC(50) ) .\n\nResponse for each culture was compared with the EGFR/PDGFR immunocytochemical pathway profile using hierarchical cluster analysis ( HCA ) and principal component analysis ( PCA ) .\n\nErlotinib response was not strongly associated with high expression of the growth factor pathway components .\n\nPTEN expression did not correlate with response to any of the three TKIs .\n\nIncreased EGFR expression was associated with gefitinib response ; increased PDGFR-\u03b1 expression was associated with imatinib response .\n\nThe results of this in vitro study suggest gefitinib and imatinib may have therapeutic potential in HGG tumours with a corresponding growth factor receptor expression profile .", "output": "Sustaining proliferative signaling" }, { "input": "Epidermal growth factor receptor ( EGFR ) and human epidermal growth factor receptor 2 ( HER2 ) amplification occurs in over 30% of esophageal carcinomas .\n\nCombination therapies with EGFR and HER2-targeting agents and cytotoxic agents are considered a potential therapeutic option for esophageal cancer .\n\nWe evaluated the antitumor effects of lapatinib , a dual tyrosine kinase inhibitor which simultaneously inhibits EGFR and HER2 , 5-fluorouracil ( 5-Fu ) alone and in combination on esophageal cancer cells .\n\nThe antiproliferative activity of lapatinib , 5-Fu and lapatinib plus 5-Fu was measured by MTT assay and the combination index ( CI ) values were calculated .\n\nAdditionally , cell cycle distribution of lapatinib alone and the combination with 5-Fu were detected by flow cytometry analysis .\n\nAnnexinV-FITC and propidium iodide stain were used for analyzing the apoptotic cells after cells were treated with either agent alone or in combination .\n\nThe EGFR and HER2 activated signaling pathways were monitored by western blotting .\n\nThe combination of lapatinib and 5-Fu synergistically inhibited cell proliferation and exhibited an enhanced proapoptotic effect on esophageal cancer cells .\n\nThe potentiation effect of combined treatment was associated with downregulation of EGFR and HER2 signaling pathways because data from western blot analysis showed that lapatinib in combination with 5-Fu markedly reduced the phosphorylation of EGFR and HER2 , and inhibited the activation of downstream signaling molecules , such as AKT and ERK .\n\nA significant G1 arrest was also observed in cell cycle analysis after exposing cells to lapatinib , however , combination with 5-Fu did not enhance G1 arrest .\n\nThese results indicate that the combination of the lapatinib and 5-Fu is a promising treatment option for esophageal carcinoma with HER2 amplification .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "p53 is a well-known transcription factor that controls cell cycle arrest and cell death in response to a wide range of stresses .\n\nMoreover , p53 regulates glucose metabolism and its mutation results in the metabolic switch to the Warburg effect found in cancer cells .\n\nNucleotide biosynthesis is also critical for cell proliferation and the cell division cycle .\n\nNonetheless , little is known about whether p53 regulates nucleotide biosynthesis .\n\nHere we demonstrated that p53-inducible microRNA-34a ( miR-34a ) repressed inosine 5'-monophosphate dehydrogenase ( IMPDH ) , a rate-limiting enzyme of de novo GTP biosynthesis .\n\nTreatment with anti-miR-34a inhibitor relieved the expression of IMPDH upon DNA damage .\n\nUltimately , miR-34a-mediated inhibition of IMPDH resulted in repressed activation of the GTP-dependent Ras signaling pathway .\n\nIn summary , we suggest that p53 has a novel function in regulating purine biosynthesis , aided by miR-34a-dependent IMPDH repression .", "output": "Genomic instability and mutation" }, { "input": "Various endocrine disrupting chemicals ( EDCs ) are exogenous compounds found in the environment and have the potential to interfere with the endocrine system and hormonal regulation .\n\nAmong EDCs , bisphenolA ( BPA ) and 1,1,1-trichloro-2,2-bis(4-methoxyphenol)-ethane [ methoxychlor ( MXC) ] have estrogenic activity resulting in a variety of dysfunctions in the E2-mediated response by binding to estrogen receptors ( ERs ) , causing human health problems such as abnormal reproduction and carcinogenesis .\n\nIn this study , we investigated the effects of BPA and MXC on cell proliferation facilitated by ER signaling in human breast cancer cells .\n\nMCF-7 cells are known to be ER\u03b1-positive and to be a highly E2-responsive cancer cell line ; these cells are , therefore , a useful invitro model for detecting estrogenic activity in response to EDCs .\n\nWe evaluated cancer cell proliferation following BPA and MXC treatment using an MTT assay .\n\nWe analyzed alterations in the expression of genes associated with the cell cycle in MCF-7 cells by semi-quantitative reverse-transcription PCR following treatment with BPA or MXC compared to EtOH .\n\nTo determine whether BPA and MXC stimulate cancer cell growth though ER signaling , we co-treated the cells with agonists ( propyl pyrazoletriol , PPT ; and diarylpropionitrile , DPN ) or an antagonist ( ICI 182,780 ) of ER signaling and reduced ER\u03b1 gene expression via siRNA in MCF-7 cells before treatment with EDCs .\n\nThese studies confirmed the carcinogenicity of EDCs invitro .\n\nAs a result , BPA and MXC induced the cancer cell proliferation by the upregulation of genes that promote the cell cycle and the downregulation of anti-proliferative genes , especially ones affecting the G1/S transition via ER\u03b1 signaling .\n\nThese collective results confirm the carcinogenicity of these EDCs invitro .\n\nFurther studies are required to determine whether EDCs promote carcinogenesis invivo .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Epithelial mesenchymal transition ( EMT ) is known to be associated with chemoresistance as well as increased invasion/metastasis .\n\nHowever , the relationship between EMT and resistance to an epidermal growth factor receptor ( EGFR ) -targeting drug in head and neck squamous cell carcinoma ( HNSCC ) remains unknown .\n\nIn this study , we investigated the acquisition of EMT by gefitinib in HNSCC cell line ( UMSCC81B ) .\n\nMETHODS We isolated fibroblastoid variant ( 81B-Fb ) from gefitinib-resistant UMSCC81B-GR3 cells obtained after increasing the doses of gefitinib treatment in vitro and examined EMT and its underlying mechanism .\n\nRESULT 81B-Fb cells exhibited fibroblast-like morphology , increased motility , loss of E-cadherin , acquisition of vimentin and snail expression .\n\nIn 81B-Fb cells , downregulation of EGFR , which is mediated by increased ubiquitination , and activation of downstream protein kinase B ( Akt ) , glycogen synthase kinase-beta ( GSK-3\u03b2 ) signalling and upregulation of snail expression were observed compared with UMSCC81B cells .\n\nLY294002 , but not U0126 , suppressed foetal bovine serum or heregulin-\u03b21-induced phosphorylation of Akt/GSK-3\u03b2 and snail expression together with the inhibition of 81B-Fb cell motility .\n\nFurthermore , forced expression of EGFR resulted in partial restoration of gefitinib sensitivity and reversal of EMT .\n\nCONCLUSION These results suggest that EMT in the gefitinib-resistant cells is mediated by the downregulation of EGFR and compensatory activation of Akt/GSK-3\u03b2/snail pathway .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "D-Limonene , a common monoterepene has been shown to have antiproliferative , apoptosis-inducing and chemopreventive effects .\n\nIn the present study , we have investigated the effects of D-limonene on the growth of 7,12-dimethylbenz[a]anthracene ( DMBA)-initiated and 12-O-tetradecanoylphorbol-13-acetate ( TPA)-promoted skin tumor development .\n\nWe found that D-limonene ( 50 and 100 mg/kg body weight ) treatments to the mouse skin significantly reduced the TPA-induced ( a ) edema and hyperplasia ( p < 0.001 ) ; ( b ) expression of cyclooxygenase-2 ; ( c ) ornithine decarboxylase activity ( p < 0.001 ) ; and ( d ) [ (3)H ] thymidine incorporation into DNA ( p < 0.001 ) .\n\nIn addition , treatment of D-limonene effectively restored the level of reduced glutathione , glutathione peroxidase , glutathione reductase , glutathione S-transferase , catalase and malondialdehyde production in TPA-treated mouse skin .\n\nIn a two-stage skin tumorigenesis study , D-limonene significantly reduced the tumor burden ( p < 0.005 ) and tumor incidence as compared to DMBA/TPA-treated mice .\n\nD-Limonene treatment also extended the latency period of tumor development from 4 to 9 weeks .\n\nD-Limonene treatment decreased the expression level of Ras , Raf and phosphorylation of extracellular signal-regulated protein kinase 1/2 in DMBA/TPA-induced tumors .\n\nA decrease in the expression of Bcl-2 and an increase in Bax expression were also observed in tumor tissues of mice treated with D-limonene .\n\nTaken together , our findings suggest that D-limonene may exert its chemopreventive activity through the inhibition of inflammation , oxidative stress and Ras-signaling as well as the induction of pro-apoptotic state during TPA-mediated promotion of DMBA-induced skin cancer in mouse model .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "The Warburg effect describes a heightened propensity of tumor cells to produce lactic acid in the presence or absence of O(2) .\n\nA generally held notion is that the Warburg effect is related to energy .\n\nUsing whole-genome , proteomic MALDI-TOF-MS and metabolite analysis , we investigated the Warburg effect in malignant neuroblastoma N2a cells .\n\nThe findings show that the Warburg effect serves a functional role in regulating acidic pericellular pH ( pHe ) , which is mediated by metabolic inversion or a fluctuating dominance between glycolytic-rate substrate level phosphorylation ( SLP ) and mitochondrial ( mt ) oxidative phosphorylation ( OXPHOS ) to control lactic acid production .\n\nThe results also show that an alkaline pHe caused an elevation in SLP/OXPHOS ratio ( approximately 98% SLP/OXPHOS ) ; while the ratio was approximately 56% at neutral pHe and approximately 93% in acidic pHe .\n\nAcidic pHe paralleled greater expression of mitochondrial biogenesis and OXPHOS genes , such as complex III-V ( Uqcr10 , Atp5 and Cox7c ) , mt Fmc1 , Romo1 , Tmem 173 , Tomm6 , aldehyde dehydrogenase , mt Sod2 mt biogenesis component PPAR-\\u03b3 co-activator 1 adjunct to loss of mt fission ( Mff ) .\n\nMoreover , acidic pHe corresponded to metabolic efficiency evidenced by a rise in mTOR nutrient sensor G\\u03b2L , its downstream target ( Eif4ebp1 ) , insulin modulators ( Trib3 and Fetub ) and loss of catabolic ( Hadhb , Bdh1 and Pygl)/glycolytic processes ( aldolase C , pyruvate kinase , Nampt and aldose-reductase ) .\n\nIn contrast , alkaline pHe initiated loss of mitofusin 2 , complex II-IV ( Sdhaf1 , Uqcrq , Cox4i2 and Aldh1l2 ) , aconitase , mitochondrial carrier triple repeat 1 and mt biosynthetic ( Coq2 , Coq5 and Coq9 ) .\n\nIn conclusion , the Warburg effect might serve as a negative feedback loop that regulates the pHe toward a broad acidic range by altering lactic acid production through inversion of metabolic systems .\n\nThese effects were independent of changes in O(2) concentration or glucose supply .", "output": "Cellular energetics" }, { "input": "OBJECTIVES To investigate the immunoexpression of epidermal growth factor receptor ( EGFR ) in a sample of oral leukoplakias ( OL ) and to determine the receptor ' s association with dysplasia , tobacco consumption , lesion site , and proliferation rate .\n\nAlthough EGFR should be overexpressed in some oral leukoplakias , the factors that may interfere with this expression and the influence of this receptor on epithelial proliferation have yet to be investigated .\n\nSTUDY DESIGN Samples of oral leukoplakias ( 48 ) and of normal oral epithelium ( 10 ) were immunohistologically examined for expression of EGFR .\n\nImmunohistochemistry for Ki-67 , and p27 were also performed in leukoplakias .\n\nEGFR expression was associated with clinical and pathological features .\n\nRESULTS EGFR was positive in 62.5% of the leukoplakias and 50% of normal oral epithelium .\n\nThe number of EGFR positive OL located in high-risk sites was significantly higher than EGFR positive OL located in low-risk sites .\n\nMost of the p27 negative leukoplakias were EGFR positive , and the p27 index in the parabasal layer was diminished in the presence of dysplasia .\n\nPositivity for EGFR was not associated with dysplasia , tobacco exposure , or Ki-67 .\n\nCONCLUSION EGFR is expressed in leukoplakia regardless of dysplasia , but EGFR positivity should be more frequent in lesions sited in areas of high cancer risk .\n\nThe association between EGFR and p27 may represent an important mechanism in the control of cellular proliferation and malignant progression of oral epithelium and therefore warrants further investigation .", "output": "Sustaining proliferative signaling" }, { "input": "Vitamin D deficiency is associated with the high risk of colon cancer and a variety of other diseases .\n\nThe active vitamin D metabolite 1\u03b1,25-dihydroxyvitamin D(3) ( 1,25(OH)(2)D(3) ) regulates gene transcription via its nuclear receptor ( VDR ) , and posttranscriptional regulatory mechanisms of gene expression have also been proposed .\n\nWe have identified microRNA-22 ( miR-22 ) and several other miRNA species as 1,25(OH)(2)D(3) targets in human colon cancer cells .\n\nRemarkably , miR-22 is induced by 1,25(OH)(2)D(3) in a time- , dose- and VDR-dependent manner .\n\nIn SW480-ADH and HCT116 cells , miR-22 loss-of-function by transfection of a miR-22 inhibitor suppresses the antiproliferative effect of 1,25(OH)(2)D(3) .\n\nAdditionally , miR-22 inhibition increases cell migration per se and decreases the antimigratory effect of 1,25(OH)(2)D(3) in both cell types .\n\nIn silico analysis shows a significant overlap between genes suppressed by 1,25(OH)(2)D(3) and miR-22 putative target genes .\n\nConsistently , miR-22 inhibition abrogates the 1,25(OH)(2)D(3)-mediated suppression of NELL2 , OGN , HNRPH1 , RERE and NFAT5 genes .\n\nIn 39 out of 50 ( 78% ) human colon cancer patients , miR-22 expression was found lower in the tumour than in the matched normal tissue and correlated directly with that of VDR .\n\nOur results indicate that miR-22 is induced by 1,25(OH)(2)D(3) in human colon cancer cells and it may contribute to its antitumour action against this neoplasia .", "output": "Activating invasion and metastasis" }, { "input": "Whereas estrogen-estrogen receptor \u03b1 ( ER ) signaling plays an important role in breast cancer growth , it is also necessary for the differentiation of normal breast epithelial cells .\n\nHow this functional conversion occurs , however , remains unknown .\n\nBased on a genome-wide sequencing study that identified mutations in several breast cancer genes , we examined some of the genes for mutations , expression levels , and functional effects on cell proliferation and tumorigenesis .\n\nWe present the data for C1orf64 or ER-related factor ( ERRF ) from 31 cell lines and 367 primary breast cancer tumors .\n\nWhereas mutation of ERRF was infrequent ( 1 of 79 or 1.3% ) , its expression was up-regulated in breast cancer , and the up-regulation was more common in lower-stage tumors .\n\nIn addition , increased ERRF expression was significantly associated with ER and/or progesterone receptor ( PR ) positivity , which was still valid in human epidermal growth factor receptor 2 ( HER2)-negative tumors .\n\nIn ER-positive tumors , ERRF expression was inversely correlated with HER2 status .\n\nFurthermore , higher ERRF protein expression was significantly associated with better disease-free survival and overall survival , particularly in ER- and/or PR-positive and HER2-negative tumors ( luminal A subtype ) .\n\nFunctionally , knockdown of ERRF in two ER-positive breast cancer cell lines , T-47D and MDA-MB-361 , suppressed cell growth in vitro and tumorigenesis in xenograft models .\n\nThese results suggest that ERRF plays a role in estrogen-ER-mediated growth of breast cancer cells and could , thus , be a potential therapeutic target .", "output": "Sustaining proliferative signaling" }, { "input": "Estrogens play essential roles in the progression of mammary and prostatic diseases .\n\nThe transcriptional effects of estrogens are transduced by two estrogen receptors , ER\u03b1 and ER\u03b2 , which elicit opposing roles in regulating proliferation : ER\u03b1 is proliferative while ER\u03b2 is anti-proliferative .\n\nExogenous expression of ER\u03b2 in ER\u03b1-positive cancer cell lines inhibits cell proliferation in response to estrogen and reduces xenografted tumor growth in vivo , suggesting that ER\u03b2 might oppose ER\u03b1's proliferative effects via formation of ER\u03b1/\u03b2 heterodimers .\n\nDespite biochemical and cellular evidence of ER\u03b1/\u03b2 heterodimer formation in cells co-expressing both receptors , the biological roles of the ER\u03b1/\u03b2 heterodimer remain to be elucidated .\n\nHere we report the identification of two phytoestrogens that selectively activate ER\u03b1/\u03b2 heterodimers at specific concentrations using a cell-based , two-step high throughput small molecule screen for ER transcriptional activity and ER dimer selectivity .\n\nUsing ER\u03b1/\u03b2 heterodimer-selective ligands at defined concentrations , we demonstrate that ER\u03b1/\u03b2 heterodimers are growth inhibitory in breast and prostate cells which co-express the two ER isoforms .\n\nFurthermore , using Automated Quantitative Analysis ( AQUA ) to examine nuclear expression of ER\u03b1 and ER\u03b2 in human breast tissue microarrays , we demonstrate that ER\u03b1 and ER\u03b2 are co-expressed in the same cells in breast tumors .\n\nThe co-expression of ER\u03b1 and ER\u03b2 in the same cells supports the possibility of ER\u03b1/\u03b2 heterodimer formation at physio- and pathological conditions , further suggesting that targeting ER\u03b1/\u03b2 heterodimers might be a novel therapeutic approach to the treatment of cancers which co-express ER\u03b1 and ER\u03b2 .", "output": "Sustaining proliferative signaling" }, { "input": "Approximately 50% of human tumors have a mutation in TP53 .\n\nThe pattern and spectra of TP53 mutations often differ between cancer types , perhaps due to different etiological factors .\n\nThe Hupki ( human TP53 knock-in ) mouse embryo fibroblast ( HUF ) immortalization assay is useful for studying mutagenesis in the human TP53 gene by environmental carcinogens .\n\nPrior to initiating an immortalization assay , carcinogen treatment conditions must be optimized , which can require a large number of cells .\n\nAs primary HUF cultures senesce within 2 weeks , restricting their use , we investigated whether immortalized HUFs retaining wild-type TP53 can be surrogates for primary HUFs in initial treatment optimization .\n\nDNA damage by eight compounds found in diesel exhaust , benzo[a]pyrene , 3-nitrobenzanthrone , 1-nitropyrene , 1,3-dinitropyrene , 1,6-dinitropyrene , 1,8-dinitropyrene , 6-nitrochrysene , and 3-nitrofluorene , was assessed by ( 32 ) P-postlabeling and the alkaline comet assay in primary HUFs and in an immortal HUF cell line J201 .\n\nFor most compounds , higher levels of DNA adducts accumulated in J201 cells than in primary HUFs .\n\nThis difference was not reflected in the comet assay or by cell viability changes .\n\nExperiments in three additional immortal HUF cell lines ( AAI49 , U56 , and E2-143 ) confirmed strong differences in DNA adduct levels compared with primary HUFs .\n\nHowever , these did not correlate with the protein expression of Nqo1 or Nat1/2 , or with gene expression of Cyp1a1 or Cyp1b1 .\n\nOur results show that using immortal HUFs as surrogates for primary HUFs in genotoxicity screening has limitations and that DNA adduct formation is the best measure of genotoxicity of the nitro-polycyclic aromatic hydrocarbons tested in HUFs .", "output": "Genomic instability and mutation" }, { "input": "Cancer cells preferentially metabolize glucose through aerobic glycolysis .\n\nThis phenomenon , known as the Warburg effect , is an anomalous characteristic of glucose metabolism in cancer cells .\n\nChronic inflammation is a key promoting factor of tumourigenesis .\n\nIt remains , however , largely unexplored whether and how pro-tumourigenic inflammation regulates glucose metabolism in cancer cells .\n\nHere , we show that pro-inflammatory cytokines promote glycolysis in breast cancer cells , and that the inflammation-induced miR-155 functions as an important mediator in this process .\n\nWe further show that miR-155 acts to upregulate hexokinase 2 ( hk2 ) , through two distinct mechanisms .\n\nFirst , miR-155 promotes hk2 transcription by activation of signal transducer and activator of transcription 3 ( STAT3 ) , a transcriptional activator for hk2 .\n\nSecond , via targeting C/EBP\\u03b2 ( a transcriptional activator for mir-143 ) , miR-155 represses mir-143 , a negative regulator of hk2 , thus resulting in upregulation of hk2 expression at the post-transcriptional level .\n\nThe miR-155-mediated hk2 upregulation also appears to operate in other types of cancer cells examined .\n\nWe suggest that the miR-155/miR-143/HK2 axis may represent a common mechanism linking inflammation to the altered metabolism in cancer cells .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "Electronic properties of DNA are believed to play a crucial role in many phenomena in living organisms , for example the location of DNA lesions by base excision repair ( BER ) glycosylases and the regulation of tumor-suppressor genes such as p53 by detection of oxidative damage .\n\nHowever , the reproducible measurement and modelling of charge migration through DNA molecules at the nanometer scale remains a challenging and controversial subject even after more than a decade of intense efforts .\n\nHere we show , by analysing 162 disease-related genes from a variety of medical databases with a total of almost 20,000 observed pathogenic mutations , a significant difference in the electronic properties of the population of observed mutations compared to the set of all possible mutations .\n\nOur results have implications for the role of the electronic properties of DNA in cellular processes , and hint at the possibility of prediction , early diagnosis and detection of mutation hotspots .", "output": "Genomic instability and mutation" }, { "input": "Mast cells ( MC ) are key effector cells in allergic reactions but also involved in host defence , tissue remodeling , angiogenesis , and fibrogenesis .\n\nHere , we show that human intestinal fibroblasts ( FB ) suppress apoptosis in human intestinal MC dependent on IL-6 .\n\nIntestinal FB produced IL-6 upon direct stimulation by intestinal MC in co-culture or by MC mediators such as TNF-\u03b1 , IL-1\u03b2 , tryptase or histamine .\n\nMC incubated with IL-6 survived for up to 3 weeks similar to MC co-cultured with FB and MC survival could be blocked by neutralizing anti-IL-6 Abs .\n\nMoreover , FB stimulated by MC mediators upregulated their expression of matrix metalloproteinase-1 ( MMP-1 ) , a key fibrolytic enzyme .\n\nNoteworthy , FB co-cultured with MC or treated with MMP-1 lost confluence and showed increased numbers of apoptotic cells .\n\nOur data indicate an intimate cross talk between mucosal MC and FB resulting in MC survival and induction of a fibrolytic rather than a profibrotic state in FB .", "output": "Resisting cell death" }, { "input": "Epidemiological studies have indicated that obesity is associated with colorectal cancer .\n\nThe obesity hormone leptin is considered as a key mediator for cancer development and progression .\n\nThe present study aims to investigate regulatory effects of leptin on colorectal carcinoma .\n\nThe expression of leptin and its receptor Ob-R was examined by immunohistochemistry in 108 Chinese patients with colorectal carcinoma .\n\nThe results showed that leptin/Ob-R expression was significantly associated with T stage , TNM stage , lymph node metastasis , distant metastasis , differentiation and expression of p-mTOR , p-70S6 kinase , and p-Akt .\n\nFurthermore , the effects of leptin on proliferation and apoptosis of HCT-116 colon carcinoma cells were determined .\n\nThe results showed that leptin could stimulate the proliferation and inhibit the apoptosis of HCT-116 colon cells through the PI3K/Akt/mTOR pathway .\n\nLy294002 ( a PI3K inhibitor ) and rapamycin ( an mTOR inhibitor ) could prevent the regulatory effects of leptin on the proliferation and apoptosis of HCT-116 cells via abrogating leptin-mediated PI3K/Akt/mTOR pathway .\n\nAll these results indicated that leptin could regulate proliferation and apoptosis of colorectal carcinoma through the PI3K/Akt/ mTOR signalling pathway .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Clear cell renal cell carcinoma ( ccRCC ) is the most common pathological subtype of kidney cancer .\n\nHere , we integrated an unbiased genome-wide RNA interference screen for ccRCC survival regulators with an analysis of recurrently overexpressed genes in ccRCC to identify new therapeutic targets in this disease .\n\nOne of the most potent survival regulators , the monocarboxylate transporter MCT4 ( SLC16A3 ) , impaired ccRCC viability in all eight ccRCC lines tested and was the seventh most overexpressed gene in a meta-analysis of five ccRCC expression datasets .\n\nMCT4 silencing impaired secretion of lactate generated through glycolysis and induced cell cycle arrest and apoptosis .\n\nSilencing MCT4 resulted in intracellular acidosis , and reduction in intracellular ATP production together with partial reversion of the Warburg effect in ccRCC cell lines .\n\nIntra-tumoural heterogeneity in the intensity of MCT4 protein expression was observed in primary ccRCCs .\n\nMCT4 protein expression analysis based on the highest intensity of expression in primary ccRCCs was associated with poorer relapse-free survival , whereas modal intensity correlated with Fuhrman nuclear grade .\n\nConsistent with the potential selection of subclones enriched for MCT4 expression during disease progression , MCT4 expression was greater at sites of metastatic disease .\n\nThese data suggest that MCT4 may serve as a novel metabolic target to reverse the Warburg effect and limit disease progression in ccRCC .", "output": "Activating invasion and metastasis, Cellular energetics, Resisting cell death" }, { "input": "An acquired mutation ( T790M ) in the epidermal growth factor receptor ( EGFR ) accounts for half of all relapses in non-small cell lung cancer ( NSCLC ) patients who initially respond to EGFR kinase inhibitors .\n\nIn this study , we demonstrated for the first time that EGFR-T790M interacts with the cytoskeletal components , myosin heavy chain 9 ( MYH9 ) and \u03b2-actin , in the nucleus of H1975 cells carrying the T790M-mutant EGFR .\n\nThe interactions of EGFR with MYH9 and \u03b2-actin were reduced in the presence of blebbistatin , a specific inhibitor for the MYH9-\u03b2-actin interaction , suggesting that the EGFR interaction with MYH9 and \u03b2-actin is affected by the integrity of the cytoskeleton .\n\nThese physical interactions among MYH9 , \u03b2-actin , and EGFR were also impaired by CL-387,785 , a kinase inhibitor for EGFR-T790M .\n\nFurthermore , CL-387,785 and blebbistatin interacted in a synergistic fashion to suppress cell proliferation and induce apoptosis in H1975 cells .\n\nThe combination of CL-387,785 and blebbistatin enhanced the down-regulation of cyclooxygenase-2 ( COX-2 ) , a transcriptional target of nuclear EGFR .\n\nOverall , our findings demonstrate that disrupting EGFR interactions with the cytoskeletal components enhanced the anti-cancer effects of CL-387,785 against H1975 cells , suggesting a novel therapeutic approach for NSCLC cells that express the drug-resistant EGFR-T790M .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "A novel series of N(4)-(3-chlorophenyl)-5-(oxazol-2-yl)pyrimidine-4,6-diamines were synthesized and evaluated as dual inhibitors of HER-1/HER-2 tyrosine kinases .\n\nIn contrast to the currently approved HER-2-targeted agent ( lapatinib , 1 ) , our irreversible HER-1/HER-2 inhibitors have the potential to overcome the clinically relevant and mutation-induced drug resistance .\n\nThe selected compound ( 19a ) showed excellent inhibitory activity toward HER-1/HER-2 tyrosine kinases with selectivity over 20 other kinases and inhibited the proliferation of both cancer cell types : lapatinib-sensitive cell lines ( SK-Br3 , MDA-MB-175 , and N87 ) and lapatinib-resistant cell lines ( MDA-MB-453 , H1781 , and H1975 ) .\n\nThe excellent pharmacokinetic profiles of 19a in mice and rats led us to further investigation of a novel therapeutic agent for HER-2-targeting treatment of solid tumors , especially HER-2-positive breast/gastric cancer and HER-2-mutated lung cancer .", "output": "Sustaining proliferative signaling" }, { "input": "The progression of prostate cancers ( PCs ) to locally invasive , androgen-independent and metastatic disease states is generally associated with treatment resistance and disease relapse .\n\nThe present study was undertaken to establish the possibility of using a combination of specific oncogenic products , including epidermal growth factor receptor ( EGFR ) , pAkt , nuclear factor-kappaB ( NF-\u03baB ) and macrophage inhibitory cytokine-1 ( MIC-1 ) as biomarkers and therapeutic targets for optimizing the management of patients with localized PC at earlier disease stages .\n\nThe immunohistochemical and immunofluorescence data have revealed that the expression levels of EGFR , Ser(473)-pAkt , NF-\u03baB p65 and MIC-1 proteins were significantly enhanced in the same subset of 76 cases of prostatic adenocarcinoma specimens during the disease progression and these biomarkers were expressed in a small subpopulation of CD133(+) PC cells and the bulk tumor mass of CD133(-) PC cells .\n\nImportantly , all of these biomarkers were also overexpressed in 80-100% of 30 PC metastasis bone tissue specimens .\n\nMoreover , the results have indicated that the EGF-EGFR signaling pathway can provide critical functions for the self-renewal of side population ( SP ) cells endowed with stem cell-like features from highly invasive WPE1-NB26 cells .\n\nOf therapeutic interest , the targeting of EGFR , pAkt , NF-\u03baB or MIC-1 was also effective at suppressing the basal and EGF-promoted prostasphere formation by SP WPE1-NB26 cells , inducing disintegration of SP cell-derived prostaspheres and decreasing the viability of SP and non-SP WPE1-NB26 cell fractions .\n\nAlso , the targeting of these oncogenic products induced the caspase-dependent apoptosis in chemoresistant SP WPE1-NB26 cells and enhanced their sensibility to the cytotoxic effects induced by docetaxel .\n\nThese findings suggest that the combined use of EGFR , pAkt , NF-\u03baB and/or MIC-1 may represent promising strategies for improving the accuracy of current diagnostic and prognostic methods and efficacy of treatments of PC patients in considering the disease heterogeneity , thereby preventing PC progression to metastatic and lethal disease states .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "BACKGROUND Surgical procedures such as liver resection and liver transplantation are the first-line treatments for hepatocellular carcinoma ( HCC ) patients .\n\nHowever , the high incidence of tumor recurrence and metastasis after liver surgery remains a major problem .\n\nRecent studies have shown that hepatic ischemia-reperfusion ( I/R ) injury and endothelial progenitor cells ( EPCs ) contribute to tumor growth and metastasis .\n\nWe aim to investigate the mechanism of FTY720 , which was originally applied as an immunomodulator , on suppression of liver tumor metastasis after liver resection and partial hepatic I/R injury .\n\nMETHODOLOGY/PRINCIPAL FINDINGS An orthotopic liver tumor model in Buffalo rat was established using the hepatocellular carcinoma cell line McA-RH7777 .\n\nTwo weeks after orthotopic liver tumor implantation , the rats underwent liver resection for tumor-bearing lobe and partial hepatic I/R injury .\n\nFTY720 ( 2 mg/kg ) was administered through the inferior caval vein before and after I/R injury .\n\nBlood samples were taken at days 0 , 1 , 3 , 7 , 14 , 21 and 28 for detection of circulating EPCs ( CD133+CD34+ ) .\n\nOur results showed that intrahepatic and lung metastases were significantly inhibited together with less tumor angiogenesis by FTY720 treatment .\n\nThe number of circulating EPCs was also significantly decreased by FTY720 treatment from day 7 to day 28 .\n\nHepatic gene expressions of CXCL10 , VEGF , CXCR3 , CXCR4 induced by hepatic I/R injury were down-regulated in the treatment group .\n\nCONCLUSIONS/SIGNIFICANCE FTY720 suppressed liver tumor metastasis after liver resection marred by hepatic I/R injury in a rat liver tumor model by attenuating hepatic I/R injury and reducing circulating EPCs .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Partial or whole-brain irradiation is often required to treat both primary and metastatic brain cancer .\n\nRadiation-induced normal tissue injury , including progressive cognitive impairment , however , can significantly affect the well-being of the approximately 200,000 patients who receive these treatments each year in the United States .\n\nAlthough the exact mechanisms underlying radiation-induced late effects remain unclear , oxidative stress and inflammation are thought to play a critical role .\n\nMicroglia are key mediators of neuroinflammation .\n\nPeroxisomal proliferator-activated receptor ( PPAR ) \u03b4 has been shown to be a potent regulator of anti-inflammatory responses .\n\nThus , we hypothesized that PPAR\u03b4 activation would modulate the radiation-induced inflammatory response in microglia .\n\nIncubating BV-2 murine microglial cells with the PPAR\u03b4 agonist L-165041 prevented the radiation-induced increase in : ( i ) intracellular reactive oxygen species generation , ( ii ) Cox-2 and MCP-1 expression , and ( iii ) IL-1\u03b2 and TNF-\u03b1 message levels .\n\nThis occurred , in part , through PPAR\u03b4-mediated modulation of stress-activated kinases and proinflammatory transcription factors .\n\nPPAR\u03b4 inhibited NF-\u03baB via transrepression by physically interacting with the p65 subunit and prevented activation of the PKC\u03b1/MEK1/2/ERK1/2/AP-1 pathway by inhibiting the radiation-induced increase in intracellular reactive oxygen species generation .\n\nThese data support the hypothesis that PPAR\u03b4 activation can modulate radiation-induced oxidative stress and inflammatory responses in microglia .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND Death receptors ( DR ) of the TNF family function as anti-tumor immune effector molecules .\n\nTumor cells , however , often exhibit DR-signaling resistance .\n\nPrevious studies indicate that radiation can modify gene expression within tumor cells and increase tumor cell sensitivity to immune attack .\n\nThe aim of this study is to investigate the synergistic effect of sub-lethal doses of ionizing radiation in sensitizing colorectal carcinoma cells to death receptor-mediated apoptosis .\n\nMETHODOLOGY/PRINCIPAL FINDINGS The ability of radiation to modulate the expression of multiple death receptors ( Fas/CD95 , TRAILR1/DR4 , TRAILR2/DR5 , TNF-R1 and LT\u03b2R ) was examined in colorectal tumor cells .\n\nThe functional significance of sub-lethal doses of radiation in enhancing tumor cell susceptibility to DR-induced apoptosis was determined by in vitro functional sensitivity assays .\n\nThe longevity of these changes and the underlying molecular mechanism of irradiation in sensitizing diverse colorectal carcinoma cells to death receptor-mediated apoptosis were also examined .\n\nWe found that radiation increased surface expression of Fas , DR4 and DR5 but not LT\u03b2R or TNF-R1 in these cells .\n\nIncreased expression of DRs was observed 2 days post-irradiation and remained elevated 7-days post irradiation .\n\nSub-lethal tumor cell irradiation alone exhibited minimal cell death , but effectively sensitized three of three colorectal carcinoma cells to both TRAIL and Fas-induced apoptosis , but not LT\u03b2R-induced death .\n\nFurthermore , radiation-enhanced Fas and TRAIL-induced cell death lasted as long as 5-days post-irradiation .\n\nSpecific analysis of intracellular sensitizers to apoptosis indicated that while radiation did reduce Bcl-X(L) and c-FLIP protein expression , this reduction did not correlate with the radiation-enhanced sensitivity to Fas and/or TRAIL mediated apoptosis among the three cell types .\n\nCONCLUSIONS/SIGNIFICANCE Irradiation of tumor cells can overcome Fas and TRAIL resistance that is long lasting .\n\nOverall , results of these investigations suggest that non-lethal doses of radiation can be used to make human tumors more amenable to attack by anti-tumor effector molecules and cells .", "output": "Resisting cell death" }, { "input": "We have recently proposed a new two-compartment model for understanding the Warburg effect in tumor metabolism .\n\nIn this model , glycolytic stromal cells produce mitochondrial fuels ( L-lactate and ketone bodies ) that are then transferred to oxidative epithelial cancer cells , driving OXPHOS and mitochondrial metabolism .\n\nThus , stromal catabolism fuels anabolic tumor growth via energy transfer .\n\nWe have termed this new cancer paradigm the \" reverse Warburg effect, \" because stromal cells undergo aerobic glycolysis , rather than tumor cells .\n\nTo assess whether this mechanism also applies during cancer cell metastasis , we analyzed the bioenergetic status of breast cancer lymph node metastases , by employing a series of metabolic protein markers .\n\nFor this purpose , we used MCT4 to identify glycolytic cells .\n\nSimilarly , we used TO MM20 and COX staining as markers of mitochondrial mass and OXPHOS activity , respectively .\n\nConsistent with the \" reverse Warburg effect, \" our results indicate that metastatic breast cancer cells amplify oxidative mitochondrial metabolism ( OXPHOS ) and that adjacent stromal cells are glycolytic and lack detectable mitochondria .\n\nGlycolytic stromal cells included cancer-associated fibroblasts , adipocytes and inflammatory cells .\n\nDouble labeling experiments with glycolytic ( MCT4 ) and oxidative ( TO MM20 or COX ) markers directly shows that at least two different metabolic compartments co-exist , side-by-side , within primary tumors and their metastases .\n\nSince cancer-associated immune cells appeared glycolytic , this observation may also explain how inflammation literally \" fuels \" tumor progression and metastatic dissemination , by \" feeding \" mitochondrial metabolism in cancer cells .\n\nFinally , MCT4(+) and TO MM20(-) \" glycolytic \" cancer cells were rarely observed , indicating that the conventional \" Warburg effect \" does not frequently occur in cancer-positive lymph node metastases .", "output": "Tumor promoting inflammation, Cellular energetics, Activating invasion and metastasis" }, { "input": "BACKGROUND Estrogen receptor \u03b2 ( ER\u03b2 ) is the predominant ER in the colorectal epithelium , whose expression is greatly reduced in colorectal cancer compared with normal colon tissue .\n\nRecent in vitro studies suggested that ER\u03b2 may suppress tumor growth .\n\nNo research was reported whether ER\u03b2 can be used as therapeutic agent for colon cancer .\n\nMETHODS In this study , ER\u03b2 gene constructed into adenoviral ( Ad ) vectors was used to treat colon cancer HCT-116 cells alone or in combination with raloxifene .\n\nIn vitro and in vivo studies were conducted to investigate the therapeutic effects of ER\u03b2 and raloxifene in HCT-116 cells .\n\nRESULTS Our results indicated that , although Ad-ER\u03b2 alone had no effect on the proliferation of HCT-116 cells , the combination of Ad-ER\u03b2 with raloxifene significantly inhibited the proliferation of HCT-116 cells .\n\nThe apparently apoptotic induction effects may partly explain the cytotoxicity of the two agents .\n\nThe results of the study of ER\u03b2 on migration and invasion of HCT-116 cells demonstrated that overexpression of ER\u03b2 significantly decreased cell migration and increased invasion of cells .\n\nThe antitumor efficacies of ER\u03b2 as well as raloxifene were further investigated on HCT-116 tumor bearing mice .\n\nResults demonstrated that both Ad-ER\u03b2 and raloxifene individually inhibited tumor growth .\n\nThe combination group showed the highest inhibitory efficiency compared with other three groups .\n\nCONCLUSION These findings demonstrated that combined administration of Ad-ER\u03b2 with raloxifene represents a promising colon cancer therapeutic strategy .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "It has been shown that regulation of EGFR expression in prostate cancer cells is mostly at the transcriptional level. microRNA-133 ( miR-133 ) has long been recognized as a muscle-specific miRNA which may regulate myoblast differentiation and participate in many myogenic diseases .\n\nRecently , it has been reported that miR-133 is also involved in other tumors , such as bladder cancer , esophageal cancer and may regulate cell motility in these cancer cells .\n\nIn the present study , we examined the expression and effects of miR-133 in two hormone-insensitive prostate cancer cell lines .\n\nThe expression of miR-133a and miR-133b were analyzed by quantitative RT-PCR .\n\nAfter transfection of miR-133a and miR-133b , cell viability assay , luciferase assay , western blot analysis , cell migration and invasion assay were conducted in Du145 and PC3 cells .\n\nIn this study , we showed that miR\u2011133a and miR-133b are expressed at the detection limit in two hormone-insensitive prostate cancer cell lines , PC3 and DU145 .\n\nEctopic expression of miR-133 inhibited cell proliferation , migration and invasion in these cells .\n\nWe also provide the first evidence that miR-133 may target EGFR .\n\nOur study provided the first glimpse of the functional role of miR-133 in two hormone-independent prostate cancer cell lines .\n\nThese results may add to our knowledge on the molecular basis of prostate cancer progression .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Immortalization ( senescence bypass ) is a critical rate-limiting step in the malignant transformation of mammalian somatic cells .\n\nHuman cells must breach at least two distinct senescence barriers to permit unfettered clonal evolution during cancer development : ( 1 ) stress- or oncogene-induced premature senescence ( SIPS/OIS ) , mediated via the p16-Rb and/or ARF-p53-p21 tumour-suppressive pathways , and ( 2 ) replicative senescence triggered by telomere shortening .\n\nIn contrast , because their telomerase is constitutively active , cells from small rodents possess only the SIPS/OIS barrier , and are therefore useful for studying SIPS/OIS bypass in isolation .\n\nDermal fibroblasts from the Syrian hamster ( SHD cells ) are exceptionally resistant to spontaneous SIPS bypass , but it can be readily induced following exposure to a wide range of chemical and physical carcinogens .\n\nHere we show that a spectrum of carcinogen-specific mutational and epigenetic alterations involving the INK4A ( p16 ) , p53 and INK4B ( p15 ) genes are associated with induced SIPS bypass .\n\nWith ionizing radiation , immortalization is invariably accompanied by efficient biallelic deletion of the complete INK4/CDKN2 locus .\n\nIn comparison , SHD cells immortalized by the powerful polycyclic hydrocarbon carcinogen benzo(a)pyrene display transversion point mutations in the DNA-binding domain of p53 coupled with INK4 alterations such as loss of expression of p15 .\n\nEpimutational silencing of p16 is the primary event associated with immortalization by nickel , a human non-genotoxic carcinogen .\n\nAs SIPS/OIS bypass is a prerequisite for the immortalization of normal diploid human epithelial cells , our results with the SHD model will provide a basis for delineating combinations of key molecular changes underpinning this important event in human carcinogenesis .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "BACKGROUND Neuroblastoma ( NBL ) is a common pediatric solid tumor , and outcomes for patients with advanced neuroblastoma remain poor despite extremely aggressive treatment .\n\nChemotherapy resistance at relapse contributes heavily to treatment failure .\n\nThe poor survival of patients with high-risk NBL prompted this investigation into novel treatment options with the objective of gaining a better understanding of resistance mechanisms .\n\nOn the basis of previous work and on data from publicly available studies , the authors hypothesized that human epidermal growth factor receptor 4 ( Her4 ) contributes to resistance .\n\nMETHODS Her4 expression was reduced with small-hairpin RNA ( shRNA ) to over express intracellular HER4 , and the authors tested its impact on tumor cell survival under various culture conditions .\n\nThe resulting changes in gene expression after HER4 knockdown were measured by using a messenger RNA ( mRNA ) array .\n\nRESULTS HER4 expression was up-regulated in tumor spheres compared with the expression in monolayer culture .\n\nWith HER4 knockdown , NBL cells became less resistant to anoikis and serum starvation .\n\nMoreover , HER4 knockdown increased the chemosensitivity of NBL cells to cisplatin , doxorubicin , etoposide , and activated ifosfamide .\n\nIn mRNA array analysis , HER4 knockdown predominately altered genes related to cell cycle regulation .\n\nIn NBL spheres compared with monolayers , cell proliferation was decreased , and cyclin D expression was reduced .\n\nHER4 knockdown reversed cyclin D suppression .\n\nOverexpressed intracellular HER4 slowed the cell cycle and induced chemoresistance .\n\nCONCLUSIONS The current results indicated that HER4 protects NBL cells from multiple exogenous apoptotic stimuli , including anoikis , nutrient deficiency , and cytotoxic chemotherapy .\n\nThe intracellular fragment of HER4 was sufficient to confer this phenotype .\n\nHER4 functions as a cell cycle suppressor , maintaining resistance to cellular stress .\n\nThe current findings indicate that HER4 overexpression may be associated with refractory disease , and HER4 may be an important therapeutic target .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Tumor necrosis factor ( TNF ) is an important inflammatory cytokine and induces many cellular responses , including inflammation , cell proliferation , apoptosis , and necrosis .\n\nIt is known that receptor interacting protein ( RIP ) kinases , RIP1 and RIP3 , are key effectors of TNF-induced necrosis , but little is known about how these two RIP kinases mediate this process , although reactive oxygen species ( ROS ) generation and JNK activation have been suggested to be two downstream events of RIP kinases .\n\nHere we report the identification of mixed lineage kinase domain-like , MLKL , as a key RIP3 downstream component of TNF-induced necrosis .\n\nThrough screening a kinase/phosphatase shRNA library in human colon adenocarcinoma HT-29 cells , we found that knockdown of MLKL blocked TNF-induced necrosis .\n\nOur data suggest that MLKL functions downstream of RIP1 and RIP3 and is recruited to the necrosome through its interaction with RIP3 .\n\nFinally , we found that MLKL is required for the generation of ROS and the late-phase activation of JNK during TNF-induced necrosis .\n\nHowever , because these two events are not involved in TNF-induced necrosis in HT-29 cells , the target of MLKL during TNF-induced necrosis remains elusive .\n\nTaken together , our study suggests that MLKL is a key RIP3 downstream component of TNF-induced necrotic cell death .", "output": "Resisting cell death" }, { "input": "Stable ectopic expression of Flt3 receptor tyrosine kinase is usually performed in interleukin 3 ( IL-3)-dependent murine cell lines like Ba/F3 , resulting in loss of IL-3 dependence .\n\nSuch high-level Flt3 expression has to date not been reported in human acute myeloid leukemia ( AML ) cell lines , despite the fact that oncogenic Flt3 aberrancies are frequent in AML patients .\n\nWe show here that ectopic Flt3 expression in different human cancer cell lines might reduce proliferation and induce apoptotic cell death , involving Bax/Bcl2 modulation .\n\nSelective depletion of Flt3-expressing cells occurred in human AML cell lines transduced with retroviral Flt3 constructs , shown here using the HL-60 leukemic cell line .\n\nFlt3 expression was investigated in two cellular model systems , the SAOS-2 osteosarcoma cell line and the human embryonic kidney HEK293 cell line , and proliferation was reduced in both systems .\n\nHEK293 cells underwent apoptosis upon ectopic Flt3 expression and cell death could be rescued by overexpression of Bcl-2 .\n\nFurthermore , we observed that the Flt3-induced inhibition of proliferation in HL-60 cells appeared to be Bax-dependent .\n\nOur results thus suggest that excessive Flt3 expression has growth-suppressive properties in several human cancer cell lines .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Cigarette smoking is implicated in numerous diseases , including emphysema and lung cancer .\n\nThe clinical expression of lung disease in smokers is not well explained by currently defined variations in gene expression or simple differences in smoking exposure .\n\nAlveolar macrophages play a critical role in the inflammation and remodeling of the lung parenchyma in smoking-related lung disease .\n\nSignificant gene expression changes in alveolar macrophages from smokers have been identified .\n\nHowever , the mechanism for these changes remains unknown .\n\nOne potential mechanism for smoking-altered gene expression is via changes in cytosine methylation in DNA regions proximal to gene-coding sequences .\n\nIn this study , alveolar macrophage DNA from heavy smokers and never smokers was isolated and methylation status at 25,000 loci determined .\n\nWe found differential methylation in genes from immune-system and inflammatory pathways .\n\nAnalysis of matching gene expression data demonstrated a parallel enrichment for changes in immune-system and inflammatory pathways .\n\nA significant number of genes with smoking-altered mRNA expression had inverse changes in methylation status .\n\nOne gene highlighted by this data was the FLT1 , and further studies found particular up-regulation of a splice variant encoding a soluble inhibitory form of the receptor .\n\nIn conclusion , chronic cigarette smoke exposure altered DNA methylation in specific gene promoter regions in human alveolar macrophages .", "output": "Tumor promoting inflammation" }, { "input": "Keratinocyte growth factor ( KGF , fibroblast growth factor-7 ) is a fibroblast-derived mitogen , which stimulates proliferation of epithelial cells .\n\nThe expression of KGF by dermal fibroblasts is induced following injury and it promotes wound repair .\n\nHowever , the role of KGF in cutaneous carcinogenesis and cancer progression is not known .\n\nWe have examined the role of KGF in progression of squamous cell carcinoma ( SCC ) of the skin .\n\nThe expression of KGF receptor ( KGFR ) mRNA was lower in cutaneous SCCs ( n = 6 ) than in normal skin samples ( n = 6 ) .\n\nExpression of KGFR mRNA was detected in 6 out of 8 cutaneous SCC cell lines and the levels were downregulated by 24-h treatment with KGF .\n\nKGF did not stimulate SCC cell proliferation , but it reduced invasion of SCC cells through collagen .\n\nGene expression profiling of three cutaneous SCC cell lines treated with KGF for 24 h revealed a specific gene expression signature characterized by upregulation of a set of genes specifically downregulated in SCC cells compared to normal epidermal keratinocytes , including genes with tumor suppressing properties ( SPRY4 , DUSP4 , DUSP6 , LRIG1 , PHLDA1 ) .\n\nKGF also induced downregulation of a set of genes specifically upregulated in SCC cells compared to normal keratinocytes , including genes associated with tumor progression ( MMP13 , MATN2 , CXCL10 , and IGFBP3 ) .\n\nDownregulation of MMP-13 and KGFR expression in SCC cells and HaCaT cells was mediated via ERK1/2 .\n\nActivation of ERK1/2 in HaCaT cells and tumorigenic Ha-ras-transformed HaCaT cells resulted in downregulation of MMP-13 and KGFR expression .\n\nThese results provide evidence , that KGF does not promote progression of cutaneous SCC , but rather suppresses the malignant phenotype of cutaneous SCC cells by regulating the expression of several genes differentially expressed in SCC cells , as compared to normal keratinocytes .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Switching to a glycolytic metabolism is a rapid adaptation of tumor cells to hypoxia .\n\nAlthough this metabolic conversion may primarily represent a rescue pathway to meet the bioenergetic and biosynthetic demands of proliferating tumor cells , it also creates a gradient of lactate that mirrors the gradient of oxygen in tumors .\n\nMore than a metabolic waste , the lactate anion is known to participate to cancer aggressiveness , in part through activation of the hypoxia-inducible factor-1 ( HIF-1 ) pathway in tumor cells .\n\nWhether lactate may also directly favor HIF-1 activation in endothelial cells ( ECs ) thereby offering a new druggable option to block angiogenesis is however an unanswered question .\n\nIn this study , we therefore focused on the role in ECs of monocarboxylate transporter 1 ( MCT1 ) that we previously identified to be the main facilitator of lactate uptake in cancer cells .\n\nWe found that blockade of lactate influx into ECs led to inhibition of HIF-1-dependent angiogenesis .\n\nOur demonstration is based on the unprecedented characterization of lactate-induced HIF-1 activation in normoxic ECs and the consecutive increase in vascular endothelial growth factor receptor 2 ( VEGFR2 ) and basic fibroblast growth factor ( bFGF ) expression .\n\nFurthermore , using a variety of functional assays including endothelial cell migration and tubulogenesis together with in vivo imaging of tumor angiogenesis through intravital microscopy and immunohistochemistry , we documented that MCT1 blockers could act as bona fide HIF-1 inhibitors leading to anti-angiogenic effects .\n\nTogether with the previous demonstration of MCT1 being a key regulator of lactate exchange between tumor cells , the current study identifies MCT1 inhibition as a therapeutic modality combining antimetabolic and anti-angiogenic activities .", "output": "Inducing angiogenesis" }, { "input": "Identification of molecular target(s) and mechanism(s) of silica-induced pulmonary toxicity is important for the intervention and/or prevention of diseases associated with exposure to silica .\n\nRats were exposed to crystalline silica by inhalation ( 15 mg m(-3) , 6 h per day , 5 days ) and global gene expression profile was determined in the lungs by microarray analysis at 1 , 2 , 4 , 8 and 16 weeks following termination of silica exposure .\n\nThe number of significantly differentially expressed genes ( >1.5-fold change and <0.01 false discovery rate P-value ) detected in the lungs during the post-exposure time intervals analyzed exhibited a steady increase in parallel with the progression of silica-induced pulmonary toxicity noticed in the rats .\n\nQuantitative real-time PCR analysis of a representative set of 10 genes confirmed the microarray findings .\n\nThe number of biological functions , canonical pathways and molecular networks significantly affected by silica exposure , as identified by the bioinformatics analysis of the significantly differentially expressed genes detected during the post-exposure time intervals , also exhibited a steady increase similar to the silica-induced pulmonary toxicity .\n\nGenes involved in oxidative stress , inflammation , respiratory diseases , cancer , and tissue remodeling and fibrosis were significantly differentially expressed in the rat lungs ; however , unresolved inflammation was the single most significant biological response to pulmonary exposure to silica .\n\nExcessive mucus production , as implicated by significant overexpression of the pendrin coding gene , SLC26A4 , was identified as a potential novel mechanism for silica-induced pulmonary toxicity .\n\nCollectively , the findings of our study provided insights into the molecular mechanisms underlying the progression of crystalline silica-induced pulmonary toxicity in the rat .\n\nPublished 2012 .\n\nThis article is a US Government work and is in the public domain in the USA .", "output": "Tumor promoting inflammation" }, { "input": "Cancer cells exhibit altered glucose metabolism characterized by a preference for aerobic glycolysis or the Warburg effect , and the cells resist matrix detachment-induced apoptosis , which is called anoikis , a barrier to metastasis .\n\nIt remains largely unclear whether tumor metabolism influences anoikis and metastasis .\n\nHere we show that when detached from the matrix , untransformed mammary epithelial cells undergo metabolic reprogramming by markedly upregulating pyruvate dehydrogenase ( PDH ) kinase 4 ( PDK4 ) through estrogen-related receptor gamma ( ERR\\u03b3 ) , thereby inhibiting PDH and attenuating the flux of glycolytic carbon into mitochondrial oxidation .\n\nTo decipher the significance of this metabolic response , we found that depletion of PDK4 or activation of PDH increased mitochondrial respiration and oxidative stress in suspended cells , resulting in heightened anoikis .\n\nConversely , overexpression of PDKs prolonged survival of cells in suspension .\n\nTherefore , decreased glucose oxidation following cell detachment confers anoikis resistance .\n\nUnlike untransformed cells , most cancer cells demonstrate reduced glucose oxidation even under attached conditions , and thus they inherently possess a survival advantage when suspended .\n\nNormalization of glucose metabolism by stimulating PDH in cancer cells restores their susceptibility to anoikis and impairs their metastatic potential .\n\nThese results suggest that the Warburg effect , more specifically , diminished glucose oxidation , promotes anoikis resistance and metastasis and that PDKs are potential targets for antimetastasis therapy .", "output": "Sustaining proliferative signaling, Cellular energetics, Activating invasion and metastasis" }, { "input": "BACKGROUND The epidermal growth factor receptor ( EGFR ) is a validated therapeutic target in non-small cell lung cancer ( NSCLC ) .\n\nHowever , current single agent receptor targeting does not achieve a maximal therapeutic effect , and some mutations confer resistance to current available agents .\n\nIn the current study we have examined , in different NSCLC cell lines , the combined effect of RNA interference targeting the EGFR mRNA , and inactivation of EGFR signaling using different receptor tyrosine kinase inhibitors ( TKIs ) or a monoclonal antibody cetuximab .\n\nMETHODS NSCLC cells ( cell lines HCC827 , H292 , H358 , H1650 , and H1975 ) were transfected with EGFR siRNA and/or treated with the TKIs gefitinib , erlotinib , and afatinib , and/or with the monoclonal antibody cetuximab .\n\nThe reduction of EGFR mRNA expression was measured by real-time quantitative RT-PCR .\n\nThe down-regulation of EGFR protein expression was measured by western blot , and the proliferation , viability , caspase3/7 activity , and apoptotic morphology were monitored by spectrophotometry , fluorimetry , and fluorescence microscopy .\n\nThe combined effect of EGFR siRNA and different drugs was evaluated using a combination index .\n\nRESULTS EGFR-specific siRNA strongly inhibited EGFR protein expression almost equally in all cell lines and inhibited cell growth and induced cell apoptosis in all NSCLC cell lines studied , albeit with a different magnitude .\n\nThe effects on growth obtained with siRNA was strikingly different from the effects obtained with TKIs .\n\nThe effects of siRNA probably correlate with the overall oncogenic significance of the receptor , which is only partly inhibited by the TKIs .\n\nThe cells which showed weak response to TKIs , such as the H1975 cell line containing the T790M resistance mutation , were found to be responsive to siRNA knockdown of EGFR , as were cell lines with downstream TKI resistance mutations .\n\nThe cell line HCC827 , harboring an exon 19 deletion mutation , was more than 10-fold more sensitive to TKI proliferation inhibition and apoptosis induction than any of the other cell lines .\n\nCetuximab alone had no relevant in vitro activity at concentrations obtainable in the clinic .\n\nThe addition of EGFR siRNA to either TKIs or cetuximab additively enhanced growth inhibition and induction of apoptosis in all five cell lines , independent of the EGFR mutation status ( wild-type or sensitizing mutation or resistant mutation ) .\n\nThe strongest biological effect was observed when afatinib was combined with an EGFR-specific siRNA .\n\nCONCLUSIONS EGFR knockdown by siRNA further decreases the cell growth of lung cancer cells that are treated with TKIs or cetuximab alone , confirming that single agent drug targeting does not achieve a maximal biological effect .\n\nThe siRNA inhibits EGFR oncogenic activity that bypasses downstream \" resistance \" mutations such as KRAS and PTEN .\n\nThe combined treatment of siRNA and EGFR inhibitory agents is additive .\n\nThe combination of a potent , irreversible kinase inhibitor such as afatinib , with EGFR-specific siRNAs should be further investigated as a new strategy in the treatment of lung cancer and other EGFR dependent cancers , including those with downstream resistance mutations .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Resisting cell death" }, { "input": "EBV latent antigen EBNA3C is indispensible for in vitro B-cell immortalization resulting in continuously proliferating lymphoblastoid cell lines ( LCLs ) .\n\nEBNA3C was previously shown to target pRb for ubiquitin-proteasome mediated degradation , which facilitates G1 to S transition controlled by the major transcriptional activator E2F1 .\n\nE2F1 also plays a pivotal role in regulating DNA damage induced apoptosis through both p53-dependent and -independent pathways .\n\nIn this study , we demonstrate that in response to DNA damage LCLs knocked down for EBNA3C undergo a drastic induction of apoptosis , as a possible consequence of both p53- and E2F1-mediated activities .\n\nImportantly , EBNA3C was previously shown to suppress p53-induced apoptosis .\n\nNow , we also show that EBNA3C efficiently blocks E2F1-mediated apoptosis , as well as its anti-proliferative effects in a p53-independent manner , in response to DNA damage .\n\nThe N- and C-terminal domains of EBNA3C form a stable pRb independent complex with the N-terminal DNA-binding region of E2F1 responsible for inducing apoptosis .\n\nMechanistically , we show that EBNA3C represses E2F1 transcriptional activity via blocking its DNA-binding activity at the responsive promoters of p73 and Apaf-1 apoptosis induced genes , and also facilitates E2F1 degradation in an ubiquitin-proteasome dependent fashion .\n\nMoreover , in response to DNA damage , E2F1 knockdown LCLs exhibited a significant reduction in apoptosis with higher cell-viability .\n\nIn the presence of normal mitogenic stimuli the growth rate of LCLs knockdown for E2F1 was markedly impaired ; indicating that E2F1 plays a dual role in EBV positive cells and that active engagement of the EBNA3C-E2F1 complex is crucial for inhibition of DNA damage induced E2F1-mediated apoptosis .\n\nThis study offers novel insights into our current understanding of EBV biology and enhances the potential for development of effective therapies against EBV associated B-cell lymphomas .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Gastrin stimulates the growth of pancreatic cancer cells through the activation of the cholecystokinin-B receptor ( CCK-BR ) , which has been found to be overexpressed in pancreatic cancer .\n\nIn this study , we proposed that the CCK-BR drives growth of pancreatic cancer ; hence , interruption of CCK-BR activity could potentially be an ideal target for cancer therapeutics .\n\nThe effect of CCK-BR downregulation in the human pancreatic adenocarcinoma cells was examined by utilizing specific CCK-BR-targeted RNA interference reagents .\n\nThe CCK-BR receptor expression was both transiently and stably downregulated by transfection with selective CCK-BR small-interfering RNA or short-hairpin RNA , respectively , and the effects on cell growth and apoptosis were assessed .\n\nCCK-BR downregulation resulted in reduced cancer cell proliferation , decreased DNA synthesis , and cell cycle arrest as demonstrated by an inhibition of G(1) to S phase progression .\n\nFurthermore , CCK-BR downregulation increased caspase-3 activity , TUNEL-positive cells , and decreased X-linked inhibitor of apoptosis protein expression , suggesting apoptotic activity .\n\nPancreatic cancer cell mobility was decreased when the CCK-BR was downregulated , as assessed by a migration assay .\n\nThese results show the importance of the CCK-BR in regulation of growth and apoptosis in pancreatic cancer .\n\nStrategies to decrease the CCK-BR expression and activity may be beneficial for the development of new methods to improve the treatment for patients with pancreatic cancer .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "OBJECTIVE To investigate the effects of CdTe QDs ( cadmium telluride quantum dots ) on oxidative stress and DNA damage of liver cells in mice .\n\nMETHODS Thirty ICR male mice were randomly divided into 5 groups : one negative control ( normal saline ) group .\n\nThree CdTe QDs groups ( exposed by intravenous injection of 0.2 ml of CdTe QDs at the concentration of 3.75 , 37.5 and 375 nmol/ml respectively ) for electron paramagnetic resonance ( EPR ) test , and another positive control group ( exposed by intravenous injection of 0.2 ml of cyclophosphamide 20 mg/ml ) for single cell gel electrophoresis ( SCGE ) test .\n\nAll mice were decapitated 24h after the injection , free radicals and DNA damage of liver cells were detected by EPR and SCGE .\n\nRESULTS The levels of oxygen free radicals detected by EPR were increased with the increase of CdTe QDs .\n\nThe tail length , olive tail moment , tail DNA ( % ) and the ratio of tail/head examined by SCGE were also increased with the increase of the dosage of CdTe QDs ( P < 0.01 ) .\n\nCONCLUSION CdTe QDs could induce oxidative stress and DNA damage of liver cells in mice with a dose-effect relationship .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "The submandibular gland-derived tumor cell line SCA-9 is considered a useful tool to study the signaling pathways involved in proliferation , and their regulation , triggered by different stimuli .\n\nIt is proposed that the non neuronal cholinergic system : acethylcholine , the enzymes that synthesize and degrade it , and the nicotinic and muscarinic receptors , play a key role in tumorigenesis .\n\nHere , we investigate the role of muscarinic receptors in SCA-9 cell proliferation , and the modulation of cholinergic signaling pathways exerted by the nuclear transcription factor \u03baB ( NF-\u03baB ) .\n\nThe activation of cholinergic receptors by carbachol ( 10\u207b\u2079M ) increased cell proliferation ( P<0.001 ) .\n\nThis was prevented by preincubating cells with the muscarinic antagonist atropine but not by mecamylamine , a nicotinic receptor blocker .\n\nPhospholipase C ( PLC)/nitric oxide synthase ( NOS)/arginase pathway is involved in this effect , since carbachol stimulated nitric oxide production , increased NOS2 and NOS3 expressions , urea production , and arginase II expression ( P<0.001 ) .\n\nAlso , phospholipase A\u2082 ( PLA\u2082)/cyclooxygenase ( COX ) pathway is up-regulated in carbachol-induced SCA-9 cell proliferation , because prostaglandin E\u2082 liberation ( P<0.001 ) is increased and COX-1 expression is turned up ( P<0.001 ) .\n\nInteractions between PLC/NOS/arginases and PLA\u2082/COX pathways via its metabolites were detected .\n\nSCA-9 cells exhibit a constitutive activation of NF-\u03baB , which regulates carbachol-induced NOS2 and 3 , arginase II and COX-1 expressions .\n\nIn addition , protein kinase C is involved in the up-regulation of NOS2 and arginase II enzymes induced by carbachol via NF-\u03baB .\n\nIn conclusion , the activation of cholinergic receptors in SCA-9 tumor cells promotes proliferation via muscarinic effector enzymes , and reveals the participation of NF-\u03baB at this step of tumorigenesis .", "output": "Sustaining proliferative signaling" }, { "input": "OBJECTIVES/HYPOTHESIS Head and neck squamous cell carcinoma ( HNSCC ) is a complex disease process involving interactions with carcinoma-associated fibroblasts and endothelial cells .\n\nWe further investigated these relationships by suppressing stromal cell growth through the inhibition of fibroblast growth factor receptor ( FGFR ) .\n\nSTUDY DESIGN Preclinical investigation .\n\nMETHODS HNSCC cell lines ( FADU , OSC19 , Cal27 , SCC1 , SCC5 , SCC22A ) , fibroblast ( HS27 ) , and endothelial cells ( human umbilical vascular endothelial cell ) were cultured individually or in coculture .\n\nProliferation was assessed following treatment with a range of physiologic concentrations of FGFR inhibitor PD173074 .\n\nMice bearing established HNSCC xenografts were treated with PD173074 ( 12 mg/kg ) , and tumor histology was analyzed for stromal composition , proliferation ( Ki67 staining ) , and apoptosis ( TUNEL [ terminal deoxynucleotidyl transferase dUTP nick end labeling ] staining ) .\n\nRESULTS In vitro , inhibition of FGFR with PD173074 dramatically reduced proliferation of fibroblasts and endothelial cells compared to untreated controls .\n\nHowever , HNSCC cell proliferation was not affected by inhibition of FGFR .\n\nWhen cocultured with fibroblasts , HNSCC cells proliferation increased by 15% to 80% ( P < .01 ) .\n\nFurthermore , this fibroblast-enhanced tumor cell growth was suppressed by FGFR inhibition .\n\nAdditionally , treatment of mice bearing HNSCC xenografts with PD173074 resulted in significant growth inhibition ( P < .001 ) .\n\nAdditionally , those tumors from mice treated with PD173074 had a smaller stromal component , decreased proliferation , and increased apoptosis .\n\nCONCLUSIONS Targeting the FGFR pathway in head and neck cancer acts through the stromal components to decrease HNSCC growth in vivo and in vitro .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Certain components of apples have been shown to prevent cancer growth and impede cancer progression .\n\nWe hypothesized that extracted apple polysaccharides ( APs ) might , therefore , have anticancer effects , through a mechanism involving the induction of apoptosis in cancer cells , partly via the NF-\u03baB pathway .\n\nTwo human colorectal cancer ( CRC ) cell lines , HT-29 and SW620 , were exposed to different concentrations of APs ( 0.01 , 0.1 or 1 mg/ml ) .\n\nCell apoptosis was measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay by flow cytometry and incorporation of 5'-bromodeoxyuridine ( BrdU ) into DNA to identify the proliferating cell fraction , using fluorescence microscopy in vitro .\n\nThe protein levels of NF-\u03baB/p65 , I-\u03baB\u03b1 , pI-\u03baB\u03b1 , Bax , Bcl-xl and Bcl-2 were evaluated by western blotting .\n\nThe target sites of APs on CRC cells were assessed by flow cytometry .\n\nAt concentrations of 0.1 and 1 mg/ml , APs showed apoptosis-inducing effects , increased expressions of Bax , nuclear p65 and cytoplasmic pI-\u03baB\u03b1 , and decreased expressions of Bcl-2 , Bcl-xl and cytoplasmic I-\u03baB\u03b1 .\n\nAPs induced apoptosis by slightly activating the NF-\u03baB pathway ; the AP target site could be the Toll-like receptor 4 on the cell membrane .\n\nThese results demonstrate the potential of APs as agents for clinical prevention and treatment of CRC .", "output": "Resisting cell death" }, { "input": "Carboxypeptidase M ( CPM ) targets the basic amino acids arginine and lysine present at the C-terminus of peptides or proteins .\n\nCPM is thought to be involved in inflammatory processes .\n\nThis is corroborated by CPM-mediated trimming and modulation of inflammatory factors , and expression of the protease in inflammatory environments .\n\nSince the function of CPM in and beyond inflammation remains mainly undefined , the identification of natural substrates can aid in discovering the ( patho)physiological role of CPM .\n\nCCL1/I-309 , with its three C-terminal basic amino acids , forms a potential natural substrate for CPM .\n\nCCL1 plays a role not only in inflammation but also in apoptosis , angiogenesis and tumor biology .\n\nEnzymatic processing differently impacts the biological activity of chemokines thereby contributing to the complex regulation of the chemokine system .\n\nThe aim of the present study was to investigate whether ( i ) CCL1/I-309 is prone to trimming by CPM , and ( ii ) the biological activity of CCL1 is altered after C-terminal proteolytic processing .\n\nCCL1 was identified as a novel substrate for CPM in vitro using mass spectrometry .\n\nC-terminal clipping of CCL1 augmented intracellular calcium release mediated by CCR8 but reduced the binding of CCL1 to CCR8 .\n\nIn line with the higher intracellular calcium release , a pronounced increase of the anti-apoptotic activity of CCL1 was observed in the BW5147 cellular model .\n\nCCR8 signaling , CCR8 binding and anti-apoptotic activity were unaffected when CPM was exposed to the carboxypeptidase inhibitor DL-2-mercaptomethyl-3-guanidino-ethylthiopropanoic acid .\n\nThe results of this study suggest that CPM is a likely candidate for the regulation of biological processes relying on the CCL1-CCR8 system .", "output": "Resisting cell death" }, { "input": "OBJECTIVE Expression of N-myc downstream-regulated gene 1 ( NDRG1)/Cap43 is a prognostic indicator of human malignancies according to the tumor type in which it occurs .\n\nWe investigated how NDRG1/Cap43 could affect tumor growth and angiogenesis in non-small-cell lung cancer ( NSCLC ) in vivo using an animal experimental model , and also how it could affect tumor angiogenesis and prognosis in NSCLC patients .\n\nMETHODS AND RESULTS Knockdown of NDRG1/Cap43 in lung cancer cells using a specific small interfering RNA resulted in growth rates in culture that were similar to those of counterpart control cells , but decreased tumor growth rates in vivo markedly .\n\nStable NDRG1/Cap43 knockdown did not induce consistent changes in the expression of Epidermal growth factor receptor ( EGFR ) family proteins and c-Met in two human lung cancer cell lines in vitro .\n\nHowever , cell lines with NDRG1/Cap43 knockdown showed markedly decreased production of the potent angiogenic factors vascular endothelial growth factor-A and interleukin-8 .\n\nCells with knockdown of NDRG1/Cap43 showed marked reduction of tumor-induced angiogenesis .\n\nUsing immunohistochemistry , we examined 182 surgically resected specimens of NSCLC for expression of NDRG1/Cap43 and tumor angiogenesis .\n\nHigh microvessel density in the tumor was significantly associated with nuclear positivity for NDRG1/Cap43 in both adenocarcinoma ( p = 0.003 ) and squamous cell carcinoma ( p=0.041 ) .\n\nFor both adenocarcinoma ( p = 0.031 ) and squamous cell carcinoma ( p=0.034 ) , the survival curve of patients negative for nuclear NDRG1/Cap43 expression differed significantly from that of patients who were positive .\n\nCONCLUSION Therefore , the expression of NDRG1/Cap43 may be predictive of tumor angiogenesis and poor prognosis in NSCLC .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Although the expression of extracellular matrix protein-1 ( ECM1 ) has been documented in several tumor models , the function of ECM1 has remained unclear .\n\nIn this study , expression of ECM1 was detected by real time PCR and immunohistochemistry .\n\nThe role and mechanism of ECM1 overexpression in cholangiocarcinoma ( CCA ) cells were assessed by wound-healing , matrigel invasion assay and Western blotting .\n\nExpression of ECM1 was significantly elevated in CCA tissues than that in adjacent noncancerous , cholangitis and normal bile duct tissues .\n\nIts overexpression was associated with poor differentiation , lymph node metastasis , poor prognosis , and the level of CA199 , MMP-9 , estrogen receptor .\n\nKnockdown of ECM1 suppressed migration and invasion of CCA cells .\n\nUsing PI3K or IKK inhibitor reduced the level of phospho-Akt or phospho-I\u03baB\u03b1 as well as ECM1 .\n\nTaken together , overexpression of ECM1 may contribute to CCA initiation and progression through promoting migration and invasion of CCA cells , its overexpression was associated with Akt/NF-\u03baB signaling axis .", "output": "Activating invasion and metastasis" }, { "input": "Curcumin , a well-known chemopreventive agent from turmeric , inhibits the expression of several oncogenes and cell proliferation genes in tumor cells .\n\nThis study aims to understand the precise molecular mechanism by which curcumin exerts its effects on retinoblastoma cells , by performing whole genome microarray analysis to determine the gene expression profiles altered by curcumin treatment .\n\nCurcumin suppressed cell viability and altered the cell cycle of retinoblastoma cells .\n\nWe identified 903 downregulated genes and 1,319 upregulated genes when compared with the control cells after treatment with 20 \u03bcM curcumin concentration for 48 h .\n\nThese genes were grouped into respective functional categories according to their biological function .\n\nWe found that curcumin regulated the expression of genes that are involved in the regulation of apoptosis , tumor suppressor , cell-cycle arrest , transcription factor , and angiogenesis .\n\nQuantitative real-time polymerase chain reaction ( qRT-PCR ) analysis was used to validate the results of genome array , and the results were consistent with the obtained data .\n\nIn conclusion , treatment of curcumin affects the expression of genes involved in various cellular functions and plays an important role in tumor metastasis and apoptosis .\n\nThus , curcumin might be an effective chemopreventive agent for retinoblastoma cancer .", "output": "Activating invasion and metastasis, Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Adenosine receptors ( A1 , A2A , A2B , A3 ) play an important role in the regulation of growth , proliferation and death of cancer and normal cells .\n\nWe recently showed the expression profile of A2A and A2B receptors in normal and tumor breast tissues .\n\nIn the present study , we used semiquantitative RT-PCR to measure the A1 and A3 gene expression levels in normal and tumor breast tissues .\n\nMETHODS Breast tumors ( n = 18 ) and non-neoplastic mammary tissues ( n = 10 ) were collected and histologically confirmed to be neoplastic or non-neoplastic , respectively .\n\nTotal RNA was extracted and reverse transcribed into cDNA , and PCR was performed under optimized condition for each receptor subtype .\n\nAmplification of beta-actin mRNA served as control for RT-PCR .\n\nThe PCR products were separated on 1.7% agarose gels .\n\nThe intensity of the bands was quantitated with ImageJ software after normalization against beta-actin expression .\n\nRESULTS All breast tumor and normal tissue specimens expressed A1 and A3 adenosine receptor transcripts .\n\nHowever , we observed that the expression level of the A3 receptor in tumor tissues was 1.27-fold that of normal tissues , whereas there was no significant difference between the expression levels of A1 in normal and tumor tissues .\n\nCONCLUSIONS Interestingly , the results of the present study indicate that breast tumors exhibit a higher level of A3 transcripts ( than normal tissues ) and support the possible key role of A3 adenosine receptor in tumor development .\n\nHowever , further studies based on real-time quantitative RT-PCR are needed to identify the exact gene expression levels .", "output": "Sustaining proliferative signaling" }, { "input": "Manipulating acetylation status of key gene targets is likely to be crucial for effective cancer therapy .\n\nIn this study , we utilized green tea catechins , epicatechin ( EC ) , epigallocatechin ( EGC ) and epigallocatechin-3-gallate ( EGCG ) to examine the regulation of androgen receptor acetylation in androgen-dependent prostate cancer cells by histone acetyl-transferase ( HAT ) activity .\n\nEC , EGC and EGCG induced prostate cancer cell death , suppressed agonist-dependent androgen receptor ( AR ) activation and AR-regulated gene transcription .\n\nThese results demonstrated a similar tendency to HAT inhibitory activities ; EGCG>EGC>EC .\n\nThe strongest HAT inhibitor among them , EGCG ( 50 \ufffdM ) , downregulated AR acetylation and finally , AR protein translocation to nucleus from the cytoplasmic compartment was effectively inhibited in the presence of the agonist .\n\nThese results suggest another mechanism to develop effective therapeutics based on green tea catechins .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Furan , a potent rodent liver carcinogen , is found in many cooked food items and thus represents a human cancer risk .\n\nMechanisms for furan carcinogenicity were investigated in male F344 rats using the in vivo Comet and micronucleus assays , combined with analysis of histopathological and gene expression changes .\n\nIn addition , formamidopyrimidine DNA glycosylase ( Fpg ) and endonuclease III ( EndoIII)-sensitive DNA damage was monitored as a measure of oxidative DNA damage .\n\nRats were treated by gavage on four consecutive days with 2 , 4 , and 8mg/kg bw furan , doses that were tumorigenic in 2-year cancer bioassays , and with two higher doses , 12 and 16mg/kg .\n\nRats were killed 3h after the last dose , a time established as producing maximum levels of DNA damage in livers of furan-treated rats .\n\nLiver Comet assays indicated that both DNA strand breaks and oxidized purines and pyrimidines increased in a near-linear dose-responsive fashion , with statistically significant increases detected at cancer bioassay doses .\n\nNo DNA damage was detected in bone marrow , a non-target tissue for cancer , and peripheral blood micronucleus assays were negative .\n\nHistopathological evaluation of liver from furan-exposed animals produced evidence of inflammation , single-cell necrosis , apoptosis , and cell proliferation .\n\nIn addition , genes related to apoptosis , cell-cycle checkpoints , and DNA-repair were expressed at a slightly lower level in the furan-treated livers .\n\nAlthough a mixed mode of action involving direct DNA binding cannot be ruled out , the data suggest that furan induces cancer in rat livers mainly through a secondary genotoxic mechanism involving oxidative stress , accompanied by inflammation , cell proliferation , and toxicity .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Evading growth suppressors, Resisting cell death" }, { "input": "Many cancer cells metabolize glucose preferentially via pyruvate to lactate instead to CO(2) and H(2)O ( oxidative phosphorylation ) even in the presence of oxygen ( Warburg effect ) .\n\nDichloroacetate ( DCA ) is a drug which is able to shift pyruvate metabolism from lactate to acetyl-CoA ( tricarboxylic acid cycle ) by indirect activation of pyruvate dehydrogenase ( PDH ) .\n\nThis can subsequently lead to an increased flow of oxygen in the respiratory chain , associated with enhanced generation of reactive oxygen species ( ROS ) which may cause apoptosis .\n\nIn order to investigate if DCA may be suitable for neuroblastoma therapy , it was investigated on three human neuroblastoma cell lines whether DCA can reduce lactate production and enhance oxygen consumption .\n\nThe data show , that DCA ( in the low millimolar range ) is able to reduce lactate production , but there was only a slight shift to increased oxygen consumption and almost no effect on cell vitality , proliferation and apoptosis of the three cell lines investigated .\n\nTherefore , DCA at low millimolar concentrations seems to be only of minor efficacy for neuroblastoma treatment .", "output": "Cellular energetics, Resisting cell death" }, { "input": "BACKGROUND Brain-metastatic breast cancer ( BMBC ) is increasing and poses a severe clinical problem because of the lack of effective treatments and because the underlying molecular mechanisms are largely unknown .\n\nRecent work has demonstrated that deregulation of epidermal growth factor receptor ( EGFR ) may correlate with BMBC progression .\n\nHowever , the exact contribution that EGFR makes to BMBC remains unclear .\n\nMETHODS The role of EGFR in BMBC was explored by serial analyses in a brain-trophic clone of human MDA-MB-231 breast carcinoma cells ( 231-BR cells ) .\n\nEGFR expression was inhibited by stable short-hairpin RNA transfection or by the kinase inhibitor erlotinib , and it was activated by heparin-binding epidermal growth factor-like growth factor ( HB-EGF ) .\n\nCell growth and invasion activities also were analyzed in vitro and in vivo .\n\nRESULTS EGFR inhibition or activation strongly affected 231-BR cell migration/invasion activities as assessed by an adhesion assay , a wound-healing assay , a Boyden chamber invasion assay , and cytoskeleton staining .\n\nAlso , EGFR inhibition significantly decreased brain metastases of 231-BR cells in vivo .\n\nSurprisingly , changes to EGFR expression affected cell proliferation activities less significantly as determined by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide ( MTT ) assay , an anchorage-independent growth assay , and cell cycle analysis .\n\nImmunoblot analysis suggested that EGFR drives cells ' invasiveness capability mainly through phosphoinositide 3-kinase/protein kinase B and phospholipase C \u03b3 downstream pathways .\n\nIn addition , EGFR was involved less in proliferation because of the insensitivity of the downstream mitogen-activated protein kinase pathway .\n\nCONCLUSIONS The current results indicated that EGFR plays more important roles in cell migration and invasion to the brain than in cell proliferation progression on 231-BR cells , providing new evidence of the potential value of EGFR inhibition in treating BMBC .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "The Salmonella type III secretion system ( T3SS ) efficiently translocates heterologous proteins into the cytosol of eukaryotic cells .\n\nThis leads to an antigen-specific CD8 T-cell induction in mice orally immunized with recombinant Salmonella .\n\nRecently , we have used Salmonella's T3SS as a prophylactic and therapeutic intervention against a murine fibrosarcoma .\n\nIn this study , we constructed a recombinant Salmonella strain translocating the immunogenic H-2D(b)-specific CD8 T-cell epitope VILTNPISM ( KDR2 ) from the murine vascular endothelial growth factor receptor 2 ( VEGFR2 ) .\n\nVEGFR2 is a member of the tyrosine protein kinase family and is upregulated on proliferating endothelial cells of the tumor vasculature .\n\nAfter single orogastric vaccination , we detected significant numbers of KDR2-tetramer-positive CD8 T cells in the spleens of immunized mice .\n\nThe efficacy of these cytotoxic T cells was evaluated in a prophylactic setting to protect mice from challenges with B16F10 melanoma cells in a flank tumor model , and to reduce dissemination of spontaneous pulmonary melanoma metastases .\n\nVaccinated mice revealed a reduction of angiogenesis by 62% in the solid tumor and consequently a significant decrease of tumor growth as compared to non-immunized mice .\n\nMoreover , in the lung metastasis model , immunization with recombinant Salmonella resulted in a reduction of the metastatic melanoma burden by approximately 60% .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "BACKGROUND Colorectal cancer is one of the leading causes of cancer-related death throughout the world , and the risk to develop this malignant disease seems to be associated with long-term cigarette smoking .\n\nNicotine , one of the major components of cigarette smoking , can stimulate cell proliferation and suppress apoptosis both in normal cells and in several human cancer cell lines derived from various organs .\n\nHowever , although nicotine appears to have a role in stimulating cell proliferation of colon cancer cells , there is no information on its role in inhibiting apoptosis in these cells .\n\nMATERIALS AND METHODS Human colorectal cancer cell lines Caco-2 and HCT-8 were treated with 1 \u03bcM nicotine alone or in combination with 1 \u03bcM \u03b1-BTX in complete or in serum free medium .\n\nCell proliferation and apoptosis were determined by cell count performed with a cell counter and by cytofluorimetric assay respectively .\n\nPI3K/Akt and PKC/ERK1/2 pathways , survivin , and P-Bcl2 ( Ser70 ) were investigated by Western blot analysis .\n\nRESULTS Nicotine induced an increase in cell proliferation and a decrease of apoptosis in Caco-2 and HCT-8 cells .\n\nBoth cell growth and apoptosis appear to be mediated by \u03b17-nicotinic acetylcholine receptors , since treatment with \u03b1-Bungarotoxin inhibited these processes .\n\nNicotine induced a statistically significant increase in the expression of PI3K and in P-Akt/Akt ratio as well as in the expression of PKC , ERK1/2 , survivin , and P-Bcl2 ( Ser70 ) in both cell lines .\n\nCONCLUSIONS Nicotine , contained in cigarette smoking , could participate in colon cancer development and progression by stimulating cell proliferation and suppressing physiological apoptosis .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Neurofibromatosis type 2 patients develop schwannomas , meningiomas and ependymomas resulting from mutations in the tumor suppressor gene , NF2 , encoding a membrane-cytoskeleton adapter protein called merlin .\n\nMerlin regulates contact inhibition of growth and controls the availability of growth factor receptors at the cell surface .\n\nWe tested if microtubule-based vesicular trafficking might be a mechanism by which merlin acts .\n\nWe found that schwannoma cells , containing merlin mutations and constitutive activation of the Rho/Rac family of GTPases , had decreased intracellular vesicular trafficking relative to normal human Schwann cells .\n\nIn Nf2-/- mouse Schwann ( SC4 ) cells , re-expression of merlin as well as inhibition of Rac or its effector kinases , MLK and p38(SAPK) , each increased the velocity of Rab6 positive exocytic vesicles .\n\nConversely , an activated Rac mutant decreased Rab6 vesicle velocity .\n\nVesicle motility assays in isolated squid axoplasm further demonstrated that both mutant merlin and active Rac specifically reduce anterograde microtubule-based transport of vesicles dependent upon the activity of p38(SAPK) kinase .\n\nTaken together , our data suggest loss of merlin results in the Rac-dependent decrease of anterograde trafficking of exocytic vesicles , representing a possible mechanism controlling the concentration of growth factor receptors at the cell surface .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Bladder cancer is one of the most common tumors of the genitourinary tract ; however , the molecular events underlying growth and invasion of the tumor remain unclear .\n\nHere , role of the CXCR7 receptor in bladder cancer was further explored .\n\nCXCR7 protein expression was examined using high-density tissue microarrays .\n\nExpression of CXCR7 showed strong epithelial staining that correlated with bladder cancer progression .\n\nIn vitro and in vivo studies in bladder cancer cell lines suggested that alterations in CXCR7 expression were associated with the activities of proliferation , apoptosis , migration , invasion , angiogenesis and tumor growth .\n\nMoreover , CXCR7 expression was able to regulate expression of the proangiogenic factors IL-8 or VEGF , which may involve in the regulation of tumor angiogenesis .\n\nFinally , we found that signaling by the CXCR7 in bladder cancer cells activates AKT , ERK and STAT3 pathways .\n\nThe AKT and ERK pathways may reciprocally regulate , which are responsible for in vitro and in vivo epithelial to mesenchymal transition ( EMT ) process of bladder cancer .\n\nSimultaneously targeting the two pathways by using U0126 and LY294002 inhibitors or using CCX733 , a selective CXCR7 antagonist drastically reduced CXCR7-induced EMT process .\n\nTaken together , our data show for the first time that CXCR7 plays a role in the development of bladder cancer .\n\nTargeting CXCR7 or its downstream-activated AKT and ERK pathways may prove beneficial to prevent metastasis and provide a more effective therapeutic strategy for bladder cancer .", "output": "Activating invasion and metastasis, Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Recent data indicate the Signal Transducer and Activator of Transcription 3 ( STAT3 ) pathway is required for VEGF production and angiogenesis in various types of cancers .\n\nSTAT3 inhibitors have been shown to reduce tumor microvessel density in tumors but a direct anti-angiogenic activity has not been described .\n\nMETHODOLOGY/PRINCIPAL FINDINGS We investigated the direct action of a small molecule inhibitor of STAT3 ( LLL12 ) in human umbilical cord vascular endothelial cells ( HUVECs ) in vitro , in a Matrigel model for angiogenesis in vivo , and its antitumor activity in a xenograft model of osteosarcoma .\n\nLLL12 ( 100 nM ) significantly inhibited VEGF-stimulated STAT3 phosphorylation in HUVECs , reduced their proliferation/migration and inhibited VEGF-induced tube formation .\n\nMorphologic analysis of LLL12 treated HUVECs demonstrated marked changes in actin/tubulin distribution and bundling .\n\nIn scid mice , LLL12 reduced microvessel invasion into VEGF-infused Matrigel plugs by \u223c90% at a dose of 5 mg/kg daily .\n\nFollowing a period of tumor progression ( 2 weeks ) , LLL12 completely suppressed further growth of established OS-1 osteosarcoma xenografts .\n\nPharmacodynamic studies showed robust phosphorylated STAT3 in control tumors , whereas phospho-STAT3 was not detected in LLL12-treated OS-1 tumors .\n\nTreated tumors demonstrated decreased proliferation ( Ki67 staining ) , and decreased microvessel density ( CD34 staining ) , but no significant increase in apoptosis ( TUNEL staining ) , relative to controls .\n\nAssay of angiogenic factors , using an antibody array , showed VEGF , MMP-9 , Angiopoietin1/2 , Tissue Factor and FGF-1 expression were dramatically reduced in LLL12-treated tumors compared to control tumors .\n\nCONCLUSIONS These findings provide the first evidence that LLL12 effectively inhibits tumor angiogenesis both in vitro and in vivo .", "output": "Inducing angiogenesis, Resisting cell death, Activating invasion and metastasis" }, { "input": "BACKGROUND Tissue factor ( TF ) , an initiator of blood coagulation , participates in cancer progression and metastasis .\n\nWe recently found that inhibition of MAPK/ERK upregulated both full length TF ( flTF ) and soluble isoform TF ( asTF ) gene expression and cell-associated TF activity in breast cancer MDA-MB-231 cells .\n\nWe explored the possible mechanisms , especially the possible interaction with EGFR and PI3K/Akt pathways .\n\nMETHODS A plasmid containing TF promoter -2174\u2009 plus luciferase reporter gene was introduced into MDA-MB-231 cells to evaluate TF promoter activity .\n\nIn order to study the interaction of these pathways , ERK inhibitor ( PD98059 ) , PI3K inhibitors ( LY294002 , wortmannin ) , Akt inhibitor ( A6730 ) , and EGFR inhibitor ( erlotinib ) as well as the corresponding siRNAs were used to treat MDA-MB-231 cells , and ovarian cancer OVCAR-3 and SKOV-3 cells .\n\nQuantitative PCR and western blot were used to determine TF expression .\n\nOne stage clotting assays were used to measure pro-coagulation activity of the MDA-MB-231 cells .\n\nRESULTS We show that PI3K inhibitors LY294002 , wortmannin and A6730 significantly inhibited TF promoter activity , and reduced TF mRNA and protein levels due to the inhibition of Akt phosphorylation .\n\nIn contrast , ERK inhibitor PD98059 and ERK siRNA enhanced TF promoter activity by 2.5 fold and induced an increase in TF mRNA and protein levels in a dose dependent manner in these cells .\n\nThe PI3K/Akt pathway was shown to be involved in PD98059-induced TF expression because the induction was inhibited by PI3K/Akt inhibitors .\n\nMost interestingly , the EGFR inhibitor erlotinib and EGFR siRNA also significantly suppressed PD98059- or ERK siRNA-induced TF promoter activity and TF protein expression .\n\nSimilar results were found with ovarian cancer cells SKOV-3 and OVCAR-3 .\n\nFurthermore , in MDA-MB-231 , mRNA levels of asTF were regulated in a similar way to that of TF in response to the cell treatment .\n\nCONCLUSIONS This study showed a regulatory mechanism in which MAPK/ERK signals inhibit EGFR/PI3K/Akt-mediated TF expression in breast cancer MDA-MB-231 cells .\n\nThe same regulation was observed in ovarian cancer OVCAR-3 and SKOV-3 cells .\n\nInterestingly , we observed that both flTF and asTF could be regulated in a parallel manner in MDA-MB-231 .\n\nAs the PI3K/Akt pathway and EGFR regulate TF expression in cancer cells , targeting these signaling components is expected to potentially inhibit TF expression-associated tumor progression .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND The isothiocyanate sulforaphane ( SFN ) possesses interesting anticancer activities .\n\nHowever , recent studies reported that SFN promotes the formation of reactive oxygen species ( ROS ) as well as DNA breakage .\n\nMETHODOLOGY/PRINCIPAL FINDINGS We investigated whether SFN is able to damage RNA , whose loss of integrity was demonstrated in different chronic diseases .\n\nConsidering the ability of SFN to protect from genotoxicity , we also examined whether SFN is able to protect from RNA damage induced by different chemicals ( doxorubicin , spermine , S-nitroso-N-acetylpenicillamine , H(2)O(2) ) .\n\nWe observed that SFN was devoid of either RNA damaging and RNA protective activity in human leukemic cells .\n\nIt was able to potentiate the RNA damage by doxorubicin and spermine .\n\nIn the first case , the effect was attributable to its ability of modulating the bioreductive activation of doxorubicin .\n\nFor spermine , the effects were mainly due to its modulation of ROS levels produced by spermine metabolism .\n\nAs to the cytotoxic relevance of the RNA damage , we found that the treatment of cells with a mixture of spermine or doxorubicin plus SFN increased their proapoptotic potential .\n\nThus it is conceivable that the presence of RNA damage might concur to the overall toxic response induced by a chemical agent in targeted cells .\n\nCONCLUSIONS/SIGNIFICANCE Since RNA is emerging as a potential target for anticancer drugs , its ability to enhance spermine- and doxorubicin-induced RNA damage and cytotoxicity could represent an additional mechanism for the potentiating effects of SFN associated with anticancer drugs .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "AIM We studied expression of molecules of the vascular endothelial growth factor ( VEGF ) pathway and its relation to vascularization , cell proliferation and patient outcome in recurring non-anaplastic meningioma .\n\nWe studied 29 tumor specimens of 8 patients with recurring meningiomas and of 8 age- and gender-matched control patients with non-recurring meningiomas ( including meningothelial , transitional , fibroblastic and atypical subtypes ) using immunohistochemistry and in-situ hybridization .\n\nRESULTS VEGF protein , VEGF-mRNA , VEGF receptor ( VEGFR)-1 mRNA , VEGFR-2 mRNA and hypoxia-inducible factor ( HIF)-1-\u03b1 protein were expressed in 27/29 ( 93% ) , 20/27 ( 74% ) , 9/27 ( 33.3% ) , 12/27 ( 44.4% ) and 5/29 ( 17.2% ) specimens , respectively .\n\nVEGFR- 2 mRNA expression was found in 6/8 tumors extracted at first operation in patients with recurring tumors and in none of the control cases ( p = 0.007 ) .\n\nMicrovessel density ( MVD ) and Ki-67 index values were generally higher in meningiomas with expression of angiogenic factors .\n\nThe association of high Ki-67 index values with VEGF-mRNA expression was significant ( p = 0.04 ) .\n\nTime to recurrence was shorter in patients with high MVD than in patients with low MVD ( p = 0.027 ) .\n\nCONCLUSIONS High MVD correlates with unfavorable prognosis in our series of recurring meningioma .\n\nVEGF and its receptors are frequently expressed in meningiomas and seem important for tumor growth and recurrence .\n\nThus , anti-VEGF therapy in aggressive meningioma seems rational from a pathobiological point of view .", "output": "Inducing angiogenesis" }, { "input": "Even when patients with nonsmall cell lung cancer undergo surgical resection at an early stage , recurrent disease often impairs the clinical outcome .\n\nThere are numerous causes potentially responsible for a relapse of the disease , one of them being extensive angiogenesis .\n\nThe balance of at least two systems , VEGF VEGFR and Ang Tie , regulates vessel formation .\n\nThe aim of this study was to determine the impact of surgery on the plasma levels of the main angiogenic factors during the first month after surgery in nonsmall cell lung cancer patients .\n\nThe study group consisted of 37 patients with stage I nonsmall cell lung cancer .\n\nPlasma concentrations of Ang1 , Ang2 , sTie2 , VEGF , and sVEGF R1 were evaluated by ELISA three times : before surgical resection and on postoperative days 7 and 30 .\n\nThe median of Ang2 and VEGF concentrations increased on postoperative day 7 and decreased on day 30 .\n\nOn the other hand , the concentration of sTie2 decreased on the 7th day after resection and did not change statistically later on .\n\nThe concentrations of Ang1 and sVEGF R1 did not change after the surgery .\n\nLung cancer resection results in proangiogenic plasma protein changes that may stimulate tumor recurrences and metastases after early resection .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "The phosphoinositide 3-kinase ( PI3K)/AKT and RAF/MEK/ERK signaling pathways are activated in a wide range of human cancers .\n\nIn many cases , concomitant inhibition of both pathways is necessary to block proliferation and induce cell death and tumor shrinkage .\n\nSeveral feedback systems have been described in which inhibition of one intracellular pathway leads to activation of a parallel signaling pathway , thereby decreasing the effectiveness of single-agent targeted therapies .\n\nIn this study , we describe a feedback mechanism in which MEK inhibition leads to activation of PI3K/AKT signaling in EGFR and HER2-driven cancers .\n\nWe found that MEK inhibitor-induced activation of PI3K/AKT resulted from hyperactivation of ERBB3 as a result of the loss of an inhibitory threonine phosphorylation in the conserved juxtamembrane domains of EGFR and HER2 .\n\nMutation of this amino acid led to increased ERBB receptor activation and upregulation of the ERBB3/PI3K/AKT signaling pathway , which was no longer responsive to MEK inhibition .\n\nTaken together , these results elucidate an important , dominant feedback network regulating central oncogenic pathways in human cancer .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Apigenin , a natural plant flavone , may have chemopreventive and therapeutic potentials for anti-inflammatory , antioxidant , and anti-cancer .\n\nNevertheless , the anti-tumor effect of apigenin on human head and neck squamous cell carcinoma ( HNSCC ) is not fully understood .\n\nMETHODS The antioxidant capacity and protective effects of apigenin against oxidative stress in murine normal embryonic liver BNLCL2 cells are examined .\n\nCell viability , morphologic change , clonogenic survival , cell cycle distribution , reactive oxygen species ( ROS ) production , glutathione formation , and death receptors- and Bcl-2-mediated caspase pathways of HNSCC SCC25 cells and A431 cells with apigenin are investigated .\n\nRESULTS Apigenin inhibits the growth of SCC25 and A431 cells and induces cell cycle arrest in the G2/M phase .\n\nApigenin has an antioxidant capacity as well as the ability to inhibit lipid peroxidation .\n\nIt protects BNLCL2 cells against oxidative damage , and is potentially able to prevent cancer .\n\nApigenin increases intracellular ROS levels and reduces levels of glutathione ; it also induces cell apoptosis via tumor necrosis factor receptor ( TNF-R)- , TNF-related apoptosis-inducing ligand receptor ( TRAIL-R)- , and Bcl-2-mediated caspase-dependent cell death pathways in SCC25 cells .\n\nThe combination of apigenin with 5-fluorouracil ( 5-Fu ) or cisplatin induces the dramatic death of SCC25 cells .\n\nCONCLUSIONS Apigenin induces SCC25 cell apoptosis via the up-regulation of both TNF-R and TRAIL-R signaling pathways , and has a synergistic effect on the inhibition of cell proliferation in combination with 5-Fu or cisplatin .\n\nGENERAL SIGNIFICANCE These analytical findings suggest that apigenin may be a good therapeutic agent against HNSCC cells .", "output": "Genomic instability and mutation, Tumor promoting inflammation, Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND Peritoneal transport status is important not only for prescription , but also as a prognostic index .\n\nFlt-1 and Flk-1 , the major vascular endothelial growth factor receptors involved in angiogenesis and hyperpermeability , may play a potent role in determining peritoneal transport characteristics .\n\nHowever , the relationship between them has not been studied to date .\n\nWe hypothesized that Flt-1 and Flk-1 expression in the peritoneal vasculature of uremic patients could be closely related to baseline peritoneal transport status .\n\nMETHODS Thirty-six new patients without a previous history of peritonitis were enrolled .\n\nClinical parameters such as age , sex , height , weight , causes of renal failure , and residual renal function were assessed .\n\nParietal peritoneal biopsies were obtained during implantation of peritoneal dialytic catheters .\n\nFlt-1 and Flk-1 were semi-quantitatively evaluated by immunohistochemical staining .\n\nPeritoneal microvascular density ( MVD ) was counted .\n\nWithin 6 weeks after commencing peritoneal dialysis , a standard peritoneal equilibration test was performed , and the dialysate-to-plasma concentration ratio for creatinine at 4 h ( D4/P Cr ) was determined .\n\nThe patients were divided into two groups based on the D4/P Cr : more than 0.65 ( Group H , n = 22 ) and less than or equal to 0.65 ( Group L , n = 14 ) .\n\nThe 24-h peritoneal protein excretion ( PPE ) was assayed .\n\nFlt-1 and Flk-1 were correlated with peritoneal MVD , D4/P Cr , and PPE .\n\nRESULTS Flt-1 and Flk-1 were detected in the peritoneal vasculature of uremic patients .\n\nFlt-1 expression was similar between the two groups , but Flk-1 expression in Group H was significantly higher than that in Group L ( p = 0.001 ) .\n\nFlt-1 expression did not show significant correlations with peritoneal MVD , D4/P Cr , and PPE .\n\nHowever , Flk-1 expression showed significant correlations with the above three parameters ( p < 0.001 for all ) .\n\nCONCLUSIONS For the first time , the expressions of Flt-1 and Flk-1 in peritoneal vasculature of uremic patients were detected .\n\nFlk-1 expression in peritoneal vasculature of uremic patients is closely correlated with the number of peritoneal microvessels , peritoneal small solute transport rate , and PPE .\n\nOur findings strongly suggest that Flk-1 may be a crucial determinant of baseline peritoneal transport characteristics .\n\nFurther interventional studies are needed .", "output": "Inducing angiogenesis" }, { "input": "Cyclin E2 , but not cyclin E1 , is included in several gene signatures that predict disease progression in either tamoxifen-resistant or metastatic breast cancer .\n\nWe therefore examined the role of cyclin E2 in antiestrogen resistance in vitro and its potential for therapeutic targeting through cyclin-dependent kinase ( CDK ) inhibition .\n\nHigh expression of CCNE2 , but not CCNE1 , was characteristic of the luminal B and HER2 subtypes of breast cancer and was strongly predictive of shorter distant metastasis-free survival following endocrine therapy .\n\nAfter antiestrogen treatment of MCF-7 breast cancer cells , cyclin E2 mRNA and protein were downregulated and cyclin E2-CDK2 activity decreased .\n\nHowever , this regulation was lost in tamoxifen-resistant ( MCF-7 TAMR ) cells , which overexpressed cyclin E2 .\n\nExpression of either cyclin E1 or E2 in T-47D breast cancer cells conferred acute antiestrogen resistance , suggesting that cyclin E overexpression contributes to the antiestrogen resistance of tamoxifen-resistant cells .\n\nEctopic expression of cyclin E1 or E2 also reduced sensitivity to CDK4 , but not CDK2 , inhibition .\n\nProliferation of tamoxifen-resistant cells was inhibited by RNAi-mediated knockdown of cyclin E1 , cyclin E2 , or CDK2 .\n\nFurthermore , CDK2 inhibition of E-cyclin overexpressing cells and tamoxifen-resistant cells restored sensitivity to tamoxifen or CDK4 inhibition .\n\nCyclin E2 overexpression is therefore a potential mechanism of resistance to both endocrine therapy and CDK4 inhibition .\n\nCDK2 inhibitors hold promise as a component of combination therapies in endocrine-resistant disease as they effectively inhibit cyclin E1 and E2 overexpressing cells and enhance the efficacy of other therapeutics .", "output": "Sustaining proliferative signaling" }, { "input": "We investigated the effects of valproic acid ( VPA ) , a histone deacetylase inhibitor , in combination with hydralazine , a DNA methylation inhibitor , on the expression of cell-surface Fas and MHC-class I-related chain molecules A and B ( MICA and B ) , the ligands of NKG2D which is an activating receptor of NK cells , and on production of their soluble forms in HOS , U-2 OS and SaOS-2 human osteosarcoma cell lines .\n\nWe also examined the susceptibility of these cells to Fas- and NK cell-mediated cell death .\n\nVPA did not increase the expression of Fas on the surface of osteosarcoma cells , while hydralazine did , and the combination of VPA with hydralazine increased the expression of cell-surface Fas .\n\nIn contrast , the combination of VPA with hydralazine did not increase the production of soluble Fas by osteosarcoma cells .\n\nBoth VPA and hydralazine increased the expression of cell-surface MICA and B in osteosarcoma cells , and their combination induced a greater increase in their expression .\n\nVPA inhibited the production of both soluble MICA and MICB by osteosarcoma cells while hydralazine produced no effect .\n\nBoth VPA and hydralazine enhanced the susceptibility of osteosarcoma cells to Fas- and NK cell-mediated cell death and the combination of VPA with hydralazine further enhanced the effects .\n\nThe present results suggest that combined administration of VPA and hydrazine is valuable for enhancing the therapeutic effects of immunotherapy for osteosarcomas .", "output": "Resisting cell death" }, { "input": "Surgery is the most effective therapy for cancer in the United States , but disease still recurs in more than 40% of patients within 5 years after resection .\n\nChemotherapy is given postoperatively to prevent relapses ; however , this approach has had marginal success .\n\nAfter surgery , recurrent tumors depend on rapid neovascular proliferation to deliver nutrients and oxygen .\n\nPhosphatidylserine ( PS ) is exposed on the vascular endothelial cells in the tumor microenvironment but is notably absent on blood vessels in normal tissues .\n\nThus , PS is an attractive target for cancer therapy after surgery .\n\nSyngeneic mice bearing TC1 lung cancer tumors were treated with mch1N11 ( a novel mouse chimeric monoclonal antibody that targets PS ) , cisplatin ( cis ) , or combination after surgery .\n\nTumor relapses and disease progression were decreased 90% by combination therapy compared with a 50% response rate for cis alone ( P = .02 ) .\n\nMice receiving postoperative mch1N11 had no wound-related complications or added systemic toxicity in comparison to control animals .\n\nMechanistic studies demonstrated that the effects of mch1N11 were associated with a dense infiltration of inflammatory cells , particularly granulocytes .\n\nThis strategy was independent of the adaptive immune system .\n\nTogether , these data suggest that vascular-targeted strategies directed against exposed PS may be a powerful adjunct to postoperative chemotherapy in preventing relapses after cancer surgery .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "Oxidative stress is the main etiological factor behind the pathogenesis of various diseases including inflammation , cancer , cardiovascular and neurodegenerative disorders .\n\nDue to the spin trapping abilities and various pharmacological properties of nitrones , their application as therapeutic agent has been gaining attention .\n\nThough the antioxidant properties of the nitrones are well known , the mechanism by which they modulate the cellular defense machinery against oxidative stress is not well investigated and requires further elucidation .\n\nHere , we have investigated the mechanisms of cytoprotection of the nitrone spin traps against oxidative stress in bovine aortic endothelial cells ( BAEC ) .\n\nCytoprotective properties of both the cyclic nitrone 5,5-dimethyl-pyrroline N-oxide ( DMPO ) and linear nitrone \u03b1-phenyl N-tert-butyl nitrone ( PBN ) against H\u2082O\u2082-induced cytotoxicity were investigated .\n\nPreincubation of BAEC with PBN or DMPO resulted in the inhibition of H\u2082O\u2082-mediated cytotoxicity and apoptosis .\n\nNitrone-treatment resulted in the induction and restoration of phase II antioxidant enzymes via nuclear translocation of NF-E2-related factor 2 ( Nrf-2 ) in oxidatively-challenged cells .\n\nFurthermore , the nitrones were found to inhibit the mitochondrial depolarization and subsequent activation of caspase-3 induced by H\u2082O\u2082 .\n\nSignificant down-regulation of the pro-apoptotic proteins p53 and Bax , and up-regulation of the anti-apoptotic proteins Bcl-2 and p-Bad were observed when the cells were preincubated with the nitrones prior to H\u2082O\u2082-treatment .\n\nIt was also observed that Nrf-2 silencing completely abolished the protective effects of nitrones .\n\nHence , these findings suggest that nitrones confer protection to the endothelial cells against oxidative stress by modulating phase II antioxidant enzymes and subsequently inhibiting mitochondria-dependent apoptotic cascade .", "output": "Resisting cell death" }, { "input": "BACKGROUND Special AT-rich binding protein-1 ( SATB1 ) reprograms chromatin organization and transcription profiles to promote tumour growth and metastasis .\n\nAIMS This study aimed to confirm the effects of SATB1 on the growth and metastasis of liver cancer and its specific regulation mechanism .\n\nMETHODS SATB1 expression was evaluated in human hepatoma tissue , adjacent noncancerous tissue and seven kinds of liver cancer cell lines .\n\nCell cycle , cell proliferation , apoptosis and epithelial-mesenchymal transition ( EMT ) was investigated after enhanced or silenced expression of SATB1 .\n\nThe regulatory action of SATB1 on the expression of genes that are known to regulate cell cycle progression , apoptosis and EMT and the specific apoptotic pathway on which it acts were further analysed .\n\nNude mice that received subcutaneous implantation were used to study the effects of SATB1 on tumour growth in vivo .\n\nRESULTS Our data show that the high expression of SATB1 was observed in the human hepatocellular carcinoma tissue ( 26/45 ) and liver cancer cell lines with high metastatic potential .\n\nSATB1 upregulated CDK4 and downregulated p16 ( INK ) ( 4A ) to promote cell cycle progression and cell proliferation and prevented apoptosis by inhibiting the FADD-caspase-8-caspase-3 death receptor-mediated apoptosis pathway .\n\nSATB1 also induced EMT concomitant with increased expression of Snail1 , Slug , Twist and vimentin and decreased expression of E-cadherin , tight junction protein ZO-1 and desmoplakin .\n\nSATB1 promoted the growth of tumour in vivo .\n\nCONCLUSION These data suggest that the SATB1 gene may play an important role in the development and progression of liver cancer by regulation of genes related to cell cycle progression , apoptosis and EMT .", "output": "Evading growth suppressors, Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Elevated aerobic glycolysis in cancer cells ( the Warburg effect ) may be attributed to respiration injury or mitochondrial dysfunction , but the underlying mechanisms and therapeutic significance remain elusive .\n\nHere we report that induction of mitochondrial respiratory defect by tetracycline-controlled expression of a dominant negative form of DNA polymerase \\u03b3 causes a metabolic shift from oxidative phosphorylation to glycolysis and increases ROS generation .\n\nWe show that upregulation of NOX is critical to support the elevated glycolysis by providing additional NAD+ .\n\nThe upregulation of NOX is also consistently observed in cancer cells with compromised mitochondria due to the activation of oncogenic Ras or loss of p53 , and in primary pancreatic cancer tissues .\n\nSuppression of NOX by chemical inhibition or genetic knockdown of gene expression selectively impacts cancer cells with mitochondrial dysfunction , leading to a decrease in cellular glycolysis , a loss of cell viability , and inhibition of cancer growth in vivo .\n\nOur study reveals a previously unrecognized function of NOX in cancer metabolism and suggests that NOX is a potential novel target for cancer treatment .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "Contact-inhibition ubiquitously exists in non-transformed cells and explains the poor regenerative capacity of in vivo human retinal pigment epithelial cells ( RPE ) during aging , injury and diseases .\n\nRPE injury or degeneration may unlock mitotic block mediated by contact inhibition but may also promote epithelial-mesenchymal transition ( EMT ) contributing to retinal blindness .\n\nHerein , we confirmed that EMT ensued in post-confluent ARPE-19 cells when contact inhibition was disrupted with EGTA followed by addition of EGF and FGF-2 because of activation of canonical Wnt and Smad/ZEB signaling .\n\nIn contrast , knockdown of p120-catenin ( p120 ) unlocked such mitotic block by activating p120/Kaiso , but not activating canonical Wnt and Smad/ZEB signaling , thus avoiding EMT .\n\nNuclear BrdU labeling was correlated with nuclear release of Kaiso through p120 nuclear translocation , which was associated with activation of RhoA-ROCK signaling , destabilization of microtubules .\n\nProlonged p120 siRNA knockdown followed by withdrawal further expanded RPE into more compact monolayers with a normal phenotype and a higher density .\n\nThis new strategy based on selective activation of p120/Kaiso but not Wnt/\\u03b2-catenin signaling obviates the need of using single cells and the risk of EMT , and may be deployed to engineer surgical grafts containing RPE and other tissues .", "output": "Evading growth suppressors, Activating invasion and metastasis" }, { "input": "High glycolysis , well known as \" Warburg effect, \" is frequently observed in a variety of cancers .\n\nWhether the deregulation of miRNAs contributes to the Warburg effect remains largely unknown .\n\nBecause miRNA regulates gene expression at both mRNA and protein levels , we constructed a gene functional association network , which allows us to detect the gene activity instead of gene expression , to integratively analyze the microarray data for gene expression and miRNA expression profiling and identify glycolysis-related gene-miRNA pairs deregulated in cancer .\n\nHexokinase 2 ( HK2 ) , coding for the first rate-limiting enzyme of glycolysis , is among the top list of genes predicted and potentially regulated by multiple miRNAs including miR-143 .\n\nInterestingly , miR-143 expression was inversely associated with HK2 protein level but not mRNA level in human lung cancer samples. miR-143 , down-regulated by mammalian target of rapamycin activation , reduces glucose metabolism and inhibits cancer cell proliferation and tumor formation through targeting HK2 .\n\nCollectively , we have not only established a novel methodology for gene-miRNA pair prediction but also identified miR-143 as an essential regulator of cancer glycolysis via targeting HK2 .", "output": "Cellular energetics" }, { "input": "The aryl hydrocarbon receptor ( AHR ) is a ligand-activated transcription factor that mediates the effects of the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin ( TCDD ) .\n\nRecently , AHR has emerged as a potential therapeutic target for breast cancer by virtue of its ability to modulate estrogen receptor-\u03b1 ( ER\u03b1 ) signalling and/or its ability to block cell proliferation .\n\nOur previous studies identified cyclin G2 ( CCNG2 ) , an inhibitor of cell-cycle progression , as an AHR target gene ; however , the mechanism of this regulation is unknown .\n\nChromatin immunoprecipitation assays in T-47D human breast cancer cells revealed a TCDD-dependent recruitment of AHR , nuclear co-activator 3 ( NCoA3 ) and the transcription factor forkhead box A1 ( FOXA1 ) , a key regulator of breast cancer cell signaling , to CCNG2 resulting in increases in CCNG2 mRNA and protein levels .\n\nMutation of the AHR response element ( AHRE ) and forkhead-binding sites abolished TCDD-induced CCNG2-regulated reporter gene activity .\n\nRNA interference-mediated knockdown of FOXA1 prevented the TCDD-dependent recruitment of AHR and NCoA3 to CCNG2 and reduced CCNG2 mRNA levels .\n\nInterestingly , knockdown of FOXA1 also caused a marked decrease in ER\u03b1 , but not AHR protein levels .\n\nHowever , RNA interference-mediated knockdown of ER\u03b1 , a negative regulator of CCNG2 , had no effect on TCDD-dependent AHR or NCoA3 recruitment to or expression of CCNG2 .\n\nThese findings show that FOXA1 , but not ER\u03b1 , is essential for AHR-dependent regulation of CCNG2 , assigning a role for FOXA1 in AHR action .", "output": "Sustaining proliferative signaling" }, { "input": "PURPOSE Precise monitoring of active angiogenesis in neovascular eye diseases such as age-related macular degeneration ( AMD ) enables sensitive use of antiangiogenic drugs and reduces adverse side effects .\n\nSo far , no in vivo imaging methods are available to specifically label active angiogenesis .\n\nHere , we report such a technique using fluorophore-labeled cationic liposomes ( CL ) detected with a standard clinical in vivo scanning laser ophthalmoscope ( SLO ) .\n\nMETHODS C57Bl/6 mice underwent laser coagulations at day 0 ( d0 ) to induce choroidal neovascularization ( CNV ) .\n\nLiposomes labeled with Oregon green , rhodamine ( Rh ) , or indocyanine green ( ICG ) were injected into the tail vein at various time points after laser coagulation , and their fluorescence was observed in vivo 60 min later using an SLO , or afterwards in choroidal flatmounts or cryosections .\n\nRESULTS SLO detected accumulated fluorescence only in active CNV lesions with insignificant background noise .\n\nThe best signal was obtained with CL-ICG .\n\nChoroidal flatmounts and cryosections of the eye confirmed the location of retained CL in CNV lesions .\n\nNeutral liposomes , in contrast , showed no accumulation .\n\nCONCLUSIONS These results establish fluorophore-labeled CL as high affinity markers to selectively stain active CNV .\n\nThis novel , non-invasive SLO imaging technique could improve risk assessment and indication for current intraocular antiangiogenic drugs in neovascular eye diseases , as well as monitor therapeutic outcomes .\n\nLabeling of angiogenic vessels using CL can be of interest not only for functional imaging in ophthalmology but also for other conditions where localization of active angiogenesis is desirable .", "output": "Inducing angiogenesis" }, { "input": "The Salvador/Warts/Hippo ( Hippo ) signaling pathway defines a novel signaling cascade regulating cell contact inhibition , organ size control , cell growth , proliferation , apoptosis and cancer development in mammals .\n\nThe upstream regulation of this pathway has been less well defined than the core kinase cassette .\n\nKIBRA has been shown to function as an upstream member of the Hippo pathway by influencing the phosphorylation of LATS and YAP , but functional consequences of these biochemical changes have not been previously addressed .\n\nWe show that in MCF10A cells , loss of KIBRA expression displays epithelial-to-mesenchymal transition ( EMT ) features , which are concomitant with decreased LATS and YAP phosphorylation , but not MST1/2 .\n\nIn addition , ectopic KIBRA expression antagonizes YAP via the serine 127 phosphorylation site and we show that KIBRA , Willin and Merlin differentially regulate genes controlled by YAP .\n\nFinally , reduced KIBRA expression in primary breast cancer specimens correlates with the recently described claudin-low subtype , an aggressive sub-group with EMT features and a poor prognosis .", "output": "Activating invasion and metastasis" }, { "input": "The effects of 17beta-estradiol ( E2 ) are mediated through activation of estrogen receptors ( ER ) : ERalpha and ERbeta .\n\nIt is known that ERalpha/ERbeta ratio is higher in breast tumors than in normal tissue .\n\nSince antioxidant enzymes and uncoupling proteins ( UCPs ) are reactive oxygen species ( ROS ) production and mitochondrial biogenesis regulators , our aim was to study the E2-effect on oxidative stress , antioxidant enzyme expression , and UCPs in breast cancer cell lines with different ERalpha/ERbeta ratios .\n\nThe lower ERalpha/ERbeta ratio T47D cell line showed low ROS production and high UCP5 levels .\n\nHowever , the higher ERalpha/ERbeta ratio MCF-7 cell line showed an up-regulation of antioxidant enzymes and UCPs , yet exhibited high oxidative stress .\n\nAs a result , a decrease in antioxidant enzyme activities and UCP2 protein levels , coupled with an increase in oxidative damage was found .\n\nOn the whole , these results show different E2-effects on oxidative stress regulation , modulating UCPs , and antioxidant enzymes , which were ERalpha/ERbeta ratio dependent in breast cancer cell lines .", "output": "Genomic instability and mutation, Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Breast cancer consists in a chronic inflammatory disease with multiple biological and clinical behaviors .\n\nBased on high throughput technologies data , this disease is currently classified according to the molecular expression of estrogen ( ER ) , progesterone ( PR ) and human epidermal growth factor ( HER-2 ) receptors .\n\nIn this study , we defined the inflammatory profile of the main molecular subtypes of breast cancer patients : luminal ( ER and PR positive , HER-2 negative ) , HER-2 enriched ( HER-2 positive ) and triple negative ( ER , PR and HER-2 negative ) .\n\nCytokines panel was assessed by measurement of TNF-\u03b1 , TGF-\u03b2 , IL-1 , IL-10 and IL-12 plasmatic levels .\n\nOxidative profile was assessed by determination of lipid peroxidation , total antioxidant capacity of plasma , malondialdehyde levels , carbonyl content and nitric oxide ( NO ) .\n\nClinical data were correlated with inflammatory findings .\n\nOur findings demonstrated that patients bearing the luminal subtype displayed high TNF-\u03b1 , TGF-\u03b2 and enhanced oxidative stress levels associated with reduced IL-12 .\n\nHER-2-enriched group exhibited higher levels of TNF-\u03b1 , IL-12 and TGF-\u03b2 associated with enhanced oxidative stress .\n\nTriple-negative subtype exhibited the most aggressive profile of disease behavior , with reduction in both TNF-\u03b1 and TGF-\u03b2 , with high levels of lipid peroxidation and NO .\n\nThe clinical importance of our findings lies in the fact that the inflammatory status varies in distinct ways due to molecular subtype of breast cancer , opening potential therapeutic targets to future therapies .", "output": "Tumor promoting inflammation" }, { "input": "Targeting receptor tyrosine kinase ( RTK ) degradation may be an interesting approach to reduce RTK cell signaling in cancer cells .\n\nHere we show that increasing E3 ubiquitin ligase casitas B-lineage lymphoma ( c-Cbl ) expression using lentiviral infection decreased osteosarcoma cell replication and survival and reduced cell migration and invasion in murine and human osteosarcoma cells .\n\nConversely , c-Cbl inhibition using short hairpin RNA ( shRNA ) increased osteosarcoma cell growth and survival , as well as invasion and migration , indicating that c-Cbl plays a critical role as a bone tumor suppressor .\n\nImportantly , the anticancer effect of increasing c-Cbl expression in osteosarcoma cells was related mainly to the downregulation of epidermal growth factor receptor ( EGFR ) and platelet-derived growth factor receptor alpha ( PDGFR\u03b1 ) .\n\nIn a murine bone tumor model , increasing c-Cbl expression also reduced RTK expression , resulting in decreased tumor cell proliferation and survival and reduced tumor growth .\n\nInterestingly , increasing c-Cbl also markedly reduced lung metastasis in mice .\n\nTissue microarray analysis revealed that low c-Cbl protein expression is associated with elevated EGFR and PDGFR\u03b1 protein levels in human osteosarcoma with poor outcome .\n\nThis study shows that increasing c-Cbl expression reduces osteosarcoma cell growth , survival , and metastasis in part through downregulation of RTKs , which supports the potential therapeutic interest of targeting c-Cbl in malignant bone diseases involving increased RTK .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Radiotherapy is commonly used for cancer treatment .\n\nHowever , it often results in side effects due to radiation damage in normal tissue , such as bone marrow ( BM ) failure .\n\nAdult hematopoietic stem and progenitor cells ( HSPC ) reside in BM next to the endosteal bone surface , which is lined primarily by hematopoietic niche osteoblastic cells .\n\nOsteoblasts are relatively more radiation-resistant than HSPCs , but the mechanisms are not well understood .\n\nIn the present study , we demonstrated that the stress response gene REDD1 ( regulated in development and DNA damage responses 1 ) was highly expressed in human osteoblast cell line ( hFOB ) cells after \\u03b3 irradiation .\n\nKnockdown of REDD1 with siRNA resulted in a decrease in hFOB cell numbers , whereas transfection of PCMV6-AC-GFP-REDD1 plasmid DNA into hFOB cells inhibited mammalian target of rapamycin ( mTOR ) and p21 expression and protected these cells from radiation-induced premature senescence ( PS ) .\n\nThe PS in irradiated hFOB cells were characterized by significant inhibition of clonogenicity , activation of senescence biomarker SA-\\u03b2-gal , and the senescence-associated cytokine secretory phenotype ( SASP ) after 4 or 8 Gy irradiation .\n\nImmunoprecipitation assays demonstrated that the stress response proteins p53 and nuclear factor \\u03ba B ( NFkB ) interacted with REDD1 in hFOB cells .\n\nKnockdown of NFkB or p53 gene dramatically suppressed REDD1 protein expression in these cells , indicating that REDD1 was regulated by both factors .\n\nOur data demonstrated that REDD1 is a protective factor in radiation-induced osteoblast cell premature senescence .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "OBJECTIVE Angiogenesis represents a key element in the pathogenesis of malignancy .\n\nThere are no robust data on prognostic factors for overall survival ( OS ) in patients with metastatic colorectal cancer treated with vascular endothelial growth factor ( VEGF)-targeted therapy .\n\nThe present study was conducted to establish a prognostic model for patients using an oxaliplatin-based or irinotecan-based chemotherapy plus bevacizumab in metastatic colorectal cancer .\n\nMETHODS Baseline characteristics and outcomes on 170 patients treated with FOLFIRI or XELOX plus anti-VEGF therapy-naive metastatic colorectal cancer were collected from three Turkey cancer centers .\n\nCox proportional hazards regression was used to identify independent prognostic factors for OS .\n\nRESULTS The median OS for the whole cohort was 19 months ( 95% CI , 14.3 to 23.6 months ) .\n\nThree of the seven adverse prognostic factors according to the Anatolian Society of Medical Oncology ( ASMO ) were independent predictors of short survival : serum lactate dehydrogenase ( LDH ) greater than the upper limit of normal ( ULN ; p<0.001 ) ; neutrophils greater than the ULN ( p<0.0014 ) ; and progression free survival ( PFS ) less than 6 months ( p =0.001 ) .\n\nCONCLUSION Serum LDH and neutrophil levels were the main prognostic factors in predicting survival , followed by PFS .\n\nThis model validates incorporation of components of the ASMO model into patient care and clinical trials that use VEGF-targeting agents .", "output": "Inducing angiogenesis" }, { "input": "Cancer cells have elevated aerobic glycolysis that is termed the Warburg effect .\n\nBut several tumor cells , including leukemic cells , also increase glutamine metabolism , which is initiated by glutaminase ( GLS ) .\n\nThe microRNA ( miRNA ) miR-23 targets GLS mRNA and inhibits expression of GLS protein .\n\nHere we show that in human leukemic Jurkat cells the NF-\u03baB p65 subunit binds to miR-23a promoter and inhibits miR-23a expression .\n\nHistone deacetylase ( HDAC ) inhibitors release p65-induced inhibition .\n\nJurkat cells growing in glutamine decrease proliferation due to cell accumulation in G0/G1 phase .\n\nNevertheless , cells get used to this new source of energy by increasing GLS expression , which correlates with an increase in p65 expression and its translocation to the nucleus , leading to a higher basal NF-\u03baB activity .\n\nJurkat cells overexpressing p65 show increase basal GLS expression and proliferate faster than control cells in glutamine medium .\n\nOverexpressing miR-23a in leukemic cells impaired glutamine use and induces mitochondrial dysfunction leading to cell death .\n\nTherefore , p65 activation decreases miR-23a expression , which facilitates glutamine consumption allowing leukemic cells to use this alternative source of carbon and favoring their adaptation to the metabolic environment .", "output": "Sustaining proliferative signaling, Cellular energetics" }, { "input": "BACKGROUND The c-Met receptor tyrosine kinase is aberrantly activated in many solid tumors .\n\nIn a prior study we showed that prostate cancer PC-3 cells exhibit constitutively activated c-Met without exogenous hepatocyte growth factor ( HGF ) ; however whether this characteristic is due to an endogenous HGF/c-Met autocrine loop remains controversial .\n\nIn the current study we examined the response of PC-3 cells to an anti-HGF neutralizing antibody or a small molecule Met kinase inhibitor ( BMS-777607 ) .\n\nMETHODS Cell scattering was tested by monitoring cell morphology after HGF stimulation .\n\nCell migration was examined by both \" wound-healing \" and transwell assasy and invasion was detected by Matrigel-coated transwell assay .\n\nProliferation , survival and anoikis were determined by MTT , colony formation and trypan blue exclusion assay , respectively .\n\nGene and protein expression were assessed by real-time PCR and Western blot , respectively .\n\nRESULTS Although HGF mRNA could be detected in PC-3 cells , the molecular weight of secreted \" HGF \" protein was inconsistent with the functional recombinant HGF .\n\nFurthermore , conditioned medium from PC-3 cell cultures was ineffective at triggering either motogenic behavior or c-Met signaling in DU145 , another prostate cancer cell line expressing c-Met but lacking basal c-Met activation .\n\nPC-3 cells also were not responsive to the anti-HGF neutralizing antibody in experiments assessing proliferation , migration , or c-Met signaling .\n\nBMS-777607 treatment with micromolar doses nonetheless led to significant inhibition of multiple PC-3 cell functions including proliferation , clonogenicity , migration and invasion .\n\nAt the molecular level , BMS-777607 suppressed autophosphorylated c-Met and downstream c-Src and Akt pathways .\n\nCONCLUSIONS These results suggest that the constitutive c-Met activation in PC-3 is independent of autocrine stimulation .\n\nBecause PC-3 cells were responsive to BMS-777607 but not the anti-HGF antibody , the findings also indicate that under circumstances where c-Met is constitutively hyperactive in the absence of functional HGF , targeting the c-Met receptor remains a viable therapeutic option to impede cancer progression .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Most ovarian cancers are estrogen-positive and hormonal treatments using anti-estrogens or aromatase inhibitors are under investigation for treating the tumors that are resistant to conventional therapies .\n\nIn this study , the long-term effects of two anti-estrogens , namely 4-hydroxytamoxifen and fulvestrant ( or ICI182,780 ) , were investigated in ER\u03b1-positive BG1 epithelial ovarian cancer cells .\n\nTo this aim , cells were grown in the presence of anti-estrogen concentrations that were sufficient to saturate the estrogen receptors , but were neither cytotoxic nor cytostatic as indicated by the absence of inhibition of cell proliferation .\n\nIn these conditions and despite the lack of cytostatic effect of the drugs , long-term treatment ( 3 months ) with the pure anti-estrogen fulvestrant induced a specific , reproducible and irreversible inhibition of ER\u03b1 expression .\n\nThis inhibition was accompanied by loss of estrogen-induced cell proliferation and gene expression as indicated by the analysis of several estrogen-responsive genes .\n\nER\u03b1 down-regulation was not linked to deregulated expression of transcription factors which drive ER\u03b1 transcription and did not involve DNA methylation or histone deacetylation .\n\nAltogether , these results demonstrate that non-cytotoxic concentrations of pure anti-estrogens affect estrogen signaling and might be relevant for the treatment for ovarian cancers .", "output": "Sustaining proliferative signaling" }, { "input": "Semiconductor nanoparticles ( NPs ) became important and wide-used tool for cell imaging because of their unique remarkable properties .\n\nNevertheless , all previous investigations in this area were done on proliferating cells .\n\nFor the first time , this work demonstrates strong influence of cell active proliferation/contact inhibition of proliferation on uptake of NPs .\n\nIn addition , we show that cell division plays key-role in penetration of silicon carbide based NPs ( SiC NPs ) inside the cell nucleus .\n\nThis may very likely concern other types of NPs able to reach the cell nuclei .\n\nIn particular , observed effect of cell division gives perspectives for future selective cancer treatment with NPs .", "output": "Evading growth suppressors" }, { "input": "We previously reported that hepatic stellate cells ( HSCs ) activated by angiotensin II ( AngII ) facilitate stromal fibrosis and tumor progression in intrahepatic cholangiocarcinoma ( ICC ) .\n\nAngII has been known as a growth factor which can promote epithelial-to-mesenchymal transition ( EMT ) in renal epithelial cells , alveolar epithelial cells and peritoneal mesothelial cells .\n\nHowever , in the past , the relationship between AngII and stromal cell-derived factor-1 ( SDF-1 ) in the microenvironment around cancer and the role of AngII on EMT of cancer cells has not been reported in detail .\n\nSDF-1 and its specific receptor , CXCR4 , are now receiving attention as a mechanism of cell progression and metastasis .\n\nIn this study , we examined whether activated HSCs promote tumor fibrogenesis , tumor progression and distant metastasis by mediating EMT via the AngII/AngII type 1 receptor ( AT-1 ) and the SDF-1/CXCR4 axis .\n\nTwo human ICC cell lines and a human HSC line , LI-90 , express CXCR4 .\n\nSignificantly higher concentration of SDF-1\u03b1 was released into the supernatant of LI-90 cells to which AngII had been added .\n\nSDF-1\u03b1 increased the proliferative activity of HSCs and enhanced the activation of HSCs as a growth factor .\n\nFurthermore , addition of SDF-1\u03b1 and AngII enhanced the increase of the migratory capability and vimentin expression , reduced E-cadherin expression , and translocated the expression of \u03b2-catenin into the nucleus and cytoplasm in ICC cells .\n\nCo-culture with HSCs also enhanced the migratory capability of ICC cells .\n\nThese findings suggest that SDF-1\u03b1 , released from activated HSCs and AngII , play important roles in cancer progression , tumor fibrogenesis , and migration in autocrine and paracrine fashion by mediating EMT .\n\nOur mechanistic findings may provide pivotal insights into the molecular mechanism of the AngII and SDF-1\u03b1-initiated signaling pathway that regulates fibrogenesis in cancerous stroma , tumor progression and meta-stasis of tumor cells expressing AT-1 and CXCR4 .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Cancer cells typically display altered glucose metabolism characterized by a preference of aerobic glycolysis , known as the Warburg effect , which facilitates cell proliferation .\n\nHypoxia-inducible factor ( HIF ) and oncoprotein Myc are two prominent transcription factors that drive glycolysis .\n\nPreviously , we reported that the estrogen-related receptors ( ERRs ) act as cofactors of HIF and enhance HIF-dependent transcription of glycolytic genes under hypoxia .\n\nERRs are orphan nuclear receptors and key regulators of energy metabolism by orchestrating mitochondrial biogenesis , fatty acid oxidation ( FAO ) and oxidative phosphorylation .\n\nHere , we show that ERRs also stimulate glycolysis under normoxia .\n\nERRs directly bind to and activate promoters of many genes encoding glycolytic enzymes , and the ERR-binding sites in such promoters are essential for ERR-mediated transcriptional activation .\n\nERRs interact with Myc , and the two factors synergistically activate transcription of glycolytic genes .\n\nFurthermore , overexpression of ERRs increases glycolytic gene expression and lactate production .\n\nConversely , depletion of ERRs in cancer cells reduces expression of glycolytic genes and glucose uptake , resulting in decreased aerobic glycolysis and cell growth .\n\nTaken together , these results suggest that ERRs are important transcriptional activators of the glycolytic pathway and contribute to the Warburg effect in cancer cells .", "output": "Sustaining proliferative signaling, Cellular energetics" }, { "input": "The results of experimental studies have indicated the pleiotropic effects of statins in organism , e.g. the influence on cell cycle , apoptosis or angiogenesis .\n\nIn this study , the effects of simvastatin on selected parameters of apoptosis and proliferation in chemocarcinogen-induced mammary tumorigenesis in female rats were determined .\n\nSimvastatin was administered dietary at a dose of 18 mg/kg and highly effective dose of 180 mg/kg the entire experiment ( 18 weeks ) .\n\nAt autopsy mammary tumors were removed and prepared for immunohistochemical and histomorphological analysis .\n\nIn treated animals ( simvastatin 180 mg/kg ) , significant decrease by 12% in Bcl-2 protein expression and non-significant decrease by 27% of Ki67 protein expression in tumor cells compared to tumor cells in control animals were observed after semiquantitative evaluation .\n\nMorphometrical analysis has shown significant proapototic shift in Bcl-2/Bax ratio in tumor cells .\n\nIn high grade control carcinoma cells , the expression of Ki67 increased by 37% ( non-significantly ) in comparison with control low grade carcinomas .\n\nA histomorphological analysis of malignant tumors has revealed a shift from high grade to low grade carcinomas after simvastatin treatment .\n\nThe noticeable decrease of mammary tumor frequency and incidence in rats after simvastatin treatment was accompanied with antiapoptotic Blc-2 protein decrease and proapoptotic Bax protein increase in this experiment .", "output": "Resisting cell death" }, { "input": "Head and neck squamous cell carcinoma ( HNSCC ) is a major cause of morbidity and mortality underscoring the need for safe and effective chemopreventive strategies .\n\nTargeting epidermal growth factor receptor ( EGFR ) is attractive in that it is an early critical event in HNSCC pathogenesis .\n\nHowever , current agents lack efficacy or have unacceptable toxicity .\n\nSeveral groups have demonstrated that the over-the-counter medication , polyethylene glycol ( PEG ) has remarkable chemopreventive efficacy against colon carcinogenesis .\n\nImportantly , we reported that this effect is mediated through EGFR internalization/degradation .\n\nIn the current study , we investigated the chemopreventive efficacy of this agent against HNSCC , using both the well validated animal model 4-NQO ( 4-nitroquinoline 1-oxide ) rat model and cell culture with the human HNSCC cell line SCC-25 .\n\nWe demonstrated that daily topical application of 10% PEG-8000 in the oral cavity ( tongue and cavity wall ) post 4NQO initiation resulted in a significant reduction in tumor burden ( both , tumor size and tumors/tumor bearing rat ) without any evidence of toxicity .\n\nImmunohistochemical studies depicted decreased proliferation ( number of Ki67-positive cells ) and reduced expression of EGFR and its downstream effectors cyclin D1 in the tongue mucosa of 4NQO-rats treated with PEG .\n\nWe showed that EGFR was also markedly downregulated in SCC-25 cells by PEG-8000 with a concomitant induction of G1-S phase cell-cycle arrest , which was potentially mediated through upregulated p21(cip1/waf1) .\n\nIn conclusion , we demonstrate , for the first time , that PEG has promising efficacy and safety as a chemopreventive efficacy against oral carcinogenesis .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "The therapeutic principle of allogeneic haematopoietic cell transplantation ( allo-HCT ) is based on an active donor immune system that eliminates host-derived tumour cells .\n\nWe hypothesized that in addition to the alloantigen-driven anti-tumour response , disruption of the immunological microenvironment within the tumour is responsible for its elimination after allo-HCT .\n\nWe observed that induction of graft-versus-host disease ( GvHD ) significantly reduced the abundance of luc(+) FoxP3(+) regulatory T ( Treg ) cells in the tumour tissue , which is indicative of impaired or over-ridden tumour recruitment signals towards Treg cells .\n\nAnalysis of the intestines and liver revealed chemokines and purine nucleotides as candidates for attracting Treg to these sites of inflammation .\n\nDespite its expression on tissue-residing Treg cells , the chemokine receptor CCR3 was not critical for Treg-cell function following allo-HCT .\n\nExtracellular ATP can attract immune cells via P2Y2 .\n\nP2Y2 was found to be expressed on Treg cells , and we found a partial reduction of GvHD prevention when P2Y2(-/-) rather than P2Y2(+/+) Treg cells were given .\n\nExogenous local inflammation reduced Treg-cell accumulation in the tumour , suggesting a potential clinical approach to prevent Treg-cell-mediated tumour escape .\n\nIn conclusion , we demonstrate that GvHD-related inflammation reduced Treg-cell numbers at the tumour sites , which may in turn help to explain the observation that patients with GvHD have a lower risk of tumour relapse .", "output": "Tumor promoting inflammation" }, { "input": "Adults and children with high-risk CRLF2-rearranged acute lymphoblastic leukemia ( ALL ) respond poorly to current cytotoxic chemotherapy and suffer unacceptably high rates of relapse , supporting the need to use alternative therapies .\n\nCRLF2 encodes the thymic stromal lymphopoietin ( TSLP ) receptor , which activates cell signaling in normal lymphocytes on binding its ligand , TSLP .\n\nWe hypothesized that aberrant cell signaling occurs in CRLF2-rearranged ALL and can be targeted by signal transduction inhibitors of this pathway .\n\nIn a large number of primary CRLF2-rearranged ALL samples , we observed increased basal levels of pJAK2 , pSTAT5 , and pS6 .\n\nWe thus characterized the biochemical sequelae of CRLF2 and JAK alterations in CRLF2-rearranged ALL primary patient samples via analysis of TSLP-mediated signal transduction .\n\nTSLP stimulation of these leukemias further induced robust JAK/STAT and PI3K/mTOR pathway signaling .\n\nJAK inhibition abrogated phosphorylation of JAK/STAT and , surprisingly , of PI3K/mTOR pathway members , suggesting an interconnection between these signaling networks and providing a rationale for testing JAK inhibitors in clinical trials .\n\nThe PI3K/mTOR pathway inhibitors rapamycin , PI103 , and PP242 also inhibited activated signal transduction and translational machinery proteins of the PI3K/mTOR pathway , suggesting that signal transduction inhibitors targeting this pathway also may have therapeutic relevance for patients with CRLF2-rearranged ALL and merit further preclinical testing .", "output": "Sustaining proliferative signaling" }, { "input": "To determine the threshold dose of \u03b2-Naphthoflavone ( BNF ) that induces hepatocellular tumor promoting effects , reactive oxygen species ( ROS ) generation and thiobarbituric acid-reactive substance ( TBARS ) formation , and drug-metabolizing enzymes that protect against ROS generation , two-stage liver carcinogenesis model was used .\n\nPartial hepatectomized rats ( n = 11 to 12 ) were fed diets containing 0 , 0.03 , 0.06 , 0.125 or 0.25% BNF for 6 weeks after an intraperitoneal injection of N-diethylnitrosamine ( DEN ) to initiate hepatocarcinogenesis .\n\nHistopathologically , glutathione S-transferase placental form ( GST-P)-positive foci significantly increased in rats given 0.25% BNF .\n\nNo marked changes in ROS production and TBARS contents were observed between the BNF treated and DEN alone groups .\n\nReal-time RT-PCR showed that the expression of Cyp1a1 , Cyp1a2 , Cyp1b1 and Nqo1 significantly increased in the groups given 0.03% BNF or more , but Ugt1a6 , Akr7a3 and Gstm1 significantly increased in the groups given 0.125% BNF or more .\n\nGpx2 and Yc2 significantly increased in the groups given 0.06% BNF or more and 0.25% BNF , respectively .\n\nInflammation-related genes such as Ccl2 , Mmp12 , Serpine1 and Cox-2 significantly increased in the 0.25% BNF group .\n\nIn immunohistochemistry , the number of cyclooxygenase-2 ( COX-2)-positive cells increased in rats given 0.25% BNF .\n\nThese results suggest that 0.25% BNF is the threshold dose for liver tumor promotion , and the fact that inflammation-related genes and COX-2 protein increased in the 0.25% BNF group strongly suggests that inflammation is involved in the liver tumor promoting effect of BNF in rats .", "output": "Tumor promoting inflammation" }, { "input": "The functional and therapeutic importance of the Warburg effect is increasingly recognized , and glycolysis has become a target of anticancer strategies .\n\nWe recently reported the identification of a group of novel small compounds that inhibit basal glucose transport and reduce cancer cell growth by a glucose deprivation-like mechanism .\n\nWe hypothesized that the compounds target Glut1 and are efficacious in vivo as anticancer agents .\n\nHere , we report that a novel representative compound WZB117 not only inhibited cell growth in cancer cell lines but also inhibited cancer growth in a nude mouse model .\n\nDaily intraperitoneal injection of WZB117 at 10 mg/kg resulted in a more than 70% reduction in the size of human lung cancer of A549 cell origin .\n\nMechanism studies showed that WZB117 inhibited glucose transport in human red blood cells ( RBC ) , which express Glut1 as their sole glucose transporter .\n\nCancer cell treatment with WZB117 led to decreases in levels of Glut1 protein , intracellular ATP , and glycolytic enzymes .\n\nAll these changes were followed by increase in ATP-sensing enzyme AMP-activated protein kinase ( AMPK ) and declines in cyclin E2 as well as phosphorylated retinoblastoma , resulting in cell-cycle arrest , senescence , and necrosis .\n\nAddition of extracellular ATP rescued compound-treated cancer cells , suggesting that the reduction of intracellular ATP plays an important role in the anticancer mechanism of the molecule .\n\nSenescence induction and the essential role of ATP were reported for the first time in Glut1 inhibitor-treated cancer cells .\n\nThus , WZB117 is a prototype for further development of anticancer therapeutics targeting Glut1-mediated glucose transport and glucose metabolism .", "output": "Enabling replicative immortality, Sustaining proliferative signaling, Cellular energetics, Resisting cell death" }, { "input": "Helicobacter hepaticus-infected Rag2(-/-) mice emulate many aspects of human inflammatory bowel disease , including the development of colitis and colon cancer .\n\nTo elucidate mechanisms of inflammation-induced carcinogenesis , we undertook a comprehensive analysis of histopathology , molecular damage , and gene expression changes during disease progression in these mice .\n\nInfected mice developed severe colitis and hepatitis by 10wk post-infection , progressing into colon carcinoma by 20wk post-infection , with pronounced pathology in the cecum and proximal colon marked by infiltration of neutrophils and macrophages .\n\nTranscriptional profiling revealed decreased expression of DNA repair and oxidative stress response genes in colon , but not in liver .\n\nMass spectrometric analysis revealed higher levels of DNA and RNA damage products in liver compared to colon and infection-induced increases in 5-chlorocytosine in DNA and RNA and hypoxanthine in DNA .\n\nParadoxically , infection was associated with decreased levels of DNA etheno adducts .\n\nLevels of nucleic acid damage from the same chemical class were strongly correlated in both liver and colon .\n\nThe results support a model of inflammation-mediated carcinogenesis involving infiltration of phagocytes and generation of reactive species that cause local molecular damage leading to cell dysfunction , mutation , and cell death .\n\nThere are strong correlations among histopathology , phagocyte infiltration , and damage chemistry that suggest a major role for neutrophils in inflammation-associated cancer progression .\n\nFurther , paradoxical changes in nucleic acid damage were observed in tissue- and chemistry-specific patterns .\n\nThe results also reveal features of cell stress response that point to microbial pathophysiology and mechanisms of cell senescence as important mechanistic links to cancer .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Resisting cell death" }, { "input": "The secretin receptor ( SR ) , a G protein-coupled receptor , mediates the effects of the gastrointestinal hormone secretin on digestion and water homeostasis .\n\nRecently , high SR expression has been observed in pancreatic ductal adenocarcinomas , cholangiocellular carcinomas , gastrinomas , and bronchopulmonary carcinoid tumors .\n\nReceptor overexpression associates with enhanced secretin-mediated signaling , but whether this molecule plays an independent role in tumorigenesis is currently unknown .\n\nWe recently discovered that pheochromocytomas developing in rats affected by the MENX ( multiple endocrine neoplasia-like ) syndrome express at very high-level Sctr , encoding SR .\n\nWe here report that SR are also highly abundant on the membranes of rat adrenal and extraadrenal pheochromocytoma , starting from early stages of tumor development , and are functional .\n\nPC12 cells , the best characterized in vitro pheochromocytoma model , also express Sctr at high level .\n\nThus , we used them as model to study the role of SR in neoplastic transformation .\n\nSmall interfering RNA-mediated knockdown of Sctr decreases PC12 cells proliferation and increases p27 levels .\n\nThe proproliferative effect of SR in PC12 cells is mediated , in part , by the phosphatidylinositol 3 kinase ( PI3K)/serine-threonine protein kinase ( AKT ) pathway .\n\nTransfection of Sctr in Y1 adrenocortical carcinoma cells , expressing low endogenous levels of Sctr , stimulates cell proliferation also , in part , via the PI3K/AKT signaling cascade .\n\nBecause of the link between SR and PI3K/AKT signaling , tumor cells expressing high levels of the receptor ( MENX-associated primary pheochromocytoma and NCI-H727 human bronchopulmonary carcinoid cells ) respond well and in a SR-dependent manner to PI3K inhibitors , such as NVP-BEZ235 .\n\nThe association between SR levels and response to PI3K inhibition might open new avenues for the treatment of tumors overexpressing this receptor .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "CDKN1C/P57 is a cyclin-dependent kinase inhibitor implicated in different human cancers , including hepatocellular carcinoma ( HCC ) ; however , little is known regarding the role of CDKN1C/P57 and its regulation in HCC .\n\nIn this study , we show that the down-regulation of Notch1 and Notch3 in two HCC cell lines resulted in Hes1 down-regulation , CDKN1C/P57 up-regulation , and reduced cell growth .\n\nIn line with these data , we report that CDKN1C/P57 is a target of transcriptional repression by the Notch effector , Hes1 .\n\nWe found that the up-regulation of CDKN1C/P57 by cDNA transfection decreased tumor growth , as determined by growth curve , flow cytometry analysis , and cyclin D1 down-regulation , without affecting the apoptosis machinery .\n\nIndeed , the expression of Bax , Noxa , PUMA , BNIP(3) , and cleaved caspase-3 was not affected by CDKN1C/P57 induction .\n\nMorphologically CDKN1C/p57-induced HCC cells became flat and lengthened in shape , accumulated the senescence-associated \u03b2-galactosidase marker , and increased P16 protein expression .\n\nEvaluation of senescence in cells depleted both for Hes1 and CDKN1C/P57 revealed that the senescent state really depends on the accumulation of CDKN1C/p57 .\n\nFinally , we validated our in vitro results in primary HCCs , showing that Hes1 protein expression inversely correlates with CDKN1C/P57 mRNA levels .\n\nIn addition , reduced Hes1 protein expression is accompanied by a shorter time to recurrence after curative resection , suggesting that Hes1 may represent a biomarker for prediction of patients with poor prognosis .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "OBJECTIVE Hepatocellular carcinoma ( HCC ) is a highly vascularized tumor in which neoangiogenesis contributes to growth and metastasis .\n\nWe assessed the safety , efficacy , and potential biomarkers of activity of bevacizumab in patients with advanced HCC .\n\nMETHODS In this phase II trial , eligible patients received bevacizumab , 5 mg/kg or 10 mg/kg every 2 weeks .\n\nThe disease-control rate at 16 weeks ( 16W-DCR ) was the primary endpoint .\n\nCirculating endothelial cells ( CECs ) and plasma cytokines and angiogenic factors ( CAFs ) were measured at baseline and throughout treatment .\n\nRESULTS The 16W-DCR was 42% ( 95% confidence interval , 27%-57% ) .\n\nSix of the 43 patients who received bevacizumab achieved a partial response ( objective response rate [ ORR ] , 14% ) .\n\nGrade 3-4 asthenia , hemorrhage , and aminotransferase elevation occurred in five ( 12% ) , three ( 7% ) , and three ( 7% ) patients , respectively .\n\nDuring treatment , placental growth factor markedly increased , whereas vascular endothelial growth factor ( VEGF)-A dramatically decreased ( p < .0001 ) ; soluble VEGF receptor-2 ( p < .0001 ) and CECs ( p = .03 ) transiently increased on day 3 .\n\nHigh and increased CEC counts at day 15 were associated with the ORR ( p = .04 ) and the 16W-DCR ( p = .02 ) , respectively .\n\nLower interleukin ( IL)-8 levels at baseline ( p = .01 ) and throughout treatment ( p \u2264 .04 ) were associated with the 16W-DCR .\n\nHigh baseline IL-8 and IL-6 levels predicted shorter progression-free and overall survival times ( p \u2264 .04 ) .\n\nCONCLUSION Bevacizumab is active and well tolerated in patients with advanced HCC .\n\nThe clinical value of CECs , IL-6 , and IL-8 warrants further investigation .", "output": "Inducing angiogenesis" }, { "input": "Silver nanoparticles ( Ag-np ) have been used in medicine and commercially due to their anti-microbial properties .\n\nTherapeutic potentials of these nanoparticles are being explored extensively despite the lack of information on their mechanism of action at molecular and cellular level .\n\nHere , we have investigated the DNA damage response and repair following Ag-np treatment in mammalian cells .\n\nStudies have shown that Ag-np exerts genotoxicity through double-strand breaks ( DSBs ) .\n\nDNA-PKcs , the catalytic subunit of DNA dependent protein kinase , is an important caretaker of the genome which is known to be the main player mediating Non-homologous End-Joining ( NHEJ ) repair pathway .\n\nWe hypothesize that DNA-PKcs is responsible for the repair of Ag-np induced DNA damage .\n\nIn vitro studies have been carried out to investigate both cytotoxicity and genotoxicity induced by Ag-np in normal human cells , DNA-PKcs proficient , and deficient mammalian cells .\n\nChemical inhibition of DNA-PKcs activity with NU7026 , an ATP-competitive inhibitor of DNA-PKcs , has been performed to further validate the role of DNA-PKcs in this model .\n\nOur results suggest that Ag-np induced more prominent dose-dependent decrease in cell viability in DNA-PKcs deficient or inhibited cells .\n\nThe deficiency or inhibition of DNA-PKcs renders the cells with higher susceptibility to DNA damage and genome instability which in turn contributed to greater cell cycle arrest/cell death .\n\nThese findings support the fact that DNA-PKcs is involved in the repair of Ag-np induced genotoxicity and NHEJ repair pathway and DNA-PKcs particularly is activated to safeguard the genome upon Ag-np exposure .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Proliferating cells consume more glucose to cope with the bioenergetics and biosynthetic demands of rapidly dividing cells as well as to counter a shift in cellular redox environment .\n\nThis study investigates the hypothesis that manganese superoxide dismutase ( MnSOD ) regulates cellular redox flux and glucose consumption during the cell cycle .\n\nA direct correlation was observed between glucose consumption and percentage of S-phase cells in MnSOD wild-type fibroblasts , which was absent in MnSOD homozygous knockout fibroblasts .\n\nResults from electron paramagnetic resonance spectroscopy and flow cytometric assays showed a significant increase in cellular superoxide levels in S-phase cells , which was associated with an increase in glucose and oxygen consumption , and a decrease in MnSOD activity .\n\nMass spectrometry results showed a complex pattern of MnSOD-methylation at both lysine ( 68 , 89 , 122 , and 202 ) and arginine ( 197 and 216 ) residues .\n\nMnSOD protein carrying a K89A mutation had significantly lower activity compared with wild-type MnSOD .\n\nComputational-based simulations indicate that lysine and arginine methylation of MnSOD during quiescence would allow greater accessibility to the enzyme active site as well as increase the positive electrostatic potential around and within the active site .\n\nMethylation-dependent changes in the MnSOD conformation and subsequent changes in the electrostatic potential around the active site during quiescence versus proliferation could increase the accessibility of superoxide , a negatively charged substrate .\n\nThese results support the hypothesis that MnSOD regulates a \" metabolic switch \" during progression from quiescent through the proliferative cycle .\n\nWe propose MnSOD as a new molecular player contributing to the Warburg effect .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Cellular energetics" }, { "input": "Past studies have shown that amplified insulin-like growth factor 1 ( IGF1)/IGF1 receptor ( IGF1-R ) signalling has an important role in colorectal cancer ( CRC ) development , progression and resistance to treatment .\n\nIn this report , we demonstrate that downregulation of microRNA-497 ( miR-497 ) as a result of DNA copy number reduction is involved in upregulation of IGF1-R in CRC cells .\n\nMiR-497 and miR-195 of the miR-15/16/195/424/497 family that share the same 3 ' untranslated region ( 3'UTR ) binding seed sequence and are predicted to target IGF1-R were concurrently downregulated in the majority of CRC tissues relative to paired adjacent normal mucosa .\n\nHowever , only overexpression of miR-497 led to suppression of the IGF1-R 3'UTR activity and downregulation of the endogenous IGF1-R protein in CRC cells .\n\nThis was associated with inhibition of cell survival , proliferation and invasion , and increased sensitivity to apoptosis induced by various stimuli including the chemotherapeutic drugs cisplatin and 5-fluorouracil , and the death ligand tumour necrosis factor-related apoptosis-inducing ligand .\n\nThe biological effect of miR-497 on CRC cells was largely mediated by inhibition of phosphatidylinositol 3-kinase/Akt signalling , as overexpression of an active form of Akt reversed its impact on cell survival and proliferation , recapitulating the effect of overexpression of IGF1-R .\n\nDownregulation of miR-497 and miR-195 appeared to associate with copy number loss of a segment of chromosome 17p13.1 , where these miRs are located at proximity .\n\nSimilarly to miR-195 , the members of the same miR family , miR-424 that was upregulated , and miR-15a , miR-15b and miR-16 that were unaltered in expression in CRC tissues compared with paired adjacent normal mucosa , did not appear to have a role in regulating the expression of IGF1-R .\n\nTaken together , these results identify downregulation of miR-497 as an important mechanism of upregulation of IGF1-R in CRC cells that contributes to malignancy of CRC.Oncogene advance online publication , 18 June 2012 ; doi:10.1038/onc.2012.214 .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "To elucidate the function of MAS-related GPCR , member D ( MRGD ) in cancers , we investigated the in vitro and in vivo oncogenic function of MRGD using murine fibroblast cell line NIH3T3 in which MRGD is stably expressed .\n\nThe expression pattern of MRGD in clinical samples was also analyzed .\n\nWe found that overexpression of MRGD in NIH3T3 induced focus formation and multi-cellular spheroid formation , and promoted tumors in nude mice .\n\nIn other words , overexpression of MRGD in NIH3T3 induced the loss of contact inhibition , anchorage-independent growth and in vivo tumorigenesis .\n\nFurthermore , it was found that the ligand of MRGD , beta-alanine , enhanced spheroid formation in MRGD-expressing NIH3T3 cells .\n\nFrom investigation of clinical cancer tissues , we found high expression of MRGD in several lung cancers by immunohistochemistry as well as real time PCR .\n\nBased on these results , MRGD could be involved in tumorigenesis and could also be a novel anticancer drug target .", "output": "Evading growth suppressors" }, { "input": "Contact inhibition of locomotion ( CIL ) is the process by which cells stop the continual migration in the same direction after collision with another cell .\n\nHighly invasive malignant cells exhibit diminished CIL when they contact stromal cells , which allows invasion of the tissue by tumors .\n\nWe show that Nm23-H1 is essential for the suppression of Rac1 through inactivation of Tiam1 at the sites of cell-cell contact , which plays a pivotal role in CIL .\n\nU87MG cells show CIL when they contact normal glia .\n\nIn spheroid confrontation assays U87MG cells showed only limited invasion of the glial population , but reduction of Nm23-H1 expression in U87MG cells abrogated CIL resulting in invasion .\n\nIn U87MG cells , Nm23-H1 is translocated to the sites of contact with glia through association with \\u03b1-catenin and N-cadherin .\n\nMutants of Nm23-H1 , which lacked the binding ability with Tiam1 , or \\u03b1-catenin did not restore CIL .\n\nMoreover , the expression of ephrin-B1 in tumor cells disrupted CIL and promoted invasion .\n\nAs one mechanism , ephrin-B1 inhibits the association of Nm23-H1 with Tiam1 , which contributes for activation of Rac1 .\n\nThese results indicate a novel function of Nm23-H1 to control CIL , and its negative regulation by ephrin-B1 .", "output": "Evading growth suppressors, Activating invasion and metastasis" }, { "input": "Recent evidence indicates that the estrogen receptor-\u03b1-negative , androgen receptor ( AR)-positive molecular apocrine subtype of breast cancer is driven by AR signaling .\n\nThe MDA-MB-453 cell line is the prototypical model of this breast cancer subtype ; its proliferation is stimulated by androgens such as 5\u03b1-dihydrotestosterone ( DHT ) but inhibited by the progestin medroxyprogesterone acetate ( MPA ) via AR-mediated mechanisms .\n\nWe report here that the AR gene in MDA-MB-453 cells contains a G-T transversion in exon 7 , resulting in a receptor variant with a glutamine to histidine substitution at amino acid 865 ( Q865H ) in the ligand binding domain .\n\nCompared with wild-type AR , the Q865H variant exhibited reduced sensitivity to DHT and MPA in transactivation assays in MDA-MB-453 and PC-3 cells but did not respond to non-androgenic ligands or receptor antagonists .\n\nLigand binding , molecular modeling , mammalian two-hybrid and immunoblot assays revealed effects of the Q865H mutation on ligand dissociation , AR intramolecular interactions , and receptor stability .\n\nMicroarray expression profiling demonstrated that DHT and MPA regulate distinct transcriptional programs in MDA-MB-453 cells .\n\nGene Set Enrichment Analysis revealed that DHT- but not MPA-regulated genes were associated with estrogen-responsive transcriptomes from MCF-7 cells and the Wnt signaling pathway .\n\nThese findings suggest that the divergent proliferative responses of MDA-MB-453 cells to DHT and MPA result from the different genetic programs elicited by these two ligands through the AR-Q865H variant .\n\nThis work highlights the necessity to characterize additional models of molecular apocrine breast cancer to determine the precise role of AR signaling in this breast cancer subtype .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Metastatic epithelioid hemangioendothelioma of the kidney is a rare malignant vascular tumor with a wide spectrum of behaviors .\n\nCASE REPORT We present the case of a 53-year-old male patient with tumor metastases which developed after radical nephrectomy .\n\nWe describe the immunohistochemistry profile of the tumor and the successful long-term results of treatment with sunitinib , a multi-targeted tyrosine kinase inhibitor .\n\nAlso , we discuss the rationale for using this type of medication based on immunohistochemistry results .\n\nCONCLUSION To the best of our knowledge , this is the first reported case of metastatic renal epithelioid hemangioendothelioma treated successfully with sunitinib .", "output": "Activating invasion and metastasis" }, { "input": "Sirtuin proteins regulate diverse cellular pathways that influence genomic stability , metabolism and ageing .\n\nSIRT7 is a mammalian sirtuin whose biochemical activity , molecular targets and physiological functions have been unclear .\n\nHere we show that SIRT7 is an NAD(+)-dependent H3K18Ac ( acetylated lysine 18 of histone H3 ) deacetylase that stabilizes the transformed state of cancer cells .\n\nGenome-wide binding studies reveal that SIRT7 binds to promoters of a specific set of gene targets , where it deacetylates H3K18Ac and promotes transcriptional repression .\n\nThe spectrum of SIRT7 target genes is defined in part by its interaction with the cancer-associated E26 transformed specific ( ETS ) transcription factor ELK4 , and comprises numerous genes with links to tumour suppression .\n\nNotably , selective hypoacetylation of H3K18Ac has been linked to oncogenic transformation , and in patients is associated with aggressive tumour phenotypes and poor prognosis .\n\nWe find that deacetylation of H3K18Ac by SIRT7 is necessary for maintaining essential features of human cancer cells , including anchorage-independent growth and escape from contact inhibition .\n\nMoreover , SIRT7 is necessary for a global hypoacetylation of H3K18Ac associated with cellular transformation by the viral oncoprotein E1A .\n\nFinally , SIRT7 depletion markedly reduces the tumorigenicity of human cancer cell xenografts in mice .\n\nTogether , our work establishes SIRT7 as a highly selective H3K18Ac deacetylase and demonstrates a pivotal role for SIRT7 in chromatin regulation , cellular transformation programs and tumour formation in vivo .", "output": "Evading growth suppressors" }, { "input": "Elevated phosphorylation of estrogen receptor \u03b1 ( ER\u03b1 ) at serines 118 ( S118 ) and 167 ( S167 ) is associated with favorable outcome for tamoxifen adjuvant therapy and may serve as surrogate markers for a functional ER\u03b1 signaling pathway in breast cancer .\n\nIt is possible that loss of phosphorylation at S118 and/or S167 could disrupt ER\u03b1 signaling , resulting in aggressive ER\u03b1-independent breast cancer cells .\n\nTo this end , MCF-7 breast cancer cells were stably transfected with an ER\u03b1-specific short hairpin RNA that reduced endogenous ER\u03b1 .\n\nThe resulting cell line was stably transfected with wild-type ER\u03b1 ( ER-AB cells ) , or ER\u03b1 containing serine to alanine mutation at S118 or S167 ( S118A cells and S167A cells , respectively ) .\n\nThese stable cell lines expressed approximately equivalent ER\u03b1 compared with parental MCF-7 cells and were evaluated for growth , morphology , migration/invasion , and ER\u03b1-regulated gene expression .\n\nS118A cells and S167A cells exhibited increased growth and migration/invasion in vitro .\n\nForward- and side-scatter flow cytometry revealed that S167A cells were smaller in size , and both S118A and S167A cells exhibited less cellular complexity .\n\nS118A and S167A cells expressed pancytokeratin and membrane localization of \u03b2-catenin and did not express vimentin , indicating retention of epithelial lineage markers .\n\nExpression of ER\u03b1-target genes and other genes regulated by ER\u03b1 signaling or involved in breast cancer were markedly altered in both S118A and S167A cells .\n\nIn summary , attenuated phosphorylation of ER\u03b1 at S118 and S167 significantly affected cellular physiology and behavior in MCF-7 breast cancer cells , resulting in increased growth , migration/invasion , compromised expression of ER\u03b1 target genes , and markedly altered gene expression patterns .", "output": "Activating invasion and metastasis" }, { "input": "Hepatocellular carcinoma ( HCC ) is the fifth most common cancer worldwide .\n\nMajor risk factors of HCC include infection with hepatitis B or C viruses , alcohol and non-alcoholic fatty liver disease .\n\nHCC is difficult to diagnose at early stage , and has a very poor survival rate when diagnosed at a late stage .\n\nThe majority of HCC-related deaths result from local invasion ( to cause liver failure ) or distant metastases .\n\nThere is an urgent need to identify effective molecular targets for the treatment of the disease .\n\nAs the target of an established class of therapeutic agent thiazolidinediones ( TZDs ) , peroxisome-proliferator-activated receptor \u03b3 ( PPAR\u03b3 ) has been widely studied for its role in the development of HCC .\n\nA substantial body of evidence based on in vitro and in vivo models indicates that the activation of PPAR\u03b3 is able to inhibit HCC cell proliferation and tumor growth through inducing cell cycle arrest and apoptosis via the regulation of a panel of downstream effector molecules .\n\nPPAR\u03b3 activation also induces an inhibitory effect on HCC metastasis .\n\nMeanwhile , there is new evidence suggesting that PPAR\u03b3 inhibition could also be anti-tumorigenic .\n\nIn the present review , we summarize the available information on the role of PPAR\u03b3 in HCC development and spread , and discuss whether PPAR\u03b3 activation by TZDs could play a role in the treatment of HCC , summarizing both in vitro and in vivo .\n\nConsidering the available data , PPAR\u03b3 seems to exert beneficial effects against HCC and may therefore represent as a therapeutic target .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "MicroRNAs ( miRNAs ) are small noncoding RNAs , 19-24 nucleotides in length , that regulate gene expression and are expressed aberrantly in most types of cancer .\n\nMiRNAs also have been detected in the blood of cancer patients and can serve as circulating biomarkers .\n\nIt has been shown that secreted miRNAs within exosomes can be transferred from cell to cell and can regulate gene expression in the receiving cells by canonical binding to their target messenger RNAs .\n\nHere we show that tumor-secreted miR-21 and miR-29a also can function by another mechanism , by binding as ligands to receptors of the Toll-like receptor ( TLR ) family , murine TLR7 and human TLR8 , in immune cells , triggering a TLR-mediated prometastatic inflammatory response that ultimately may lead to tumor growth and metastasis .\n\nThus , by acting as paracrine agonists of TLRs , secreted miRNAs are key regulators of the tumor microenvironment .\n\nThis mechanism of action of miRNAs is implicated in tumor-immune system communication and is important in tumor growth and spread , thus representing a possible target for cancer treatment .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "Non-small cell lung cancer ( NSCLC ) , accounting for 80% of lung cancers , is the leading cause of all cancer deaths .\n\nPreviously , we demonstrated that delta-tocotrienol inhibits NSCLC cell proliferation , invasion and induces apoptosis by down-regulation of the Notch-1 signaling pathway .\n\nThe objective of this study was to investigate whether delta-tocotrienol , could enhance the anticancer effects of cisplatin .\n\nTreatment with a combination of delta-tocotrienol and cisplatin resulted in a dose-dependent , significant inhibition of cell growth , migration , invasiveness , and induction of apoptosis in NSCLC cells , as compared to the single agents .\n\nThis was associated with a decrease in NF-\u03baB DNA binding activity , decrease in Notch-1 , Hes-1 , Bcl-2 and increase in cleaved Caspase-3 and PARP expressions .\n\nThese results suggest that down-regulation of Notch-1 , via inhibition of NF-\u03baB signaling pathways by delta-tocotrienol and cisplatin , in combination , could provide a potential novel approach for tumor arrest in NSCLC , while lowering the effective dose of cisplatin .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Geraniol ( GOH ) , a naturally occurring monoterpene , has been shown to have antiproliferative , cell cycle arrest and apoptosis-inducing effects , and represents a promising cancer chemopreventive agent .\n\nIn the present study , we investigated the chemopreventive potential of GOH ( 50 and 100\u2009mg\u2009kg(-1) body weight ) against 7,12-dimethylbenz[a]anthracene ( DMBA)/12-O-tetradecanoylphorbol 13-acetate ( TPA)-mediated skin tumorigenesis in Swiss albino mice .\n\nThe topical treatment of GOH , 30\u2009min prior to TPA ( 2\u2009\u00b5g per 200\u2009\u00b5l of acetone ) treatment significantly inhibited TPA-induced skin edema , hyperplasia , COX-2 induction and oxidative stress response .\n\nThe GOH treatment also resulted in reduction of TPA-induced ornithine decarboxylase activity and [ (3) H ] thymidine incorporation by 53% ( P\u2009<\u20090.001 ) and 41% ( P\u2009<\u20090.001 ) , respectively .\n\nWe found that GOH treatment significantly inhibited the tumor incidence and number of tumors ( P\u2009<\u20090.001 ) and extended the latency period from 4\u2009weeks in DMBA/TPA treatment group to 10\u2009weeks in GOH-pretreated mice .\n\nFurthermore , we observed that GOH treatment significantly suppressed the Ras/Raf/ERK1/2 signaling pathway in skin tumor .\n\nConsistently , GOH-treated skin tumors showed reduced expression of Bcl-2 and increased expression of Bax in these lesions .\n\nThus , it was concluded that GOH inhibits DMBA/TPA-mediated skin tumorigenesis by attenuating the Ras proliferation pathway and inducing pro-apoptotic state via inhibition of oxidative stress response and inflammation .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Circulating tumour cells ( CTCs ) shed into blood from primary cancers include putative precursors that initiate distal metastases .\n\nAlthough these cells are extraordinarily rare , they may identify cellular pathways contributing to the blood-borne dissemination of cancer .\n\nHere , we adapted a microfluidic device for efficient capture of CTCs from an endogenous mouse pancreatic cancer model and subjected CTCs to single-molecule RNA sequencing , identifying Wnt2 as a candidate gene enriched in CTCs .\n\nExpression of WNT2 in pancreatic cancer cells suppresses anoikis , enhances anchorage-independent sphere formation , and increases metastatic propensity in vivo .\n\nThis effect is correlated with fibronectin upregulation and suppressed by inhibition of MAP3K7 ( also known as TAK1 ) kinase .\n\nIn humans , formation of non-adherent tumour spheres by pancreatic cancer cells is associated with upregulation of multiple WNT genes , and pancreatic CTCs revealed enrichment for WNT signalling in 5 out of 11 cases .\n\nThus , molecular analysis of CTCs may identify candidate therapeutic targets to prevent the distal spread of cancer .", "output": "Activating invasion and metastasis" }, { "input": "The apoptotic effects of interferon lambdas ( IFN\u03bbs ) have been described in several types of cancers .\n\nHowever , their effects on human lung cancer cells and the mechanisms are elusive .\n\nIn addition , the interaction between IFN\u03bbs and other interferons remains unclear .\n\nThe interplay between IFN\u03b1 and IFN\u03bb has been reported .\n\nHowever , although IFN\u03b3 is a well-known regulatory interferon , the mechanisms through which it regulates IFN\u03bbs in lung cancer cells are unknown .\n\nThese issues are critical for the application of IFN\u03bbs in lung cancer therapy .\n\nIn this study , we used A549 , a cell line derived from a human lung carcinoma , to characterize the antiproliferative and apoptotic effects of IFN\u03bbs on lung cancer , and the interplay between IFN\u03b3 and IFN\u03bb .\n\nBecause overexpression of full-length ectopic IFN\u03bbR1 led to cell death , we generated A549 cells stably expressing a chimeric receptor ( 10R1/\u03bbR1 ) , which is composed of the extracellular domain of IL-10 receptor ( IL10R1 ) fused in tandem to the transmembrane and intracellular domains of the IFN\u03bb receptor ( IFN\u03bbR1 ) .\n\nBy comparing with A549 cells stably expressing its cognate vector , we demonstrated that IL-10 stimulation triggered the intracellular IFN\u03bb signaling via 10R1/\u03bbR1 receptor .\n\nBy using A549 cells expressing 10R1/\u03bbR1 , we report that the IFN\u03bbR1 chain of IFN\u03bb receptor possesses an intrinsic ability to trigger apoptosis in human lung cancer cells .\n\nAlthough it did not suppress cell proliferation , IFN\u03bb signaling via 10R1/\u03bbR1 receptor induced cell cycle arrest , externalization of phosphatidylserine , DNA fragmentation , activation of caspase-3 , caspase-8 and caspase-9 .\n\nHowever , the caspase inhibitor Z-VAD-FMK did not prevent apoptosis .\n\nIn addition , the extent of induced apoptosis correlate with the expression levels of the IFN\u03bb receptor and the levels of STAT1 activation .\n\nLastly , we demonstrated that IFN\u03b3 sensitized A549 cells to IFN\u03bb-induced apoptosis , via upregulation of IFN\u03bbR1 .\n\nThese data indicate the potential of IFN\u03bb , alone or in combination with IFN\u03b3 , in the treatment of human lung carcinoma .", "output": "Resisting cell death" }, { "input": "Glycolysis is the initial step of glucose catabolism and is up-regulated in cancer cells ( the Warburg Effect ) .\n\nSuch shifts toward a glycolytic phenotype have not been explored widely in other biological systems , and the molecular mechanisms underlying the shifts remain unknown .\n\nWith proteomics , we observed increased glycolysis in disused human diaphragm muscle .\n\nIn disused muscle , lung cancer , and H(2)O(2)-treated myotubes , we show up-regulation of the rate-limiting glycolytic enzyme muscle-type phosphofructokinase ( PFKm , >2 fold , P<0.05 ) and accumulation of lactate ( >150% , P<0.05 ) .\n\nUsing microRNA profiling , we identify miR-320a as a regulator of PFKm expression .\n\nReduced miR-320a levels ( to \\u223c50% of control , P<0.05 ) are associated with the increased PFKm in each of these diverse systems .\n\nManipulation of miR-320a levels both in vitro and in vivo alters PFKm and lactate levels in the expected directions .\n\nFurther , miR-320a appears to regulate oxidative stress-induced PFKm expression , and reduced miR-320a allows greater induction of glycolysis in response to H(2)O(2) treatment .\n\nWe show that this microRNA-mediated regulation occurs through PFKm's 3 ' untranslated region and that Ets proteins are involved in the regulation of PFKm via miR-320a .\n\nThese findings suggest that oxidative stress-responsive microRNA-320a may regulate glycolysis broadly within nature .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "Vascular endothelial growth factor ( VEGF ) is one of the most important mediators of angiogenesis .\n\nSingle-chain ( sc)-VEGF protein containing an N-terminal Cys-tag has been designed for site-specific modification with a variety of imaging and therapeutic moieties .\n\nSite-specific labeling of scVEGF with thiol-reactive prosthetic group , N-[2-(4-(18)F-fluorobenzamido) ethyl ] maleimide ( [ (18)F]FBEM ) for positron emission tomography ( PET ) imaging of VEFGR may provide a new tracer which has great potential for clinical translation.Methods : [ (18)F]FBEM-scVEGF was synthesized by site-specific conjugation of ( 18)F-FBEM to a thiol group in Cys-tag of scVEGF at room temperature .\n\nThe functional activity after labeling was tested by immunofluorescence staining , cellular uptake and efflux .\n\nThe tumor targeting and in vivo properties were evaluated by biodistribution and microPET studies in tumor-bearing mice.Results : The radiolabeling yield and specific activity of [ (18)F]FBEM-scVEGF were 20.6 \ufffd 15.1% ( based on starting [ (18)F]FBEM , uncorrected , n = 5 ) and 58.8 \ufffd 12.4 GBq/\ufffdmol , respectively .\n\nNoninvasive microPET and direct tissue sampling experiments demonstrated that [ (18)F]FBEM-scVEGF had VEGFR specific tumor uptake in MDA-MB-435 , U87MG and 4T1 xenograft models .\n\nThe optimal tumor uptake was achieved at 2 h p.i. , which can be partially , but significantly blocked by co-injection of non-labeled scVEGF protein .\n\nOverall , [ (18)F]FBEM-scVEGF showed VEGFR specific tumor uptake.Conclusion : The scVEGF was site-specifically labeled with ( 18)F via [ (18)F]FBEM prosthetic group and the tracer [ (18)F]FBEM-scVEGF exhibited high receptor binding affinity and tumor targeting efficacy .\n\nFurther study of [ (18)F ] FBEM-scVEGF to evaluate angiogenesis in cancer and other disease types is warranted .", "output": "Inducing angiogenesis" }, { "input": "Epstein-Barr virus ( EBV ) induces an uncoordinated S-phase-like cellular environment coupled with multiple prophase-like events in cells replicating the virus .\n\nThe EBV encoded Ser/Thr kinase BGLF4 has been shown to induce premature chromosome condensation through activation of condensin and topoisomerase II and reorganization of the nuclear lamina to facilitate the nuclear egress of nucleocapsids in a pathway mimicking Cdk1 .\n\nHowever , the observation that RB is hyperphosphorylated in the presence of BGLF4 raised the possibility that BGLF4 may have a Cdk2-like activity to promote S-phase progression .\n\nHere , we investigated the regulatory effects of BGLF4 on cell cycle progression and found that S-phase progression and DNA synthesis were interrupted by BGLF4 in mammalian cells .\n\nExpression of BGLF4 did not compensate Cdk1 defects for DNA replication in S. cerevisiae .\n\nUsing time-lapse microscopy , we found the fate of individual HeLa cells was determined by the expression level of BGLF4 .\n\nIn addition to slight cell growth retardation , BGLF4 elicits abnormal chromosomal structure and micronucleus formation in 293 and NCP-TW01 cells .\n\nIn Saos-2 cells , BGLF4 induced the hyperphosphorylation of co-transfected RB , while E2F1 was not released from RB-E2F1 complexes .\n\nThe E2F1 regulated activities of the cyclin D1 and ZBRK1 promoters were suppressed by BGLF4 in a dose dependent manner .\n\nDetection with phosphoamino acid specific antibodies revealed that , in addition to Ser780 , phosphorylation of the DNA damage-responsive Ser612 on RB was enhanced by BGLF4 .\n\nTaken together , our study indicates that BGLF4 may directly or indirectly induce a DNA damage signal that eventually interferes with host DNA synthesis and delays S-phase progression .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "While human embryonic stem cells ( hESCs ) and human embryonal carcinoma cells ( hECCs ) have been studied extensively at the levels of the genome , transcriptome , proteome and epigenome our knowledge of their corresponding metabolomes is limited .\n\nHere , we present the metabolic signatures of hESCs and hESCs obtained by untargeted gas chromatography coupled to mass spectrometry ( GC-MS ) .\n\nWhilst some metabolites are common to both cell types , representing the self-renewal and house-keeping signatures , others were either higher ( e.g. , octadecenoic acid , glycerol-3-phosphate , 4-hydroxyproline ) or lower ( e.g. , glutamic acid , mannitol , malic acid , GABA ) in hESCs ( H9 ) compared to hECCs ( NTERA2 ) , these represent cell type specific signatures .\n\nFurther , our combined results of GC-MS and microarray based gene expression profiling of undifferentiated and OCT4-depleted hESCs are consistent with the Warburg effect which is increased glycolysis in embryonic cells and tumor cells in the presence of O(2) while oxidative phosphorylation ( OXPHOS ) is impaired or even shut down .\n\nRNAi-based OCT4 knock down mediated differentiation resulted in the activation of the poised OXPHOS machinery by expressing missing key proteins such as NDUFC1 , UQCRB and COX , increase in TCA cycle activity and decreased lactate metabolism .\n\nThese results shed light on the metabolite layer of pluripotent stem cells and could potentially establish novel metabolic markers of self renewal and pluripotency .", "output": "Cellular energetics" }, { "input": "OBJECTIVES The aim of this study was to immunohistochemically evaluate the expression of matrix metalloproteinase ( MMP)-1 , MMP- 2 , tissue inhibitor of metalloproteinase ( TIMP)-1 , TIMP-2 , and podoplanin in oral squamous cell carcinoma ( OSCC ) .\n\nImmunohistochemical staining of podoplanin-positive lymphatic vessel density ( LVD ) was also assessed .\n\nSTUDY DESIGN Forty cases of OSCC were analyzed by immunohistochemistry .\n\nRESULTS MMP-2 , MMP-10 , TIMP-1 , TIMP-2 , and podoplanin were detected in each of the 40 OSCC cases .\n\nThe expression of MMP-2 was significantly correlated with histologic grade .\n\nThe expression of podoplanin was positively correlated with gender and negatively correlated with tumor size .\n\nA significant positive correlation was also detected between LVD and the presence of lymph node metastases , gender , age , and diameter of the lymph node ( if involved ) , as well as histologic grade .\n\nCONCLUSIONS The results are suggestive of important roles that MMP-2 , MMP-10 , TIMP-2 , and podoplanin play in pathologic processes of OSCC , including invasion .\n\nOur findings also suggest that LVD may play a role in lymphatic metastasis and tumor progression .", "output": "Activating invasion and metastasis" }, { "input": "Bone marrow necrosis ( BMN ) is a rare finding in children with malignancy occurring most commonly in children with acute lymphoblastic leukemia .\n\nThis article describes the first case of a girl who developed BMN during treatment for Hodgkin's disease .\n\nDuring the second cycle of chemotherapy , she experienced sudden profound bone pain in the lumbosacral region associated with elevated levels of lactate dehydrogenase ( LDH ) , fibrin degradation products ( D-Dimer ) , and alkaline phosphatase as well as pancytopenia and leukoerythroblastosis .\n\nMRI studies showed multiple confluent areas with low signal intensity and rim contrast enhancement in all vertebral bodies .\n\nBone marrow biopsy revealed focal necrosis within hypocellular bone marrow .\n\nThe patient responded quickly to symptomatic treatment with analgetics and heparin ; however , elevations of LDH and D-Dimer persisted for 1.5 and 8 months , respectively .\n\nClinicians should be aware of this rare condition to establish the diagnosis and to continue oncologic treatment as early as possible .", "output": "Resisting cell death" }, { "input": "Inhibition of cell proliferation by fenoterol and fenoterol derivatives in 1321N1 astrocytoma cells is consistent with \u03b2(2)-adrenergic receptor ( \u03b2(2)-AR ) stimulation .\n\nHowever , the events that result in fenoterol-mediated control of cell proliferation in other cell types are not clear .\n\nHere , we compare the effect of the \u03b2(2)-AR agonists ( R,R')-fenoterol ( Fen ) and ( R,R')-4-methoxy-1-naphthylfenoterol ( MNF ) on signaling and cell proliferation in HepG2 hepatocarcinoma cells by using Western blotting and [ (3)H]thymidine incorporation assays .\n\nDespite the expression of \u03b2(2)-AR , no cAMP accumulation was observed when cells were stimulated with isoproterenol or Fen , although the treatment elicited both mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt activation .\n\nUnexpectedly , isoproterenol and Fen promoted HepG2 cell growth , but MNF reduced proliferation together with increased apoptosis .\n\nThe mitogenic responses of Fen were attenuated by 3-(isopropylamino)-1-[(7-methyl-4-indanyl)oxy]butan-2-ol ( ICI 118,551 ) , a \u03b2(2)-AR antagonist , whereas those of MNF were unaffected .\n\nBecause of the coexpression of \u03b2(2)-AR and cannabinoid receptors ( CBRs ) and their impact on HepG2 cell proliferation , these G\u03b1(i)/G\u03b1(o)-linked receptors may be implicated in MNF signaling .\n\nCell treatment with ( R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-napthalenylmethanone ( WIN 55,212-2 ) , a synthetic agonist of CB(1)R and CB(2)R , led to growth inhibition , whereas inverse agonists of these receptors blocked MNF mitogenic responses without affecting Fen signaling .\n\nMNF responses were sensitive to pertussis toxin .\n\nThe \u03b2(2)-AR-deficient U87MG cells were refractory to Fen , but responsive to the antiproliferative actions of MNF and WIN 55,212-2 .\n\nThe data indicate that the presence of the naphthyl moiety in MNF results in functional coupling to the CBR pathway , providing one of the first examples of a dually acting \u03b2(2)-AR-CBR ligand .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Focal inflammation causes systemic fever .\n\nCancer hyperthermia therapy results in shrinkage of tumors by various mechanisms , including induction of adaptive immune response .\n\nHowever , the physiological meaning of systemic fever and mechanisms of tumor shrinkage by hyperthermia have not been completely understood .\n\nIn this study , we investigated how heat shock influences the adaptive immune system .\n\nWe established a cytotoxic T lymphocyte ( CTL ) clone ( #IM29 ) specific for survivin , one of the tumor-associated antigens ( TAAs ) , from survivin peptide-immunized cancer patients ' peripheral blood , and the CTL activities were investigated in several temperature conditions ( 37-41\ufffdC ) .\n\nCytotoxicity and IFN-\u03b3 secretion of CTL were greatest under 39\ufffdC condition , whereas they were minimum under 41\ufffdC .\n\nTo address the molecular mechanisms of this phenomenon , we investigated the apoptosis status of CTLs , expression of CD3 , CD8 , and TCR\u03b1\u03b2 by flow cytometry , and expression of perforin , granzyme B , and Fas ligand by western blot analysis .\n\nThe expression of perforin and granzyme B were upregulated under temperature conditions of 39 and 41\ufffdC .\n\nOn the other hand , CTL cell death was induced under 41\ufffdC condition with highest Caspase-3 activity .\n\nTherefore , the greatest cytotoxicity activity at 39\ufffdC might depend on upregulation of cytotoxic granule proteins including perforin and granzyme B. These results suggest that heat shock enhances effector phase of the adaptive immune system and promotes eradication of microbe and tumor cells .", "output": "Resisting cell death" }, { "input": "Background .\n\nOver the past ten years oncological outcomes achieved by local excision techniques ( LETs ) as the sole treatment for early stages of rectal cancer ( ESRC ) have been often disappointing .\n\nThe reasons for these poor results lie mostly in the high risk of the disease's diffusion to local-regional lymph nodes even in ESRC .\n\nAims .\n\nThis study aims to find the correct indications for LET in ESRC taking into consideration clinical-pathological features of tumours that may reduce the risk of lymph node metastasis to zero .\n\nMethods .\n\nSystematic literature review and meta-analysis of casistics of ESRC treated with total mesorectal excision with the aim of identifying risk factors for nodal involvement .\n\nResults .\n\nThe risk of lymph node metastasis is higher in G \u2265 2 and T \u2265 2 tumours with lymphatic and/or vascular invasion .\n\nOther features which have not yet been sufficiently investigated include female gender , TSM stage >1 , presence of tumour budding and/or perineural invasion .\n\nConclusions .\n\nResults comparable to radical surgery can be achieved by LET only in patients with T(1) N(0) G(1) tumours with low-risk histological features , whereas deeper or more aggressive tumours should be addressed by radical surgery ( RS ) .", "output": "Activating invasion and metastasis" }, { "input": "Src kinase is elevated in breast tumors that are ER/PR negative and do not overexpress HER2 , but clinical trials with Src inhibitors have shown little activity .\n\nThe present study evaluated preclinical efficacy of a novel peptidomimetic compound , KX-01 ( KX2-391 ) , that exhibits dual action as an Src and pretubulin inhibitor .\n\nKX-01 was evaluated as a single-agent and in combination with paclitaxel in MDA-MB-231 , MDA-MB-157 , and MDA-MB-468 human ER/PR/HER2-negative breast cancer cells .\n\nTreatments were evaluated by growth/apoptosis , isobologram analysis , migration/invasion assays , tumor xenograft volume , metastasis , and measurement of Src , focal adhesion kinase ( FAK ) , microtubules , Ki67 , and microvessel density .\n\nKX-01 inhibited cell growth in vitro and in combination with paclitaxel resulted in synergistic growth inhibition .\n\nKX-01 resulted in a dose-dependent inhibition of MDA-MB-231 and MDA-MB-157 tumor xenografts ( 1 and 5 mg/kg , twice daily ) .\n\nKX-01 inhibited activity of Src and downstream mediator FAK in tumors that was coincident with reduced proliferation and angiogenesis and increased apoptosis .\n\nKX01 also resulted in microtubule disruption in tumors .\n\nCombination of KX-01 with paclitaxel resulted in significant regression of MDA-MB-231 tumors and reduced metastasis to mouse lung and liver .\n\nKX-01 is a potently active Src/pretubulin inhibitor that inhibits breast tumor growth and metastasis .\n\nAs ER/PR/HER2-negative patients are candidates for paclitaxel therapy , combination with KX-01 may potentiate antitumor efficacy in management of this aggressive breast cancer subtype .", "output": "Activating invasion and metastasis, Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "Lentinula edodes mycelia ( L.E.M. ) is a dried powder extracted from shiitake mushrooms ( Lentinula edodes ) .\n\nWe previously demonstrated that it has immunomodulatory effects .\n\nIn this paper , the direct cytotoxic effects of the polysaccharide-rich fraction of L.E.M .\n\n( L.E.M. ethanol precipitate ; LEP ) on HepG2 human hepatocellular carcinoma ( HCC ) cells were investigated .\n\nLEP directly killed the HepG2 cells efficaciously , but had only minor effects on normal rat hepatocytes and normal mouse dermal cells under the same conditions .\n\nCharacteristic morphological changes associated with apoptosis such as shrinkage , rounding , and floating as well as chromatin condensation were confirmed ; terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling ( TUNEL ) staining was positive as determined by fluorescence microscopy analyses .\n\nThe caspase-3 and -8 death receptor pathway was found largely responsible for the apoptotic death of HepG2 cells treated with LEP .\n\nIn conclusion , LEP can directly induce apoptosis of HepG2 cells , and thus may have potential chemotherapeutic applications for the treatment of HCC .", "output": "Resisting cell death" }, { "input": "Epigenetic deregulation of gene expression has a role in the initiation and progression of prostate cancer ( PCa ) .\n\nThe histone methyltransferase MMSET/WHSC1 ( Multiple Myeloma SET domain ) is overexpressed in a number of metastatic tumors , but its mechanism of action has not been defined .\n\nIn this work , we found that PCa cell lines expressed significantly higher levels of MMSET compared with immortalized , non-transformed prostate cells .\n\nKnockdown experiments showed that , in metastatic PCa cell lines , dimethylation of lysine 36 and trimethylation of lysine 27 on histone H3 ( H3K36me2 and H3K27me3 , respectively ) depended on MMSET expression , whereas depletion of MMSET in benign prostatic cells did not affect chromatin modifications .\n\nKnockdown of MMSET in DU145 and PC-3 tumor cells decreased cell proliferation , colony formation in soft agar and strikingly diminished cell migration and invasion .\n\nConversely , overexpression of MMSET in immortalized , non-transformed RWPE-1 cells promoted cell migration and invasion , accompanied by an epithelial-mesenchymal transition ( EMT ) .\n\nAmong a panel of EMT-promoting genes analyzed , TWIST1 expression was strongly activated in response to MMSET .\n\nChromatin immunoprecipitation analysis demonstrated that MMSET binds to the TWIST1 locus and leads to an increase in H3K36me2 , suggesting a direct role of MMSET in the regulation of this gene .\n\nDepletion of TWIST1 in MMSET-overexpressing RWPE-1 cells blocked cell invasion and EMT , indicating that TWIST1 was a critical target of MMSET , responsible for the acquisition of an invasive phenotype .\n\nCollectively , these data suggest that MMSET has a role in PCa pathogenesis and progression through epigenetic regulation of metastasis-related genes .", "output": "Activating invasion and metastasis" }, { "input": "MicroRNAs ( miRs ) are small non-coding RNAs that recently emerged as potent regulators of gene expression .\n\nThe members of the miR-17-92 cluster have been shown to control endothelial cell functions and neovascularization ; however , the regulation and function of the cluster in endothelial cell lineage commitment has not been explored .\n\nThis project aimed to test the role of the miR-17-92 cluster during endothelial differentiation .\n\nWe demonstrate that miR-17 , miR-18 , miR-19 and miR-20 are increased upon the induction of endothelial cell differentiation of murine embryonic stem cells or induced pluripotent stem cells .\n\nIn contrast , miR-92a and the primary miR-17-92 transcript were downregulated .\n\nThe inhibition of each individual miR of the cluster by cholesterol-modified antagomirs did not affect endothelial marker gene expression .\n\nMoreover , the combination of all antagomirs had no effect .\n\nThese findings illustrate that although the miR-17-92 cluster regulates vascular integrity and angiogenesis , none of the members has a significant impact on the endothelial differentiation of pluripotent stem cells .", "output": "Inducing angiogenesis" }, { "input": "Fagonia indica is a small spiny shrub of great ethnopharmacological importance in folk medicine .\n\nThe aqueous decoction of aerial parts is a popular remedy against various skin lesions , including cancer .\n\nWe used a biological activity-guided fractionation approach to isolate the most potent fraction of the crude extract on three cancer cell lines : MCF-7 oestrogen-dependent breast cancer , MDA-MB-468 oestrogen-independent breast cancer , and Caco-2 colon cancer cells .\n\nA series of chromatographic and spectroscopic procedures were utilised on the EtOAc fraction , which resulted in the isolation of a new steroidal saponin glycoside .\n\nThe cytotoxic activity of the saponin glycoside was determined in cancer cells using the MTT and neutral red uptake assays .\n\nAfter 24h treatment , the observed IC(50) values of the saponin glycoside were 12.5 \u03bcM on MDA-MB-468 and Caco-2 cells , but 100 \u03bcM on MCF-7 cells .\n\nSeveral lines of evidence : PARP cleavage , caspase-3 cleavage , DNA ladder assays , and reversal of growth inhibition with the pan-caspase inhibitor Z-VAD-fmk , suggested stimulation of apoptosis in MDA-MB-468 and Caco-2 cells , but not in MCF-7 cells , which do not express caspase-3 .\n\nThe haemolytic activity of the saponin glycoside was confirmed in sheep red blood cells , with cell lysis observed at >100 \u03bcM , suggesting that , at this concentration , the saponin glycoside caused necrosis through cell lysis in MCF-7 cells .\n\nUsing the DNA ladder assay , the saponin glycoside ( 12.5 \u03bcM ) was not toxic to HUVEC ( human umbilical vein endothelial cells ) or U937 cells , indicating some selectivity between malignant and normal cells .\n\nWe conclude that the steroidal saponin glycoside isolated from F. indica is able to induce apoptosis or necrosis in cancer cells depending on the cell type .", "output": "Resisting cell death" }, { "input": "Angiogenesis is critical for cancer growth and metastasis .\n\nSteps of angiogenesis are energy consuming , while vascular endothelial cells are highly glycolytic .\n\nGlioblastoma multiforme ( GBM ) is a highly vascular tumor and this enhances its aggressiveness .\n\nD-amino acid oxidase ( DAO ) is a promising therapeutic protein that induces oxidative stress upon acting on its substrates .\n\nOxidative stress-energy depletion ( OSED ) therapy was recently reported ( El Sayed et al. , Cancer Gene Ther , 19 , 1-18 , 2012 ) .\n\nOSED combines DAO-induced oxidative stress with energy depletion caused by glycolytic inhibitors such as 3-bromopyruvate ( 3BP ) , a hexokinase II inhibitor that depleted ATP in cancer cells and induced production of hydrogen peroxide. 3BP disturbs the Warburg effect and antagonizes effects of lactate and pyruvate ( El Sayed et al. , J Bioenerg Biomembr , 44 , 61-79 , 2012 ) .\n\nCitrate is a natural organic acid capable of inhibiting glycolysis by targeting phosphofructokinase .\n\nHere , we report that DAO , 3BP and citrate significantly inhibited angiogenesis , decreased the number of vascular branching points and shortened the length of vascular tubules .\n\nOSED delayed the growth of C6/DAO glioma cells. 3BP combined with citrate delayed the growth of C6 glioma cells and decreased significantly the number and size of C6 glioma colonies in soft agar .\n\nHuman GBM cells ( U373MG ) were resistant to chemotherapy e.g. cisplatin and cytosine arabinoside , while 3BP was effective in decreasing the viability and disturbing the morphology of U373MG cells .", "output": "Inducing angiogenesis" }, { "input": "OBJECTIVE To evaluate the Expression and correlation of cyclooxygenase-2 ( COX-2 ) and vascular endothelial growth factor receptor ( VEGF ) in nasopharyngeal carcinoma .\n\nMETHOD In this study , expression levels of COX-2 , VEGF were examined in 58 patients with nasopharyngeal carcinoma and 38 patients with inflammation in nasopharyngeal mucosa by immunohistochemistry method .\n\nRESULT The expression of COX-2 , VEGF were higher in nasopharyngeal carcinoma than those in nasopharyngeal mucosa ( P < 0.05 ) , and they had some correlation with the invasion and lymphatic metastasis and with the clinical stage of nasopharyngeal carcinoma ( P < 0.05 ) .\n\nThe expression of COX-2 was positively correlated with that of VEGF ( P < 0.05 ) .\n\nCONCLUSION The coexpression of COX-2 and VEGF may play animportant role in the carcinogenesis and development of nasopharyngeal carcinoma , and they may prom ( see text ) lymph node metastasis of nasopharyngeal carcinoma .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "BACKGROUND It has been proven that metastasis-associated in colon cancer 1 ( MACC1 ) is a new gene that is related to the invasion and metastasis of tumors .\n\nMACC1 also regulates c-met expression .\n\nThe aim of this study is to explore the expressions of MACC1 and hepatocyte growth factor receptor ( c-met ) , and its relationship with invasion , metastasis , and prognosis of non-small cell lung cancer ( NSCLC ) .\n\nMETHODS MACC1 and c-met expressions were detected in 103 cases of NSCLC and 40 cases of neighboring normal lung cancer tissue using immunohistochemistry .\n\nRESULTS MACC1 and c-met expressions were significantly higher in lung cancer tissues than that in neighboring normal tissue ( P<0.001 ) .\n\nMACC1 and c-met expressions were associated with poor differentiation , advanced T stages , lymph node metastasis , and advanced TNM stages ( P<0.05 ) of NSCLC , but not with sex , age , smoking , and histological classification ( P>0.05 ) .\n\nIn addition , a positive correlation between MACC1 and c-met expressions was observed ( r=0.403 , P<0.001 ) .\n\nThe result from the Kaplan-Meier survival analysis showed that the five-year survival rate in patients with positive MACC1 and c-met expressions was remarkanly lower than that in patients with negative expressions ( P<0.05 ) .\n\nThe result from the Cox regression analysis showed that MACC1 expression was an independent prognostic factor for NSCLC ( P=0.026 ) .\n\nCONCLUSIONS MACC1 and c-met have an important function in the differentiation , invasion , and metastasis of NSCLC .\n\nMACC1 and c-met have poor prognosis in patients with NSCLC .\n\nMoreover , MACC1 expression is an independent prognostic factor for NSCLC .", "output": "Activating invasion and metastasis" }, { "input": "Epigenetic changes in pediatric neuroblastoma may contribute to the aggressive pathophysiology of this disease , but little is known about the basis for such changes .\n\nIn this study , we examined a role for the DNA methyltransferase DNMT3B , in particular , the truncated isoform DNMT3B7 , which is generated frequently in cancer .\n\nTo investigate if aberrant DNMT3B transcripts alter DNA methylation , gene expression , and phenotypic character in neuroblastoma , we measured DNMT3B expression in primary tumors .\n\nHigher levels of DNMT3B7 were detected in differentiated ganglioneuroblastomas compared to undifferentiated neuroblastomas , suggesting that expression of DNMT3B7 may induce a less aggressive clinical phenotype .\n\nTo test this hypothesis , we investigated the effects of enforced DNMT3B7 expression in neuroblastoma cells , finding a significant inhibition of cell proliferation in vitro and angiogenesis and tumor growth in vivo .\n\nDNMT3B7-positive cells had higher levels of total genomic methylation and a dramatic decrease in expression of the FOS and JUN family members that comprise AP1 transcription factors .\n\nConsistent with an established antagonistic relationship between AP1 expression and retinoic acid receptor activity , increased differentiation was seen in the DNMT3B7-expressing neuroblastoma cells following treatment with all-trans retinoic acid ( ATRA ) compared to controls .\n\nOur results indicate that DNMT3B7 modifies the epigenome in neuroblastoma cells to induce changes in gene expression , inhibit tumor growth , and increase sensitivity to ATRA .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Tumor cells are surrounded by infiltrating inflammatory cells , such as lymphocytes , neutrophils , macrophages , and mast cells .\n\nA body of evidence indicates that mast cells are associated with various types of tumors .\n\nAlthough role of mast cells can be directly related to their granule content , their function in angiogenesis and tumor progression remains obscure .\n\nThis study aims to understand the role of mast cells in these processes .\n\nTumors were chemically induced in BALB/c mice and tumor progression was divided into Phases I , II and III .\n\nPhase I tumors exhibited a large number of mast cells , which increased in phase II and remained unchanged in phase III .\n\nThe expression of mouse mast cell protease ( mMCP)-4 , mMCP-5 , mMCP-6 , mMCP-7 , and carboxypeptidase A were analyzed at the 3 stages .\n\nOur results show that with the exception of mMCP-4 expression of these mast cell chymase ( mMCP-5 ) , tryptases ( mMCP-6 and 7 ) , and carboxypeptidase A ( mMC-CPA ) increased during tumor progression .\n\nChymase and tryptase activity increased at all stages of tumor progression whereas the number of mast cells remained constant from phase II to III .\n\nThe number of new blood vessels increased significantly in phase I , while in phases II and III an enlargement of existing blood vessels occurred .\n\nIn vitro , mMCP-6 and 7 are able to induce vessel formation .\n\nThe present study suggests that mast cells are involved in induction of angiogenesis in the early stages of tumor development and in modulating blood vessel growth in the later stages of tumor progression .", "output": "Inducing angiogenesis" }, { "input": "Semaphorin 5A , a member of semaphorin family , was originally identified as axonal guidance factor functioning during neuronal development .\n\nPreviously , we showed that the expression of semaphorin 5A might contribute to the metastasis of gastric cancer .\n\nHowever , its functional roles and mechanism(s) in invasion and metastasis of gastric cancer remain unclear .\n\nBy using human gastric caner cell lines Parental SGC7901 , SGC7901-siScrambled and SGC7901-siSema 5A , we found that semaphorin 5A significantly promoted the invasive and metastatic abilities of gastric cancer cell in vitro .\n\nSemaphorin 5A increased the expression of MMP9 by activating phosphorylated ErK1/2 in gastric cancer cell .\n\nFurthermore , MEK inhibitor PD98059 and MMP9 antibody ( Ab ) significantly inhibited in vitro invasive and metastatic abilities induced by semaphorin 5A .\n\nTaken together , the present work revealed a novel function of semaphorin 5A that the existence of semaphorin 5A could promote invasion and metastasis of gastric cancer by regulating MMP9 expression , at least partially , via the MEK/ERKs signal transduction pathway .\n\nSemaphorin 5A and its regulated molecules could be the potential targets for cancer therapy .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Gadd45a , the first well-defined p53 downstream gene , can be induced by multiple DNA-damaging agents , which plays important roles in the control of cell cycle checkpoint , DNA repair process and signaling transduction .\n\nOur previous findings suggested that Gadd45a maintains cell-cell adhesion and cell contact inhibition .\n\nHowever , little is known about how Gadd45a participates in the suppression of malignancy in human cancer cells .\n\nTo examine the functions of Gadd45a in cell invasion and metastasis , we performed the adhesion , wound-healing and transwell assays in Gadd45a ( +/+ ) and Gadd45a ( -/- ) MEF cell lines .\n\nWe found the adhesion , migration and invasive abilities were much higher in Gadd45a deficient cells .\n\nWe furthermore applied high-throughput cDNA microarray analysis and bioinformatics analysis to analyze the mechanisms of Gadd45a gene in invasion and metastasis .\n\nCompared with the Gadd45a wild type cells , the Gadd45a deficient cells showed a wide range of transcripts alterations .\n\nThe altered gene pathways were predicted by the MAS software , which indicated focal adhesion,cell communication,ECM-receptor interaction as the three main pathways .\n\nReal-time PCR was employed to validate the differentially expressed genes .\n\nInterestingly , we figured out that the deregulations of these genes are caused neither by genomic aberrations nor methylation status .\n\nThese findings provided a novel insight that Gadd45a may involve in tumor progression by regulating related genes expressions .", "output": "Activating invasion and metastasis" }, { "input": "Genetic and epigenetic changes in the von Hippel-Lindau ( VHL ) tumour suppressor gene are common in sporadic conventional ( clear cell ) renal cell carcinoma ( ccRCC ) .\n\nThe effects on VHL expression are unknown but increased understanding may be relevant clinically , either in terms of prognosis or in therapy selection .\n\nWe have examined the expression of VHL mutant RNA in 84 ccRCC tumours previously screened for mutations in genomic DNA , 56 of which contained 52 unique mutations or polymorphisms .\n\nBased on the predicted change to the primary amino acid sequence , 24 of the mutations were missense , 11 resulted in frameshifts with premature truncation , 9 resulted in immediate truncation at the site of the mutation and 2 were frameshifts which extended the reading frame beyond the normal stop codon .\n\nNine tumours had intronic variants , including substitution of invariant residues at splice sites , deletion of nucleotides spanning the exon-intron junction , an intronic variant of unknown function and the polymorphism c.463+43A>G .\n\nFour variants were identified which were present in genomic DNA but not in mRNA .\n\nThree of these , all encoding apparent missense changes to the primary amino acid sequence , were located close to the ends of exons , reduced the strength of the splice site and function as null rather than missense variants .\n\nOne nonsense variant was not detectable in mRNA but all other mutations resulting in premature truncation codons ( PTCs ) were , suggesting truncating VHL mutations may potentially generate truncated VHL protein .\n\nAn intronic variant , c.341\\u201111T>A , previously regarded as of unknown function , is associated with an increased level of skipping of exon 2 and may , therefore , reduce production of pVHL .\n\nOur data show that the biological consequences of VHL mutations are not necessarily predictable from the sequence change of the mutation and that for the majority of VHL mutations , the potential for the generation of mutant protein exists .", "output": "Genomic instability and mutation" }, { "input": "Although interleukin-28A ( IL-28A ) is believed to have an antiviral effect , its role in tumor migration requires further examination .\n\nThe present study was intended to verify the effect of IL-28A on the migration of UMUC-3 bladder cancer cells .\n\nIL-28A and its receptor IL-28AR1 mRNA were detected in UMUC-3 cells .\n\nAlthough exogenous IL-28A showed no effect on cell proliferation , a wound-healing migration assay showed that the migration of UMUC-3 cells was induced by IL-28A .\n\nFurthermore , treatment of the cells with IL-28A significantly promoted MMP-9 expression via binding activities of NF-\u03baB and AP-1 .\n\nIL-28A also induced the activation of p38MAPK and Jak2-Stat2 signaling .\n\nUsing the p38MAPK inhibitor SB203580 and the dominant-negative plasmid DN-p38 , we found evidence that the inhibition of p38MAPK signaling suppressed the effects of IL-28A including wound-healing migration and MMP-9 expression by activation of NF-\u03baB and AP-1 binding in UMUC-3 cells .\n\nHowever , Jak-2 inhibition by AG490 did not affect IL-28A-induced migration of UMUC-3 cells .\n\nCollectively , we suggest for the first time that the p38MAPK pathway mediates IL-28A-induced cell migration through MMP-9 expression by activating NF-\u03baB and AP-1 binding motifs .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Epithelial-to-mesenchymal transition ( EMT ) in cancer cells is considered to be a prerequisite for acquiring invasive/migratory phenotype and subsequent metastasis .\n\nThis study provides molecular evidence associated with the antimetastatic effect of black tea polyphenol extracts ( BTE ) , which contain polyphenols including gallic acid , gallocatechin , catechin , epigallocatechin-3-gallate , epicatechin-3-gallate , and theaflavin 3,3'-digallate , in an an oral squamous cell culture system by showing a nearly complete inhibition on the invasion ( p < 0.001 ) of SCC-4 cells via reduced activities of MMP-2 ( p < 0.001 ) and u-PA ( p < 0.001 ) .\n\nImmunoblot was performed to find that BTE could induce up-regulation of epithelial markers such as E-cadherin and inhibit mesenchymal markers such as snail-1 and vimentin .\n\nBTE inhibited p-FAK and p-paxillin , indicating the anti-EMT effect of BTE in oral squamous cell carcinoma .\n\nBTE was evidenced by its inhibition of the tumor growth of SCC-4 cells via cancer cell xenografted nude mice mode .\n\nThese results suggested that BTE could reduce invasion by reversing EMT in human oral cancer cells .", "output": "Activating invasion and metastasis" }, { "input": "We have evaluated DNA damage ( DNA adduct formation ) after feeding benzo[a]pyrene ( BP ) to wild-type ( WT ) and cancer-susceptible Xpa(-/-)p53(+/-) mice deficient in nucleotide excision repair and haploinsufficient for the tumor suppressor p53 .\n\nDNA damage was evaluated by high-performance liquid chromatography/electrospray ionization tandem mass spectrometry ( HPLC/ES-MS/MS ) , which measures r7,t8,t9-trihydroxy-c-10-(N ( 2)-deoxyguanosyl)-7,8,9,10-tetrahydrobenzo[a]pyrene ( BPdG ) , and a chemiluminescence immunoassay ( CIA ) , using anti-r7,t8-dihydroxy-t-9,10-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene ( BPDE)-DNA antiserum , which measures both BPdG and the other stable BP-DNA adducts .\n\nWhen mice were fed 100 ppm BP for 28 days , BP-induced DNA damage measured in esophagus , liver and lung was typically higher in Xpa(-/-)p53(+/-) mice , compared with WT mice .\n\nThis result is consistent with the previously observed tumor susceptibility of Xpa(-/-)p53(+/-) mice .\n\nBPdG , the major DNA adduct associated with tumorigenicity , was the primary DNA adduct formed in esophagus ( a target tissue in the mouse ) , whereas total BP-DNA adducts predominated in higher levels in the liver ( a non-target tissue in the mouse ) .\n\nIn an attempt to lower BP-induced DNA damage , we fed the WT and Xpa(-/-)p53(+/-) mice 0.3% chlorophyllin ( CHL ) in the BP-containing diet for 28 days .\n\nThe addition of CHL resulted in an increase of BP-DNA adducts in esophagus , liver and lung of WT mice , a lowering of BPdG in esophagi of WT mice and livers of Xpa(-/-)p53(+/-) mice and an increase of BPdG in livers of WT mice .\n\nTherefore , the addition of CHL to a BP-containing diet showed a lack of consistent chemoprotective effect , indicating that oral CHL administration may not reduce PAH-DNA adduct levels consistently in human organs .", "output": "Genomic instability and mutation" }, { "input": "The Alternative Lengthening of Telomeres ( ALT ) pathway is a telomerase-independent pathway for telomere maintenance that is active in a significant subset of human cancers and in vitro immortalized cell lines .\n\nALT is thought to involve templated extension of telomeres through homologous recombination , but the genetic or epigenetic changes that unleash ALT are not known .\n\nRecently , mutations in the ATRX/DAXX chromatin remodeling complex and histone H3.3 were found to correlate with features of ALT in pancreatic neuroendocrine cancers , pediatric glioblastomas , and other tumors of the central nervous system , suggesting that these mutations might contribute to the activation of the ALT pathway in these cancers .\n\nWe have taken a comprehensive approach to deciphering ALT by applying genomic , molecular biological , and cell biological approaches to a panel of 22 ALT cell lines , including cell lines derived in vitro .\n\nHere we show that loss of ATRX protein and mutations in the ATRX gene are hallmarks of ALT-immortalized cell lines .\n\nIn addition , ALT is associated with extensive genome rearrangements , marked micronucleation , defects in the G2/M checkpoint , and altered double-strand break ( DSB ) repair .\n\nThese attributes will facilitate the diagnosis and treatment of ALT positive human cancers .", "output": "Genomic instability and mutation, Enabling replicative immortality, Evading growth suppressors" }, { "input": "Nonmelanoma skin cancer ( NMSC ) is by far the most frequent type of cancer in humans .\n\nNMSC includes several types of malignancies with different clinical outcomes , the most frequent being basal and squamous cell carcinomas .\n\nWe have used the Sleeping Beauty transposon/transposase system to identify somatic mutations associated with NMSC .\n\nTransgenic mice bearing multiple copies of a mutagenic Sleeping Beauty transposon T2Onc2 and expressing the SB11 transposase under the transcriptional control of regulatory elements from the keratin K5 promoter were treated with TPA , either in wild-type or Ha-ras mutated backgrounds .\n\nAfter several weeks of treatment , mice with transposition developed more malignant tumors with decreased latency compared with control mice .\n\nTransposon/transposase animals also developed basal cell carcinomas .\n\nGenetic analysis of the transposon integration sites in the tumors identified several genes recurrently mutated in different tumor samples , which may represent novel candidate cancer genes .\n\nWe observed alterations in the expression levels of some of these genes in human tumors .\n\nOur results show that inactivating mutations in Notch1 and Nsd1 , among others , may have an important role in skin carcinogenesis .", "output": "Genomic instability and mutation" }, { "input": "A 34-year-old Japanese woman presented with left supraclavicular lymph node swelling .\n\nComputed tomography scans revealed a mass on the left lower lobe , pulmonary nodules , and pleural effusion .\n\nA lymph node biopsy revealed large-cell carcinoma with an epidermal growth factor receptor ( EGFR ) deletion mutation , L747-T751 in exon 19 .\n\nAlthough malignant pleural effusions carried the same EGFR mutation , progressive pleural effusions after treatment with chemotherapy , gefitinib , and erlotinib did not show any EGFR mutation .\n\nA cell line established from the pleural effusion 3 days before the patient expired also did not harbor the EGFR mutation .\n\nHistological sections of the lymph node of the patient were similar to those of the xenograft tumor of the cell line .\n\nThere may be genetic heterogeneity in EGFR mutant tumors .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "BACKGROUND Colorectal cancer is one of the main cancers in the Western world .\n\nAbout 90% of the deaths arise from formation of distant metastasis .\n\nThe expression of the newly identified gene metastasis associated in colon cancer 1 ( MACC1 ) is a prognostic indicator for colon cancer metastasis .\n\nHere , we analyzed for the first time the impact of single nucleotide polymorphisms ( SNPs ) in the coding region of MACC1 for clinical outcome of colorectal cancer patients .\n\nAdditionally , we screened met proto-oncogene ( Met ) , the transcriptional target gene of MACC1 , for mutations .\n\nMETHODS We sequenced the coding exons of MACC1 in 154 colorectal tumors ( stages I , II and III ) and the crucial exons of Met in 60 colorectal tumors ( stages I , II and III ) .\n\nWe analyzed the association of MACC1 polymorphisms with clinical data , including metachronous metastasis , UICC stages , tumor invasion , lymph node metastasis and patients ' survival ( n = 154 , stages I , II and III ) .\n\nFurthermore , we performed biological assays in order to evaluate the functional impact of MACC1 SNPs on the motility of colorectal cancer cells .\n\nRESULTS We genotyped three MACC1 SNPs in the coding region .\n\nThirteen % of the tumors had the genotype cg ( rs4721888 , L31V ) , 48% a ct genotype ( rs975263 , S515L ) and 84% a gc or cc genotype ( rs3735615 , R804T ) .\n\nWe found no association of these SNPs with clinicopathological parameters or with patients ' survival , when analyzing the entire patients ' cohort .\n\nAn increased risk for a shorter metastasis-free survival of patients with a ct genotype ( rs975263 ) was observed in younger colon cancer patients with stage I or II ( P = 0.041 , n = 18 ) .\n\nIn cell culture , MACC1 SNPs did not affect MACC1-induced cell motility and proliferation .\n\nCONCLUSION In summary , the identification of coding MACC1 SNPs in primary colorectal tumors does not improve the prediction for metastasis formation or for patients ' survival compared to MACC1 expression analysis alone .\n\nThe ct genotype ( rs975263 ) might be associated with a reduced survival for younger colon cancer patients in early stages .\n\nHowever , further studies with larger sample sizes are needed .", "output": "Activating invasion and metastasis" }, { "input": "Breast cancer includes high number of molecular entities targetable by specific agents .\n\nIn this study , array CGH and PIK3CA/AKT1 mutations were used to drive patients into targeted therapy .\n\nA prospective molecular analysis was offered to metastatic breast cancer patients for whom samples were collected prospectively or retrospectively either from frozen or paraffin-embedded tissue .\n\nAnalyses were performed using array CGH ( Agilent platform ) and PIK3CA ( exon 10 and 21 ) and AKT1 mutations were explored by standard Sanger sequencing .\n\nOne hundred and eight patients were included .\n\nGood quality CGH was obtained in 79% cases and was better for frozen samples .\n\nGenomic alterations were identified in 50% of patients including 11 PIK3CA and 8 AKT1 mutations .\n\nEighteen treatments ( 17 patients ) were administered according to their molecular profile with evidence of activity in nine .\n\nReasons for not providing a genomic-driven treatment included absence of progressive disease ( 38% ) , investigator's choice ( 9% ) , rapid PD ( 19% ) , and no drug access ( 21% ) .\n\nArray CGH correctly identified Her2 status in 97% cases ; failures were related to low % of tumour cells .\n\nOur study showed that array CGH is feasible in the context of daily practice and , in combination with PIK3CA/AKT1 mutations , identifies a significant number of actionable molecular alterations that allow driving patients into specific targeted agents .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND : To quantify tumour angiogenesis , microvessel density ( MVD ) has been widely used .\n\nWe here present a novel angiogenesis marker , microvessel proliferation ( MVP ) , based on dual immunohistochemical staining of nestin and Ki-67 .\n\nImmature endothelial cells express nestin , and when co-expressed with the proliferation marker Ki-67 , the number of proliferating immature blood vessels can be measured .\n\nMATERIALS AND METHODS : Microvessel proliferation was evaluated in 178 breast cancer samples and estimated by vascular proliferation index ( VPI ) , the ratio between the number of vessels containing proliferating endothelial cells and the total number of immature vessels .\n\nRESULTS : High VPI was strongly associated with several markers of aggressive breast cancer , such as negative oestrogen receptor ( ER ) status ( p=0.003 ) , high tumour cell proliferation by Ki-67 ( p=0.004 ) , high p53 expression ( p=0.001 ) , and five profiles for the basal-like phenotype ( odds ratios ( OR ) ; range 3.4-6.3 ) .\n\nAlso , high VPI was significantly associated with interval detected breast cancer compared with screening detected lesions ( p<0.0005 ) , and adverse outcome in univariate and multivariate survival analysis ( p=0.034 and p=0.022 , respectively ) .\n\nCONCLUSION : Microvessel proliferation is a novel marker of ongoing angiogenesis and was associated with aggressive tumour features , basal-like phenotypes , interval presentation , and prognosis in this series of breast cancer .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Liver cancer , predominantly hepatocellular carcinoma ( HCC ) , represents a complex and fatal malignancy driven primarily by oxidative stress and inflammation .\n\nDue to dismal prognosis and limited therapeutic intervention , chemoprevention has emerged as a viable approach to reduce the morbidity and mortality of HCC .\n\nPomegranate fruit is a rich source of phytochemicals endowed with potent antioxidant and anti-inflammatory properties .\n\nWe previously reported that pomegranate phytochemicals inhibit diethylnitrosamine ( DENA)-initiated hepatocarcinogenesis in rats though nuclear factor E2-related factor 2 ( Nrf2)-mediated antioxidant mechanisms .\n\nSince Nrf2 also acts as a key mediator of the nuclear factor-kappaB ( NF-\u03baB)-regulated inflammatory pathway , our present study investigated the anti-inflammatory mechanisms of a pomegranate emulsion ( PE ) during DENA-induced rat hepatocarcinogenesis .\n\nRats were administered with PE ( 1 or 10 g/kg ) 4 weeks before and 18 weeks following DENA initiation .\n\nThere was a significant increase in hepatic expressions of inducible nitric oxide synthase , 3-nitrotyrosine , heat shock protein 70 and 90 , cyclooxygenase-2 and NF-\u03baB in DENA-exposed rat livers .\n\nPE dose-dependently suppressed all aforementioned elevated inflammatory markers .\n\nA conspicuous finding of this study involves lack of cardiotoxicity of PE as assessed by monitoring cardiac function using noninvasive echocardiography .\n\nOur results provide substantial evidence that suppression of the inflammatory cascade through modulation of NF-\u03baB signaling pathway may represent a novel mechanism of liver tumor inhibitory effects of PE against experimental hepatocarcinogenesis .\n\nData presented here coupled with those of our earlier study underline the importance of simultaneously targeting two interconnected molecular circuits , namely , Nrf2-mediated redox signaling and NF-\u03baB-regulated inflammatory pathway , by pomegranate phytoconstituents to achieve chemoprevention of HCC .", "output": "Tumor promoting inflammation" }, { "input": "Bioenergetic profiling of tumors is a new challenge of cancer research and medicine as therapies are currently being developed .\n\nMeanwhile , methodological means must be proposed to gather information on tumor metabolism in order to adapt these potential therapies to the bioenergetic specificities of tumors .\n\nStudies performed on tumors and cancer cell lines have shown that cancer cells bioenergetics is highly variable .\n\nThis profile changes with microenvironmental conditions ( eg. substrate availability ) , the oncogenes activated ( and the tumor suppressors inactivated ) and the interaction with the stroma ( i.e. reverse Warburg effect ) .\n\nHere , we assessed the power of metabolic footprinting ( MFP ) to unravel the bioenergetics and associated anabolic changes induced by three oncogenes , c-Myc , KLF4 and Oct1 .\n\nThe MFP approach provides a quantitative analysis of the metabolites secreted and consumed by cancer cells .\n\nWe used ultra performance liquid chromatography for quantifying the amino acid uptake and secretion .\n\nTo investigate the potential oncogene-mediated alterations in mitochondrial metabolism , we measured oxygen consumption rate and ATP production as well as the glucose uptake and lactate release .\n\nOur findings show that c-Myc deficiency initiates the Warburg effect along with a reduction of mitochondrial respiration .\n\nKLF4 deficiency also stimulated glycolysis , albeit without cellular respiration impairment .\n\nIn contrast , Oct1 deficiency reduced glycolysis and enhanced oxidative phosphorylation efficiency .\n\nMFP revealed that c-Myc , KLF4 and Oct1 altered amino acid metabolism with specific patterns .\n\nWe identified isoleucine , \u03b1-aminoadipic acid and GABA ( \u03b3-aminoisobutyric acid ) as biomarkers related .\n\nOur findings establish the impact of Oct1 , KLF4 and c-Myc on cancer bioenergetics and evidence a link between oncosecretomics and cellular bioenergetics profile .", "output": "Cellular energetics" }, { "input": "In a screen for thoracic malignancy-associated markers , thyroid stimulating hormone receptor ( TSHR ) was identified as a candidate as it binds to the previously-characterized lung cancer marker NKX2-1 .\n\nWe screened for mutations in all coding regions of the TSHR gene in 96 lung adenocarcinoma samples and their matched adjacent normal lung samples .\n\nWe found one patient with a somatic mutation at codon 458 ( exon 10 ) , which is located at the transmembrane domain where most TSHR mutations have been found in thyroid-related diseases .\n\nThis patient had lung adenocarcinoma with BAC ( bronchioloalveolar carcinoma ) features in the setting of a prior medical history significant for carotid stenosis and severe chronic obstructive pulmonary disease ( COPD ) .\n\nIn order to characterize the genetic features of TSHR in lung cancer , we checked for TSHR expression and copy number in the 96 lung cancer tissues .\n\nTSHR protein expression was generally overexpressed in multiple thoracic malignancies ( adenocarcinoma , squamous cell carcinoma and malignant pleural mesothelioma ) by immunohistochemistry .\n\nOur data suggest that aberrant TSHR function may contribute to lung cancer development or a subgroup of lung cancer with specific clinical phenotypes .", "output": "Genomic instability and mutation" }, { "input": "Colorectal tumors are continuously exposed to an inflammatory environment , which together with mitogenic signals sustain several cancer hallmarks .\n\nNuclear factor-kappa B ( NF\\u03baB ) is a major regulator of inflammation and variation in NF\\u03baB-associated genes could potentially be used as biomarkers to identify patients with increased risk of colorectal cancer ( CRC ) development , and/or a rapidly progressing disease .\n\nIn this study , 348 CRC cases and 806 randomly selected healthy individuals from southeastern Sweden were examined with regard to seven polymorphisms in NF\\u03baB pathway-associated genes .\n\nLog-rank-tests and Cox proportional hazard regression analysis examined the association between the polymorphisms and CRC-specific survival , whereas chi-square tests and logistic regression analysis were used to test for associations between the polymorphisms and CRC susceptibility .\n\nGene expression and loss of heterozygosity analyses of TNFAIP3 were carried out in a subset of tumors to assess its role as a tumor suppressor in CRC .\n\nHeterozygous and polymorphic TNFAIP3 ( rs6920220 ) , heterozygous NLRP3 ( Q705K ) and polymorphic NF\\u03baB -94 ATTG ins/del genotypes were found to be associated with poorer survival in patients diagnosed with invasive CRC ( aHR = 5.2 , 95% CI : 2.5-10.9 , P < 0.001 ) .\n\nTNFAIP3 mRNA levels were significantly decreased in tumors compared with adjacent non-neoplastic mucosa ( P < 0.0001 ) and loss of heterozygosity of 6q23.3 ( TNFAIP3 ) was detected in 17% of cases , whereas only 2.5% of the investigated specimens displayed TNFAIP3 gene mutations .\n\nWe propose that TNFAIP3 ( rs6920220 ) , NLRP3 ( Q705K ) and NF\\u03baB -94 ATTG ins/del polymorphisms are associated with poor survival in patients with advanced CRC and may be used as prognostic markers .\n\nExperimental results indicate that TNFAIP3 may act as a tumor suppressor in CRC .", "output": "Genomic instability and mutation" }, { "input": "Interleukin-23 ( IL-23 ) plays an essential role in the mucosal immune system .\n\nIt has been suggested that IL-23 is able to induce carcinogenesis as well as inflammation and a recent study revealed that IL-23R is expressed in colorectal carcinoma cells .\n\nHowever , neither the differences in the IL-23R expression among the patients nor the concrete functions of IL-23 in colorectal carcinoma cells have been revealed .\n\nThe aim of the present study was to examine the characteristics of IL-23R expression in colorectal carcinoma and the direct effects of IL-23 on colorectal cancer cells .\n\nWe examined the IL-23R expression in human colorectal cancer tissue samples by immunohistochemistry .\n\nCell proliferation and invasion assays under IL-23 stimulation were performed using cultured cells derived from colorectal cancer .\n\nELISA and real-time PCR were used to evaluate the transforming growth factor ( TGF)-\u03b2 production due to IL-23 stimulation .\n\nAll of the TNM stage IV patients were positive for IL-23R .\n\nIL-23R expression in the carcinoma tissue was also relatively high at the deepest point of invasion in certain cases .\n\nThe proliferative and invasive activities and/or TGF-\u03b2 production of DLD-1 cells increased by IL-23 stimulation , whereas no change was observed in the activities of MIP101 and KM12c cells .\n\nIL-23 directly enhanced the malignancy of the colon carcinoma cells .\n\nAn autocrine mechanism via TGF-\u03b2 production may underlie these effects .\n\nIL-23 is therefore a potential target for cancer immunotherapy .\n\nHowever , the homogeneity in IL-23R expression and the effects of IL-23 on colorectal carcinoma cells should be considered .", "output": "Activating invasion and metastasis" }, { "input": "Proton radiotherapy has gained more favor among oncologists as a treatment option for localized and deep-seated tumors .\n\nIn addition , protons are a major constituent of the space radiation astronauts receive during space flights .\n\nThe potential for these exposures to lead to , or enhance cancer risk has not been well studied .\n\nOur objective is to study the biological effects of low energy protons on epithelial cells and its propensity to enhance transforming growth factor beta 1 ( TGF\u03b21)-mediated epithelial-mesenchymal transition ( EMT ) , a process occurring during tumor progression and critical for invasion and metastasis .\n\nNon-transformed mink lung epithelial cells ( Mv1Lu ) and hTERT- immortalized human esophageal epithelial cells ( EPC ) were used in this study .\n\nEMT was identified by alterations in cell morphology , EMT-related gene expression changes determined using real-time PCR , and EMT changes in specific cellular markers detected by immunostaining and western blotting .\n\nAlthough TGF\u03b21 treatment alone is able to induce EMT in both Mv1Lu and EPC cells , low energy protons ( 5 MeV ) at doses as low as 0.1 Gy can enhance TGF\u03b21 induced EMT .\n\nProtons alone can also induce a mild induction of EMT .\n\nSD208 , a potent TGF\u03b2 Receptor 1 ( TGF\u03b2R1 ) kinase inhibitor , can efficiently block TGF\u03b21/Smad signaling and attenuate EMT induction .\n\nWe suggest a model for EMT after proton irradiation in normal and cancerous tissue based on our results that showed that low and high doses of protons can sensitize normal human epithelial cells to mesenchymal transition , more prominently in the presence of TGF\u03b21 , but also in the absence of TGF\u03b21 .", "output": "Activating invasion and metastasis" }, { "input": "BACKGROUND To investigate the dynamics of inter- and intratumoral molecular alterations during tumor progression in recurrent gliomas .\n\nMETHODOLOGY/PRINCIPAL FINDINGS To address intertumoral heterogeneity we investigated non-microdissected tumor tissue of 106 gliomas representing 51 recurrent tumors .\n\nTo address intratumoral heterogeneity a set of 16 gliomas representing 7 tumor pairs with at least one recurrence , and 4 single mixed gliomas were investigated by microdissection of distinct oligodendroglial and astrocytic tumor components .\n\nAll tumors and tumor components were analyzed for allelic loss of 1p/19q ( LOH 1p/19q ) , for TP53- mutations and for R132 mutations in the IDH1 gene .\n\nThe investigation of non-microdissected tumor tissue revealed clonality in 75% ( 38/51 ) .\n\nAberrant molecular alterations upon recurrence were detected in 25% ( 13/51). 64% ( 9/14 ) of these were novel and associated with tumor progression .\n\nLoss of previously detected alterations was observed in 36% ( 5/14 ) .\n\nOne tumor pair ( 1/14 ; 7% ) was significant for both .\n\nIntratumoral clonality was detected in 57% ( 4/7 ) of the microdissected tumor pairs and in 75% ( 3/4 ) of single microdissected tumors. 43% ( 3/7 ) of tumor pairs and one single tumor ( 25% ) revealed intratumoral heterogeneity .\n\nWhile intratumoral heterogeneity affected both the TP53- mutational status and the LOH1p/19q status , all tumors with intratumoral heterogeneity shared the R132 IDH1- mutation as a common feature in both their microdissected components .\n\nCONCLUSIONS/SIGNIFICANCE The majority of recurrent gliomas are of monoclonal origin .\n\nHowever , the detection of divertive tumor cell clones in morphological distinct tumor components sharing IDH1- mutations as early event may provide insight into the tumorigenesis of true mixed gliomas .", "output": "Genomic instability and mutation" }, { "input": "The tumor suppressor gene p53 has been implicated in the regulation of epithelial-mesenchymal transition ( EMT ) and tumor metastasis by regulating microRNA ( miRNA ) expression .\n\nHere , we report that mutant p53 exerts oncogenic functions and promotes EMT in endometrial cancer ( EC ) by directly binding to the promoter of miR-130b ( a negative regulator of ZEB1 ) and inhibiting its transcription .\n\nWe transduced p53 mutants into p53-null EC cells , profiled the miRNA expression by miRNA microarray and identified miR-130b as a potential target of mutant p53 .\n\nEctopic expression of p53 mutants repressed the expression of miR-130b and triggered ZEB1-dependent EMT and cancer cell invasion .\n\nLoss of an endogenous p53 mutation increased the expression of miR-130b , which resulted in reduced ZEB1 expression and attenuation of the EMT phenotype .\n\nFurthermore , re-expression of miR-130b suppressed mutant p53-induced EMT and ZEB1 expression .\n\nImportantly , the expression of miR-130 was significantly reduced in EC tissues , and patients with higher expression levels of miR-130b survived longer .\n\nThese data provide a novel understanding of the roles of p53 gain-of-function mutations in accelerating tumor progression and metastasis through modulation of the miR-130b-ZEB1 axis .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "BACKGROUND Agricultural products and by products provide the primary materials for a variety of technological applications in diverse industrial sectors .\n\nAgro-industrial wastes , such as cotton and curaua fibers , are used to prepare nanofibers for use in thermoplastic films , where they are combined with polymeric matrices , and in biomedical applications such as tissue engineering , amongst other applications .\n\nThe development of products containing nanofibers offers a promising alternative for the use of agricultural products , adding value to the chains of production .\n\nHowever , the emergence of new nanotechnological products demands that their risks to human health and the environment be evaluated .\n\nThis has resulted in the creation of the new area of nanotoxicology , which addresses the toxicological aspects of these materials .\n\nPURPOSE AND METHODS Contributing to these developments , the present work involved a genotoxicological study of different nanofibers , employing chromosomal aberration and comet assays , as well as cytogenetic and molecular analyses , to obtain preliminary information concerning nanofiber safety .\n\nThe methodology consisted of exposure of Allium cepa roots , and animal cell cultures ( lymphocytes and fibroblasts ) , to different types of nanofibers .\n\nNegative controls , without nanofibers present in the medium , were used for comparison .\n\nRESULTS The nanofibers induced different responses according to the cell type used .\n\nIn plant cells , the most genotoxic nanofibers were those derived from green , white , and brown cotton , and curaua , while genotoxicity in animal cells was observed using nanofibers from brown cotton and curaua .\n\nAn important finding was that ruby cotton nanofibers did not cause any significant DNA breaks in the cell types employed .\n\nCONCLUSION This work demonstrates the feasibility of determining the genotoxic potential of nanofibers derived from plant cellulose to obtain information vital both for the future usage of these materials in agribusiness and for an understanding of their environmental impacts .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Flat adenomas are a subgroup of colorectal adenomas that have been associated with a distinct biology and a more aggressive clinical behavior compared to their polypoid counterparts .\n\nIn the present study , we aimed to compare the mutation spectrum of 14 cancer genes , between these two phenotypes .\n\nMETHODS A consecutive series of 106 flat and 93 polypoid adenomas was analyzed retrospectively for frequently occurring mutations in \" hot spot \" regions of KRAS , BRAF , PIK3CA and NRAS , as well as selected mutations in CTNNB1 ( \\u03b2-catenin ) , EGFR , FBXW7 ( CDC4 ) , PTEN , STK11 , MAP2K4 , SMAD4 , PIK3R1 and PDGFRA using a high-throughput genotyping technique .\n\nAdditionally , APC was analyzed using direct sequencing .\n\nRESULTS APC mutations were more frequent in polypoid adenomas compared to flat adenomas ( 48.5% versus 30.3% , respectively , p\\u200a=\\u200a0.02 ) .\n\nMutations in KRAS , BRAF , NRAS , FBXW7 and CTNNB1 showed similar frequencies in both phenotypes .\n\nBetween the different subtypes of flat adenomas ( 0-IIa , LST-F and LST-G ) no differences were observed for any of the investigated genes .\n\nCONCLUSION The lower APC mutation rate in flat adenomas compared to polypoid adenomas suggests that disruption of the Wnt-pathway may occur via different mechanisms in these two phenotypes .\n\nFurthermore , in contrast to previous observations our results in this large well-defined sample set indicate that there is no significant association between the different morphological phenotypes and mutations in key genes of the RAS-RAF-MAPK pathway .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Epstein-Barr virus ( EBV ) encodes six nuclear transformation-associated proteins that induce extensive changes in cellular gene expression and signaling and induce B-cell transformation .\n\nThe role of HIF1A in EBV-induced B-cell immortalization has not been previously studied .\n\nMETHODS AND FINDINGS Using Western blotting and Q-PCR , we found that HIF1A protein is stabilized in EBV-transformed lymphoblastoid cells .\n\nWestern blotting , GST pulldown assays , and immunoprecipitation showed that EBV-encoded nuclear antigens EBNA-5 and EBNA-3 bind to prolylhydroxylases 1 and 2 , respectively , thus inhibiting HIF1A hydroxylation and degradation .\n\nImmunostaining and Q-PCR showed that the stabilized HIF1A translocates to the nucleus , forms a heterodimer with ARNT , and transactivates several genes involved in aerobic glycolysis .\n\nUsing biochemical assays and Q-PCR , we also found that lymphoblastoid cells produce high levels of lactate , lactate dehydrogenase and pyruvate .\n\nCONCLUSIONS Our data suggest that activation of the aerobic glycolytic pathway , corresponding to the Warburg effect , occurs in EBV-transformed lymphoblastoid cells , in contrast to mitogen-activated B-cells .", "output": "Cellular energetics" }, { "input": "22Rv1 is a common prostate cancer cell line used in xenograft mouse experiments as well as in vitro cell culture assays to study aspects of prostate cancer tumorigenesis .\n\nRecently , this cell line was shown to harbor multiple copies of a gammaretrovirus , called XMRV , integrated in its genome .\n\nWhile the original prostate cancer xenograft CWR22 is free of any retrovirus , subsequently generated cell lines 22Rv1 and CWR-R1 , carry this virus and additionally shed infectious gammaretroviral particles in their supernatant .\n\nAlthough XMRV most likely was generated by recombination events in cell culture this virus has been demonstrated to infect human cells in vitro and 22Rv1 as well as CWR-R1 cells are now considered biosafety 2 reagents .\n\nHere , we demonstrate that 22Rv1 cells with reduced retroviral transcription show reduced tumor angiogenesis and increased necrosis of the primary tumor derived from xenografted cells in scid mice when compared to the parental cell line .\n\nThe presence of XMRV transcripts significantly increases secretion of osteopontin ( OPN ) , CXCL14 , IL13 and TIMP2 in 22Rv1 cells .\n\nFurthermore , these data are supported by in vitro cell invasion and differentiation assays .\n\nCollectively , our data suggest that the presence of XMRV transcripts at least partially contributes to 22Rv1 characteristics observed in vitro and in vivo with regard to migration , invasion and tumor angiogenesis .\n\nWe propose that data received with 22Rv1 cells or equivalent cells carrying xenotropic gammaretroviruses should be carefully controlled including other prostate cancer cell lines tested for viral sequences .", "output": "Activating invasion and metastasis, Inducing angiogenesis, Resisting cell death" }, { "input": "Metastatic cancer is extremely difficult to treat , and the presence of metastases greatly reduces a cancer patient's likelihood of long-term survival .\n\nThe ZEB1 transcriptional repressor promotes metastasis through downregulation of microRNAs ( miRs ) that are strong inducers of epithelial differentiation and inhibitors of stem cell factors .\n\nGiven that each miR can target multiple genes with diverse functions , we posited that the prometastatic network controlled by ZEB1 extends beyond these processes .\n\nWe tested this hypothesis using a mouse model of human lung adenocarcinoma metastasis driven by ZEB1 , human lung carcinoma cells , and human breast carcinoma cells .\n\nTranscriptional profiling studies revealed that ZEB1 controls the expression of numerous oncogenic and tumor-suppressive miRs , including miR-34a .\n\nEctopic expression of miR-34a decreased tumor cell invasion and metastasis , inhibited the formation of promigratory cytoskeletal structures , suppressed activation of the RHO GTPase family , and regulated a gene expression signature enriched in cytoskeletal functions and predictive of outcome in human lung adenocarcinomas .\n\nWe identified several miR-34a target genes , including Arhgap1 , which encodes a RHO GTPase activating protein that was required for tumor cell invasion .\n\nThese findings demonstrate that ZEB1 drives prometastatic actin cytoskeletal remodeling by downregulating miR-34a expression and provide a compelling rationale to develop miR-34a as a therapeutic agent in lung cancer patients .", "output": "Activating invasion and metastasis" }, { "input": "Identifying genomic alterations driving breast cancer is complicated by tumor diversity and genetic heterogeneity .\n\nRelevant mouse models are powerful for untangling this problem because such heterogeneity can be controlled .\n\nInbred Chaos3 mice exhibit high levels of genomic instability leading to mammary tumors that have tumor gene expression profiles closely resembling mature human mammary luminal cell signatures .\n\nWe genomically characterized mammary adenocarcinomas from these mice to identify cancer-causing genomic events that overlap common alterations in human breast cancer .\n\nChaos3 tumors underwent recurrent copy number alterations ( CNAs ) , particularly deletion of the RAS inhibitor Neurofibromin 1 ( Nf1 ) in nearly all cases .\n\nThese overlap with human CNAs including NF1 , which is deleted or mutated in 27.7% of all breast carcinomas .\n\nChaos3 mammary tumor cells exhibit RAS hyperactivation and increased sensitivity to RAS pathway inhibitors .\n\nThese results indicate that spontaneous NF1 loss can drive breast cancer .\n\nThis should be informative for treatment of the significant fraction of patients whose tumors bear NF1 mutations .", "output": "Genomic instability and mutation" }, { "input": "The metastasis-associated lung adenocarcinoma transcript 1 , MALAT1 , is a long non-coding RNA ( lncRNA ) that has been discovered as a marker for lung cancer metastasis .\n\nIt is highly abundant , its expression is strongly regulated in many tumor entities including lung adenocarcinoma and hepatocellular carcinoma as well as physiological processes , and it is associated with many RNA binding proteins and highly conserved throughout evolution .\n\nThe nuclear transcript MALAT-1 has been functionally associated with gene regulation and alternative splicing and its regulation has been shown to impact proliferation , apoptosis , migration and invasion .\n\nHere , we have developed a human and a mouse knockout system to study the loss-of-function phenotypes of this important ncRNA .\n\nIn human tumor cells , MALAT1 expression was abrogated using Zinc Finger Nucleases .\n\nUnexpectedly , the quantitative loss of MALAT1 did neither affect proliferation nor cell cycle progression nor nuclear architecture in human lung or liver cancer cells .\n\nMoreover , genetic loss of Malat1 in a knockout mouse model did not give rise to any obvious phenotype or histological abnormalities in Malat1-null compared with wild-type animals .\n\nThus , loss of the abundant nuclear long ncRNA MALAT1 is compatible with cell viability and normal development .", "output": "Sustaining proliferative signaling" }, { "input": "FMS-like tyrosine kinase 3 ( FLT3 ) normally functions in the survival/proliferation of hematopoietic stem/progenitor cells , but its constitutive activation by internal tandem duplication ( ITD ) mutations correlates with a poor prognosis in AML .\n\nThe development of FLT3 tyrosine kinase inhibitors ( TKI ) is a promising strategy , but resistance that arises during the course of treatment caused by secondary mutations within the mutated gene itself poses a significant challenge .\n\nIn an effort to predict FLT3 resistance mutations that might develop in patients , we used saturation mutagenesis of FLT3/ITD followed by selection of transfected cells in FLT3 TKI .\n\nWe identified F621L , A627P , F691L and Y842C mutations in FLT3/ITD that confer varying levels of resistance to FLT3 TKI .\n\nWestern blotting confirmed that some FLT3 TKI were ineffective at inhibiting FLT3 autophosphorylation and signaling through MAP kinase , STAT5 and AKT in some mutants .\n\nBalb/c mice transplanted with the FLT3/ITD Y842C mutation confirmed resistance to sorafenib in vivo but not to lestaurtinib .\n\nThese results indicate a growing number of FLT3 mutations that are likely to be encountered in patients .\n\nSuch knowledge , combined with known remaining sensitivity to other FLT3 TKI , will be important to establish as secondary drug treatments that can be substituted when these mutants are encountered .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Increasing evidence shows that estrogens are involved in lung cancer proliferation and progression , and most human lung tumors express estrogen receptor \u03b2 ( ER\u03b2 ) as well as aromatase .\n\nTo determine if the aromatase inhibitor anastrozole prevents development of lung tumors induced by a tobacco carcinogen , alone or in combination with the ER antagonist fulvestrant , ovariectomized female mice received treatments with the tobacco carcinogen 4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanone ( NNK ) along with daily supplements of androstenedione , the substrate for aromatase .\n\nPlacebo , anastrozole and/or fulvestrant were administered in both an initiation and a promotion protocol of lung tumorigenesis .\n\nThe combination of fulvestrant and anastrozole given during NNK exposure resulted in significantly fewer NNK-induced lung tumors ( mean = 0.5 ) compared with placebo ( mean = 4.6 , P < 0.001 ) , fulvestrant alone ( mean = 3.4 , P < 0.001 ) or anastrozole alone ( mean = 2.8 , P = 0.002 ) .\n\nA significantly lower Ki67 cell proliferation index was also observed compared with single agent and control treatment groups .\n\nBeginning antiestrogen treatment after NNK exposure , when preneoplastic lesions had already formed , also yielded maximum antitumor effects with the combination .\n\nAromatase expression was found mainly in macrophages infiltrating preneoplastic and tumorous areas of the lungs , whereas ER\u03b2 was found in both macrophages and tumor cells .\n\nAntiestrogens , especially in combination , effectively inhibited tobacco carcinogen-induced murine lung tumorigenesis and may have application for lung cancer prevention .\n\nAn important source of estrogen synthesis may be inflammatory cells that infiltrate the lungs in response to carcinogens , beginning early in the carcinogenesis process .\n\nER\u03b2 expressed by inflammatory and neoplastic epithelial cells in the lung may signal in response to local estrogen production .", "output": "Sustaining proliferative signaling" }, { "input": "A glycolytic profile unifies a group of pheochromocytomas and paragangliomas ( PHEOs/PGLs ) with distinct underlying gene defects , including von Hippel-Lindau ( VHL ) and succinate dehydrogenase B ( SDHB ) mutations .\n\nNevertheless , their tumor aggressiveness is distinct : PHEOs/PGLs metastasize rarely in VHL- , but frequently in SDHB-patients .\n\nTo date , the molecular mechanisms causing the more aggressive phenotype in SDHB-PHEOs/PGLs remain largely unknown .\n\nRecently , however , an excellent model to study aggressive PHEOs ( mouse tumor tissue ( MTT ) cells ) has been developed from mouse PHEO cells ( MPC ) .\n\nWe employed this model for a proteomics based approach to identify changes characteristic for tumor aggressiveness , which we then explored in a homogeneous set of human SDHB- and VHL-PHEOs/PGLs .\n\nThe increase of glucose transporter 1 in VHL , and of hexokinase 2 in VHL and SDHB , confirmed their glycolytic profile .\n\nIn agreement with the cell model and in support of decoupling of glycolysis , the Krebs cycle and oxidative phosphorylation ( OXPHOS ) , SDHB tumors showed increased lactate dehydrogenase levels .\n\nIn SDHB-PGLs OXPHOS complex activity was increased at complex III and , as expected , decreased at complex II .\n\nMoreover , protein and mRNA expression of all tested OXPHOS-related genes were higher in SDHB- than in VHL-derived tumors .\n\nAlthough there was no direct evidence for increased reactive oxygen species production , elevated superoxide dismutase 2 expression may reflect elevated oxidative stress in SDHB-derived PHEOs/PGLs .\n\nFor the first time , we show that despite dysfunction in complex II and evidence for a glycolytic phenotype , the Warburg effect does not seem to fully apply to SDHB-PHEOs/PGLs with respect to decreased OXPHOS .\n\nIn addition , we present evidence for increased LDHA and SOD2 expression in SDHB-PHEOs/PGLs , proteins that have been proposed as promising therapeutic targets in other cancers .\n\nThis study provides new insight into pathogenic mechanisms in aggressive human PHEOs/PGLs , which may lead to identifying new diagnostic and prognostic markers in the near future .", "output": "Tumor promoting inflammation, Cellular energetics" }, { "input": "BACKGROUND To assess the potential mechanisms that may underlie increased local failure in triple negative ( TN ) breast cancers , an analysis was performed of the risk of residual carcinoma after lumpectomy with correlation to pathologic factors , including molecular phenotype .\n\nMETHODS A review of pathologic specimens was performed for women with invasive breast cancer treated with lumpectomy followed by reexcision .\n\nData were collected on age ; tumor size , grade , and nodal stage ; estrogen receptor , progesterone receptor , and human endothelial growth factor receptor 2 ( Her2 ) ; extensive intraductal component ; lymphovascular invasion ; margins ; and reexcision findings .\n\nUnivariate and multivariate logistic regression analyses were performed to evaluate for associations between pathologic features of the lumpectomy specimen and reexcision findings .\n\nMolecular phenotypes were defined by conventionally used immunohistochemical pattern .\n\nRESULTS Data were collected on 369 patients with breast cancer .\n\nThe median age was 57 years , median tumor size was 1.5 cm , 36% had positive margins , 32% had positive lymph nodes , 73.5% had the luminal A subtype , 9.5% had the luminal B subtype , 4.5% were Her2-enriched , and 12.5% were TN .\n\nOverall , 32% of patients had invasive cancer in their reexcision specimens , and 51% of those with the TN subtype had residual invasive disease on reexcision compared with 30% to 31% for other subtypes .\n\nOn univariate analysis , age , tumor size , margin status , lymphovascular invasion , nodal status , and TN subtype were associated with elevated risk of residual invasive cancer .\n\nOn multivariate analysis using a forward stepwise model , TN subtype maintained significance , with an odds ratio of 3.28 ( P = .002 ) .\n\nCONCLUSION TN subtype has a statistically significant association with an increased risk of residual tumor .\n\nThis suggests the putative increase in the risk of local failure in TN patients may be related to increased residual tumor burden .", "output": "Activating invasion and metastasis" }, { "input": "The role of energy deregulation and altered/adapted metabolism in tumor cells is an increasingly important issue in understanding cancer .\n\nHereditary leiomyomatosis and renal cell carcinoma ( HLRCC ) is an aggressive form of RCC characterized by germline mutation of fumarate hydratase ( FH ) , followed by somatic loss of the remaining wild-type allele and known to be a highly metastatic and lethal malignancy compared to other RCCs .\n\nThe intrinsic loss of normal tricarboxylic acid ( TCA ) cycle presumably aids tumorigenesis due to the necessary metabolic alterations required and the enforced dependence on glycolysis derived energy , mimicking the Warburg effect .\n\nThus , there is considerable utility in establishing a preclinical cell model from these tumors to study energy metabolism deregulation , as well as developing new targeted therapeutic approaches for TCA cycle enzyme-deficient cancers .\n\nHere , we describe a new immortalized cell line , UOK268 , derived from a patient's primary HLRCC-associated kidney cancer .\n\nThis represents the first primary renal cell line to model TCA cycle gene loss and provides a perfect partner cell line to our previously described metastasis-derived HLRCC-associated cell line , UOK262 .\n\nWe identified a novel germline FH missense mutation , p.His192Asp , and the subsequent loss of heterozygosity in UOK268 .\n\nThe UOK268 cell line expressed mutant FH protein , which localized to the mitochondria , but with loss of almost all catalytic activity .\n\nThe UOK268 cells had severely compromised oxidative phosphorylation and increased glycolytic flux .\n\nIngenuity pathways analysis of human mitochondria-focused cDNA microarray ( hMitChip3 ) gene chip data confirmed the altered mRNA expression patterns of genes involved in several important pathways , such as lipid metabolism , apoptosis , and energy production/glycolysis .\n\nUOK268 provides a unique model of a primary cell line demonstrating an enforced , irreversible Warburg effect and , combined with UOK262 , provides a unique invitro preclinical model for studying the bioenergetics of the Warburg effect in human cancer .", "output": "Genomic instability and mutation, Cellular energetics, Resisting cell death" }, { "input": "Overexpression of growth factors and/or their receptors is a common event in malignancy and provides the underlying mechanisms for one of the hallmarks of cancer , uncontrolled proliferation .\n\nMounting evidence suggests that IGF-1 is involved in the pathogenesis and progression of different types of human cancer such as colon , breast , prostate and lung .\n\nHowever , only a few studies have investigated the association between IGF-1 levels and childhood cancer risk .\n\nWe aimed to compare the IGF-1 serum level in children with de novo malignancies to healthy children , and to assess its relationship with cancer type , stage , metastasis and different disease characteristics .\n\nThe study was carried out on 100 children ; 50 children with de novo malignancies and 50 healthy children of matched age and gender as a control group .\n\nThe patients were subjected to a routine work-up for their cancers according to our local standards .\n\nEstimation of the serum level of IGF-1 was carried out in the two groups using ELISA .\n\nOur results showed that children with cancer had significantly higher levels of IGF-1 than healthy controls of the same age and gender .\n\nNo association was found between IGF-1 and tumor type , stage , metastasis and other disease characteristics .\n\nIn conclusion , the IGF-1 serum level is an important indicator of risk for the most prevalent forms of childhood cancer .\n\nIt may be used to identify children at the highest risk for these cancers and aid in determing who may benefit most from preventive strategies .\n\nGiven the small number of children in our study , studies with larger populations are required to confirm these results .", "output": "Sustaining proliferative signaling" }, { "input": "OBJECTIVES To investigate the expression of TLR-4 ( toll-like receptor ) on human cervical cancer and find the biological function of the TLR-4 signal system .\n\nMETHODS The immunohistochemistry method was performed to study the protein expression and distribution of TLR-4 .\n\nThe viability of HeLa cells was determined by cell viability assay .\n\nCell proliferation was detected by FCM , ELISA and Western blot were used to observe the gene and protein expression of IL-6 and TGF-beta1 in Hela cell lines .\n\nRESULTS TLR-4 was over-expressed in cervix cancer , and its activation by LPS promotes proliferation and anti-apoptosis in Hela cells in vitro .\n\nMoreover the cell line proliferation increased in a dose- and time-dependent manner .\n\nThe production of IL-6 and TGF-beta1 were promoted through the activation of the NF-kappaB signaling pathway .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "PURPOSE To characterize the clinical features of a Chinese Uygur pedigree with primary open-angle glaucoma ( POAG ) and to identify mutations in two candidate genes , trabecular meshwork inducible glucocorticoid response ( MYOC/TIGR ) and human dioxin-inducible cytochrome P450 ( CYP1B1 ) .\n\nMETHODS Twenty one members from a Chinese Uygur family of four generations were included in the study .\n\nAll participants underwent complete ophthalmologic examinations .\n\nFive were diagnosed as POAG , four as glaucoma suspects , and the rest were asymptomatic .\n\nMolecular genetic analysis was performed on all subjects included in the study .\n\nAll exons of CYP1B1 and MYOC were amplified by polymerase chain reaction ( PCR ) , sequenced and compared with a reference database .\n\nThe variations detected were evaluated in available family members as well as 102 normal controls .\n\nPossible changes in structure and function of the protein induced by amino acid variance were predicted by bioinformatics analysis .\n\nRESULTS Elevated intraocular pressure and late-stage glaucomatous cupping of the optic disc were found in five patients of this family .\n\nA novel heterozygous missense mutation c.1151 A>G in exon 3 of MYOC was found in all five patients diagnosed as POAG and four glaucoma suspects , but not in the rest of the family members and 102 normal controls .\n\nThis mutation caused an amino acid substitution of aspartic acid to glycine at position 384 ( p .\n\nD384G ) of the MYOC protein .\n\nThis substitution may cause structural and functional changes of the protein based on bioinformatics analysis .\n\nNo mutations were found in CYP1B1 .\n\nCONCLUSIONS Our study suggests that the novel mutation D384G of MYOC is likely responsible for the pathogenesis of POAG in this pedigree .", "output": "Genomic instability and mutation" }, { "input": "Focal adhesion kinase ( FAK ) is an important mediator of extracellular matrix integrin signaling , cell motility , cell proliferation and cell survival .\n\nIncreased FAK expression is observed in a variety of solid human tumors and increased FAK expression and activity frequently correlate with metastatic disease and poor prognosis .\n\nHerein we identify miR-7 as a direct regulator of FAK expression. miR-7 expression is decreased in malignant versus normal breast tissue and its expression correlates inversely with metastasis in human breast cancer patients .\n\nForced expression of miR-7 produced increased E-CADHERIN and decreased FIBRONECTIN and VIMENTIN expression in breast cancer cells .\n\nThe levels of miR-7 expression was positively correlated with E-CADHERIN mRNA and negatively correlated with VIMENTIN mRNA levels in breast cancer samples .\n\nForced expression of miR-7 in aggressive breast cancer cell lines suppressed tumor cell monolayer proliferation , anchorage independent growth , three-dimensional growth in Matrigel , migration and invasion .\n\nConversely , inhibition of miR-7 in the HBL-100 mammary epithelial cell line promoted cell proliferation and anchorage independent growth .\n\nRescue of FAK expression reversed miR-7 suppression of migration and invasion. miR-7 also inhibited primary breast tumor development , local invasion and metastatic colonization of breast cancer xenografts .\n\nThus , miR-7 expression is decreased in metastatic breast cancer , correlates with the level of epithelial differentiation of the tumor and inhibits metastatic progression .", "output": "Activating invasion and metastasis" }, { "input": "Both the deregulation of microRNAs and epidermal growth factor receptor ( EGFR ) are emerging as important factors in non-small-cell lung cancer ( NSCLC ) .\n\nHere , miR-133b was found to be associated with tumor stage , the extent of regional lymph node involvement , stage , visceral pleura or vessel invasion and EGFR mRNA expression in Chinese patients with NSCLC .\n\nBioinformatic analysis and luciferase reporter assay revealed that miR-133b can interact specifically with the 3'-UTR of EGFR mRNA .\n\nFunctionally , miR-133b transfection showed regulatory activity in translationally repressing EGFR mRNA .\n\nMoreover , miR-133b transfection may modulate apoptosis , invasion and sensitivity to EGFR-TKI through the EGFR signaling pathways , especially in EGFR-addicted NSCLC cells .\n\nTaken together , our findings show that miR-133b can inhibit cell growth of NSCLC through targeting EGFR and regulating its downstream signaling pathway .\n\nThis finding has important implications for the development of targeted therapeutics for a number of EGFR-addicted cancers .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "Stress-induced phosphoprotein 1 ( STIP1 ) , a cochaperone that organizes other chaperones , heat shock proteins ( HSPs ) , was recently shown to be secreted by human ovarian cancer cells .\n\nIn neuronal tissues , binding to prion protein was required for STIP1 to activate the ERK ( extracellular-regulated MAP kinase ) signaling pathways .\n\nHowever , we report that STIP1 binding to a bone morphogenetic protein ( BMP ) receptor , ALK2 ( activin A receptor , type II-like kinase 2 ) , was necessary and sufficient to stimulate proliferation of ovarian cancer cells .\n\nThe binding of STIP1 to ALK2 activated the SMAD signaling pathway , leading to transcriptional activation of ID3 ( inhibitor of DNA binding 3 ) , promoting cell proliferation .\n\nIn conclusion , ovarian-cancer-tissue-secreted STIP1 stimulates cancer cell proliferation by binding to ALK2 and activating the SMAD-ID3 signaling pathways .\n\nAlthough animal studies are needed to confirm these mechanisms in vivo , our results may pave the way for developing novel therapeutic strategies for ovarian cancer .", "output": "Sustaining proliferative signaling" }, { "input": "Objective .\n\nThe present study was performed to investigate the effect of N-desulfated heparin on basic fibroblast growth factor ( bFGF ) expression , tumor angiogenesis and metastasis of gastric carcinoma .\n\nMethods .\n\nHuman gastric cancer SGC-7901 tissues were orthotopically implanted into the stomach of NOD SCID mice .\n\nTwenty mice were randomly divided into two groups which received either intravenous injection of 0.9% NaCl solution ( normal saline group ) or 10\u2009mg/kg N-desulfated heparin ( N-desulfated heparin group ) twice weekly for three weeks .\n\nIn vitro , human gastric carcinoma SGC-7901 cells were treated with N-desulfated heparin in different concentration ( 0.1\u2009mg/mL , 1\u2009mg/mL , N-desulfated heparin group ) , and treated with medium ( control group ) .\n\nResults .\n\nIn vivo , the tumor metastasis rates were 9/10 in normal saline group and 2/10 in N-desulfated heparin group ( P < 0.05 ) .\n\nThe intratumoral microvessel density was higher in normal saline group than in N-desulfated heparin group ( P < 0.05). bFGF expression in gastric tissue was inhibited by N-desulfated heparin ( P < 0.05 ) .\n\nThere was no bleeding in N-desulfated heparin group .\n\nIn vitro , N-desulfated heparin inhibited significantly bFGF protein and mRNA expression of gastric carcinoma cells ( P < 0.05 ) .\n\nConclusions .\n\nN-desulfated heparin can inhibit the metastasis of gastric cancer through inhibiting tumor bFGF expression and tumor angiogenesis with no obvious anticoagulant activity .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "OBJECTIVES Lymph node metastasis is among the most important prognostic factors for patients with esophageal squamous cell carcinoma after curative esophagectomy ; however , the extent of lymphadenectomy is still controversial .\n\nThe objective of the present study was to determine the frequency of lymphatic metastases and to study the pattern of lymph node metastasis in a large study population .\n\nMETHODS The data from 1361 patients with thoracic esophageal squamous cell carcinoma who underwent curative R0 esophagectomy were retrospectively examined .\n\nLogistic regression analysis was used to identify the factors associated with lymph node metastasis .\n\nRESULTS Of the 1361 patients , 714 ( 52.5% ) were found to have lymph node metastasis .\n\nThe frequency of lymph node metastasis increased as the tumor invasion increased .\n\nParatracheal nodes were the most frequent metastasis nodes ( 15.9% ) .\n\nThe frequency of lymph node metastasis was 9.8% in the neck , 18.0% in the upper mediastinum , 18.9% in the middle mediastinum , 11.8% in the lower mediastinum , and 28.4% in the abdomen .\n\nOf these 714 patients , 424 ( 31.2% ) presented with 1 field involvement , 255 ( 18.7% ) with 2 fields , and 35 ( 2.6% ) with 3 fields involvement .\n\nLogistic regression analysis revealed tumor length ( P<.001 ) , tumor invasion ( P<.001 ) , tumor differentiation ( P=.003 ) , and lymphovascular invasion ( P<.001 ) were risk factors for lymph node metastasis .\n\nTumor location ( P<.001 ) , tumor invasion ( P=.003 ) , lymphovascular invasion ( P=.004 ) , and paratracheal lymph node involvement ( P=.002 ) were identified as risk factors for cervical lymph node metastasis .\n\nCONCLUSIONS Metastases were more frequent in the abdomen than in the neck .\n\nTotal mediastinal and upper abdominal lymphadenectomy should be carefully conducted .\n\nCertain factors , such as tumor location , depth of tumor invasion , lymphovascular invasion , and paratracheal lymph node involvement , might be helpful in determining the need to perform cervical lymphadenectomy in individual patients .", "output": "Activating invasion and metastasis" }, { "input": "MDSCs and Tregs play an essential role in the immunosuppressive networks that contribute to tumor-immune evasion .\n\nThe mechanisms by which tumors promote the expansion and/or function of these suppressive cells and the cross-talk between MDSC and Treg remain incompletely defined .\n\nPrevious reports have suggested that MDSC may contribute to Treg induction in cancer .\n\nHerein , we provide evidence that tumor-induced gr-MDSCs , endowed with the potential of suppressing conventional T Lc , surprisingly impair TGF-\u03b21-mediated generation of CD4(+)CD25(+)FoxP3(+) iTregs .\n\nFurthermore , gr-MDSCs impede the proliferation of nTregs without , however , affecting FoxP3 expression .\n\nSuppression of iTreg differentiation from na\ufffdve CD4(+) cells by gr-MDSC occurs early in the polarization process , requires inhibition of early T cell activation , and depends on ROS and IDO but does not require arginase 1 , iNOS , NO , cystine/cysteine depletion , PD-1 and PD-L1 signaling , or COX-2 .\n\nThese findings thus indicate that gr-MDSCs from TB hosts have the unanticipated ability to restrict immunosuppressive Tregs .", "output": "Sustaining proliferative signaling, Avoiding immune destruction" }, { "input": "BACKGROUND Cholangiocarcinoma , a type of malignant tumor , originates from epithelial cells of the bile duct .\n\nPerineural invasion is common path for cholangiocarcinoma metastasis , and it is highly correlated with postoperative recurrence and poor prognosis .\n\nIt has been reported that muscarinic acetylcholine receptor M3 ( mAChR M3 ) is widely expressed in digestive tract cancer , and may play an important role in the proliferation , differentiation , transformation and carcinogenesis of tumors .\n\nThis study was to explore the effect of mAChR M3 on the growth of cholangiocarcinoma cells in vitro and provide a new approach to the pathogenesis and treatment of cholangiocarcinoma .\n\nMETHODS Streptavidin-biotin complex immunohistochemistry was carried out to assess the expression of mAChR M3 in surgical specimens of cholangiocarcinomas ( 40 cases ) and normal bile duct tissues ( 9 ) , as well as to investigate nerve infiltration .\n\nThe cholangiocarcinoma cells were treated with different concentrations of selective M-receptor agonist pilocarpine and M-receptor blocker atropine sulfate to induce changes in cell proliferation .\n\nThe experimental data were analyzed by the Chi-square test .\n\nRESULTS The strongly-positive expression rate of mAChR M3 was much higher in poorly-differentiated ( 69% , 9/13 ) than in well- and moderately-differentiated cholangiocarcinomas ( 30% , 8/27 ) ( X2=5.631 , P<0.05 ) .\n\nThe strongly-positive mAChR M3 expression rate in hilar cholangiocarcinoma ( 50% , 14/28 ) was higher than that in cholangiocarcinomas from the middle and lower common bile duct ( 25% , 3/12 ) ( X2=2.148 , P<0.05 ) .\n\nCholangiocarcinomas with distant metastasis had a strongly-positive expression rate ( 75% , 9/12 ) , which was much higher than those without distant metastasis ( 29% , 8/28 ) ( X2=7.410 , P<0.01 ) .\n\nThe absorbance value in the pilocarpine+atropine group was significantly higher than the corresponding value in the pilocarpine group .\n\nCONCLUSIONS The expression of mAChR M3 is influenced by the extent of differentiation , distant metastasis and the site of cholangiocarcinoma .\n\nIt also plays a key role in the proliferation and metastasis of cholangiocarcinoma .", "output": "Activating invasion and metastasis" }, { "input": "Colorectal cancer represents one of the most challenging diseases .\n\nIncreasing evidence indicates that aberrant expression of microRNAs ( miRNAs ) is related to pathogenesis of colorectal cancer .\n\nCancer cells reprogram metabolic pathways to sustain higher proliferation rates .\n\nWhether mechanisms underlying the role of miRNA in colorectal cancer are involved in metabolic reprogramming and the mechanisms through which miRNAs alter cancer metabolism are as yet unknown .\n\nHerein , we show that miR-124 , miR-137 and miR-340 are associated with poor prognosis of colorectal cancer .\n\nExpression of these miRNAs inhibits the growth of colorectal cancer cells .\n\nPKM ( pyruvate kinase isozyme ) alternative splicing proteins ( PTB1/hnRNAPA1/hnRNAPA2 ) , which control the inclusion of exon 9 ( PKM1 ) or exon 10 ( PKM2 ) , are targeted by miR-124 , miR-137 and miR-340 .\n\nConsequently , miR-124 , miR-137 and miR-340 switch PKM gene expression from PKM2 to PKM1 .\n\nHigh ratios of PKM1/PKM2 inhibit the glycolysis rate , but elevate the glucose flux into oxidative phosphorylation .\n\nThese results demonstrate that miRNAs ( miR-124 , miR-137 and miR-340 ) impair colorectal cancer growth by counteracting the Warburg effect due to regulating alternative splicing of the PKM gene .", "output": "Cellular energetics" }, { "input": "Mesenchymal stem cells ( MSCs ) are generally used in tissue engineering , regenerative medicine and therapy for immune disorder disease .\n\nMSCs are also employed as drug carriers for tumor therapy due to their ability to migrate to tumor tissue .\n\nHowever , due to the immunosuppressive function of MSCs , the application of MSCs in prostate cancer therapy remains limited .\n\nIn this study , we investigated the underlying mechanism by which MSCs enable prostate cancer cells to escape from immune surveillance in the inflammatory microenvironment .\n\nFirstly , we demonstrated that compared with the control groups , MSCs pretreated with IL-1\u03b1 effectively promoted the growth of the mouse prostate cancer cell line RM-1 invivo .\n\nFurthermore , when RM-1 prostate cancer cells were co-injected with MSCs pretreated with IL-1\u03b1 , tumor incidence significantly increased in allogeneic recipients .\n\nIn addition , we investigated the mechanism through which MSCs promote the ability of RM-1 cells to escape from immune injury .\n\nThe results revealed that IL-1\u03b1 led to the upregulation of TGF-\u03b2 in MSCs .\n\nThe inflammatory cytokine-induced promotive effect of MSCs on RM-1 cells in vivo was inhibited by TGF-\u03b2 siRNA .\n\nThe results of our study suggest that inflammatory cytokines induce the immunosuppressive function of MSCs which enables prostate cancer cells to escape from immune injury .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Avoiding immune destruction" }, { "input": "Glioblastoma is one of the most lethal and common malignant human brain tumors , with aggressive proliferation and highly invasive properties .\n\nHonokiol derived from Magnolia officinalis is able to cross the blood-brain barrier ( BBB ) and the blood-cerebrospinal fluid barrier ( BCSFB ) , suggesting a strong possibility that it could be an effective drug for the treatment of brain tumors , including glioblastoma .\n\nThus , we investigated the effects of honokiol on the expression of adhesion molecules in TNF-\u03b1-stimulated endothelial cells , and cancer growth and invasion were determined in T98G human glioblastoma cells .\n\nHonokiol dose-dependently inhibited the expression of intercellular adhesion molecule-1 ( ICAM-1 ) and vascular cell adhesion molecule-1 ( VCAM-1 ) in human umbilical vein endothelial cells ( HUVECs ) stimulated with TNF-\u03b1 for 6 h .\n\nPretreatment with honokiol significantly reduced the adhesion of T98G cells to HUVECs .\n\nMoreover , honokiol inhibited the invasion of T98G cells , suggesting that honokiol has an anti-metastatic effect .\n\nIn addition , honokiol increased the cytotoxicity of T98G cells in a dose- and time-dependent manner as assayed by MTT .\n\nTUNEL assay showed that honokiol significantly induced apoptosis in T98G cells at doses of 10 \ufffdM or more .\n\nThe induction of apoptotic cell death was mediated by the downregulation of the anti-apoptotic protein Bcl-2 and the upregulation of the pro-apoptotic protein Bax .\n\nTaken together , the results of this study suggest that honokiol exerts an anticancer effect by preventing metastasis and inducing apoptotic cell death of brain tumor cells .", "output": "Resisting cell death, Activating invasion and metastasis" }, { "input": "IFN-\u03b3 is a master regulator of the immune responses that occur in the transplanted kidney , acting both on the immune system and on the graft itself .\n\nThe cellular responses to IFN-\u03b3 are complex , and emerging evidence suggests that IFN-\u03b3 may regulate autophagic functions .\n\nConversely , autophagy modulates innate and adaptive immune functions in various contexts .\n\nIn this study , we identify a novel mechanism by which IFN-\u03b3 activates autophagy in human kidney epithelial cells and provide new insights into how autophagy regulates immune functions in response to IFN-\u03b3 .\n\nOur results indicate that IFN-\u03b3 promotes tryptophan depletion , activates the eIF2\u03b1 kinase general control nonderepressible-2 ( GCN2 ) , and leads to an increase in the autophagic flux .\n\nFurther , tryptophan supplementation and RNA interference directed against GCN2 inhibited IFN-\u03b3-induced autophagy .\n\nThis process is of functional relevance because autophagy regulates the secretion of inflammatory cytokines and growth factors by human kidney epithelial cells in response to IFN-\u03b3 .\n\nThese findings assign to IFN-\u03b3 a novel function in the regulation of autophagy , which , in turn , modulates IFN-\u03b3-induced secretion of inflammatory cytokines .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Altered p53 protein is prevalently associated with the pathologic class of triple-negative breast cancers and loss of p53 function has recently been linked to the induction of an epithelial-mesenchymal transition ( EMT ) and acquisition of stemness properties .\n\nWe explored the association between TP53 mutational status and expression of some genes involved in the canonical TGF-\\u03b2 signaling pathway ( the most potent EMT inducer ) and in two early EMT associated events : loss of cell polarity and acquisition of stemness-associated features .\n\nWe used a publicly accessible microarray dataset consisting of 251 p53-sequenced primary breast cancers .\n\nStatistical analysis indicated that mutant p53 tumors ( especially those harboring a severe mutation ) were consistent with the aggressive class of triple-negative cancers and that , differently from cell cultures , surgical tumors underexpressed some TGF-\\u03b2 related transcription factors known as involved in EMT ( ID1 , ID4 , SMAD3 , SMAD4 , SMAD5 , ZEB1 ) .\n\nThese unexpected findings suggest an interesting relationship between p53 mutation , mammary cell dedifferentiation , and the concomitant acquisition of stemlike properties ( as indicated by the overexpression of PROM1 and NOTCH1 genes ) , which improve tumor cells aggressiveness as indicated by the overexpression of genes associated with cell proliferation ( CDK4 , CDK6 , MKI67 ) and migration ( CXCR4 , MMP1 ) .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Breast cancer metastasis is the most common cause of cancer-related death in women .\n\nThus , seeking targets of breast tumor cells is an attractive goal towards improving clinical treatment .\n\nThe present study showed that CCL18 from tumor-associated macrophages could promote breast cancer metastasis via PITPNM3 .\n\nIn addition , we found that pachymic acid ( PA ) could dose-dependently inhibit migration and invasion of MDA-MB-231 cells , with or without rCCL18 stimulation .\n\nFurthermore , evidence was obtained that PA could suppress the phosphorylation of PITPNM3 and the combination of CCL18 and PITPNM3 .\n\nTherefore , we speculate that PA could inhibit breast cancer metastasis via PITPNM3 .", "output": "Activating invasion and metastasis" }, { "input": "The insulin-like growth factor ( IGF ) pathway represents one of the most studied molecular regulatory networks in oncology .\n\nClinical trials investigating the therapeutic value of anti-IGF1 receptor ( IGF1R ) therapies in cancer , including prostate cancer , are ongoing .\n\nHowever , the multiple functions of the IGF network in the prostate are not entirely known .\n\nTo elucidate the effects of IGF and insulin ( INS ) on prostate cells , we stimulated prostate cancer ( PC3 , DU145 , LNCaP , DUCaP ) and noncancerous prostate cells ( EP156T , RWPE-1 ) and observed differing responses : whereas cancer cells responded to IGF and INS exposure by way of enhanced cell proliferation and glucose consumption , basal to luminal differentiation was induced in noncancerous cells .\n\nThe same diverse responses were observed when the growth factor receptors IGF1R or INSR were overexpressed .\n\nDown-regulation of IGF1R or INSR isoform A ( INSRA ) also inhibited only proliferation of cancer cells .\n\nThe proliferative response induced by the INSR in cancer cells was mediated solely by the INSRA .\n\nMoreover we observed that the receptors of the IGF network mutually influence their expression and exert redundant functions , thus underscoring the functional molecular network formed by IGF , INS , IGF1R , and INSR .\n\nCollectively we found that both IGF1R and INSRA have oncogenic effects in prostate cancer , but the IGF network also has important physiological functions in the noncancerous prostate .\n\nThese data provide new insights into the biology of the IGF network in the prostate , thereby facilitating the design and interpretation of clinical studies investigating IGF1R targeting agents .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Although basal cell carcinoma ( BCC ) is the most common type of skin cancer , the incidence of metastasis is exceedingly low .\n\nOBJECTIVE Case presentation of a basal cell carcinoma arising on the dorsum of the foot with inguinal and pelvic lymph node metastases .\n\nMETHOD Case presentation with literature review .\n\nRESULT On the basis of our review of Japanese literature , the risk factors for BCC metastasis are occurrence on the genitalia , diameter of more than 3 cm , deep invasion of tumor cells into extradermal structures , and infiltrative/morpheic histological type .\n\nCONCLUSION Although metastasis from BCC is extremely rare , the prognosis of metastatic BCC is often poor .\n\nCareful follow-up is recommended in cases with high risk factors .", "output": "Activating invasion and metastasis" }, { "input": "Mutations in adenomatous polyposis coli ( APC ) gene are found in more than 80% of colorectal cancer ( CRC ) patients .\n\nThe nuclear transcription factor Nrf2 plays a central role in the regulation of oxidative stress and inflammation .\n\nPreviously , we have shown that chronic inflammation in Nrf2(-/-) ( Nrf2 knockout ; KO ) mice resulted in higher expression of inflammatory markers and cytokines , coupled with higher inflammatory damage to the colonic crypt cells , as compared to the Nrf2(+/+) ( wild type ; WT ) mice .\n\nInduction of mutation in the colon by administration of carcinogen , AOM prior to DSS-induced inflammation resulted in higher tumor incidence and numbers in Nrf2KO mice .\n\nThese results indicate that Nrf2-dependent inhibition of inflammation appears to be critical in inhibiting mutation-initiated colorectal carcinogenesis .\n\nIn this study , we aim to investigate if loss of Nrf2 would dose-dependently promote intestinal tumorigenesis in Apc(min/+) mice .\n\nTo demonstrate the in vivo mechanisms , we constructed both Apc mutated and Nrf2 deficient strain Apc(min/+) mice with C57BL/6 Nrf2KO mice to obtain F1 , Apc(min/+) ;Nrf2(+/-) and F2 , Apc(min/+) ;Nrf2(-/-) mice .\n\nNrf2KO decreased the protein expression of antioxidant enzyme NQO1 in Apc(min/+) .\n\nIn contrast , Nrf2KO enhanced the expression of inflammatory markers such as COX-2 , cPLA , LTB(4) in Apc(min/+) .\n\nFinally , Nrf2KO resulted in higher level of PCNA and c-Myc expression in intestinal tissue , indicating the deficiency of Nrf2 promotes proliferation of intestinal crypt cells in Apc(min/+) .\n\nTaken together , our results suggest that Nrf2KO attenuates anti-oxidative stress pathway , induces inflammation , and increases proliferative potential in the intestinal crypts leading to enhanced intestinal carcinogenesis and adenomas in Apc(min/+) . \u00a9 2012 Wiley Periodicals , Inc .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Hesperetin , a flavonoid from citrus fruits , has several bioactivities such as anti-inflammatory , antihypertensive , antiatherogenic effects .\n\nHowever , studies elucidating the role and the mechanism(s) of action of hesperetin in cervical cancer are sparse .\n\nIn this study , we investigated the mechanism of the antiproliferative and apoptotic actions exerted by hesperetin on human cervical cancer SiHa cells .\n\nThe viability of SiHa cells was evaluated using the MTT assay , apoptosis by acridine orange/ethidium bromide , propidium iodide , TUNEL assay , and Annexin V-Cy3 , cell cycle distribution and mitochondrial transmembrane potential using flow cytometry , and apoptotic marker genes using quantitative real-time PCR .\n\nThe treatment of SiHa cells with hesperetin ( IC(50,) 650\u03bcm ) showed a marked concentration- and time-dependent inhibition of proliferation and induced the G2/M phase in a dose-dependent manner after 24h .\n\nThere was an attenuation of mitochondrial membrane potential with increased expression of caspase-3 , caspase-8 , caspase-9 , p53 , Bax , and Fas death receptor and its adaptor protein Fas-associated death domain-containing protein ( FADD ) , indicating the participation of both death receptor- and mitochondria-related mechanisms .\n\nFurthermore , hesperetin-induced apoptosis was confirmed by TUNEL and Annexin V-Cy3 .\n\nThis study shows that hesperetin exhibits a potential anticancer activity against human cervical cancer cell lines in vitro through the reduction in cell viability and the induction of apoptosis .\n\nAltogether , these data sustain our contention that hesperetin has anticancer properties and merits further investigation as a potential therapeutic agent .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Placental Growth Factor ( PGF ) is a key molecule in angiogenesis .\n\nSeveral studies have revealed an important role of PGF primarily in pathological conditions ( e.g. : ischaemia , tumour formation , cardiovascular diseases and inflammatory processes ) suggesting its use as a potential therapeutic agent .\n\nHowever , to date , no information is available regarding the genetics of PGF variability .\n\nFurthermore , even though the effect of environmental factors ( e.g. : cigarette smoking ) on angiogenesis has been explored , no data on the influence of these factors on PGF levels have been reported so far .\n\nHere we have first investigated PGF variability in two cohorts focusing on non-genetic risk factors : a study sample from two isolated villages in the Cilento region , South Italy ( N=871 ) and a replication sample from the general Danish population ( N=1,812 ) .\n\nA significant difference in PGF mean levels was found between the two cohorts .\n\nHowever , in both samples , we observed a strong correlation of PGF levels with ageing and sex , men displaying PGF levels significantly higher than women .\n\nInterestingly , smoking was also found to influence the trait in the two populations , although differently .\n\nWe have then focused on genetic risk factors .\n\nThe association between five single nucleotide polymorphisms ( SNPs ) located in the PGF gene and the plasma levels of the protein was investigated .\n\nTwo polymorphisms ( rs11850328 and rs2268614 ) were associated with the PGF plasma levels in the Cilento sample and these associations were strongly replicated in the Danish sample .\n\nThese results , for the first time , support the hypothesis of the presence of genetic and environmental factors influencing PGF plasma variability .", "output": "Inducing angiogenesis" }, { "input": "To evaluate a radioprotective effect of sodium n-propyl thiosulfate ( NPTS ) and sodium 2-propenyl thiosulfate ( 2PTS ) derived from onions and garlic , respectively , rat hepatoma H4IIE cells and mouse lymphoma L5178Y cells were preincubated with each of these compounds for 48 hours at 37\u00b0C before receiving 10 Gy of X-ray irradiation .\n\nCell damage caused by the irradiation was quantified as comet tail moment , which represents the degree of DNA damage .\n\nX-ray-induced DNA damage was significantly decreased in both H4IIE and L5178Y cells by micromolar concentrations of NPTS and 2PTS compared with the control without the compounds .\n\nThe protective effect was more potent with 2PTS than NPTS .\n\nOnions and garlic have antiradiation potential .", "output": "Genomic instability and mutation" }, { "input": "Curcumin ( diferuloylmethane ) is a polyphenol derived from the plant turmeric ( Curcuma longa ) , which is commonly used as a spice .\n\nAlthough anti-carcinogenic , anti-oxidant , anti-inflammation , and anti-angiogenic properties have been reported , the effect of curcumin on breast cancer metastasis is unknown .\n\nMatrix metalloproteinase-9 ( MMP-9 ) is a major component in cancer cell invasion .\n\nIn this study , we investigated the inhibitory effect of curcumin on 12-O-tetradecanoylphorbol-13-acetate ( TPA)-induced MMP-9 expression and cell invasion and the molecular mechanisms involved in MCF-7 cells .\n\nOur results showed that curcumin inhibits TPA-induced MMP-9 expression and cell invasion through suppressing NF-\u03baB and AP-1 activation .\n\nAlso , curcumin strongly repressed the TPA-induced phosphorylation of p38 and JNK and inhibited TPA-induced translocation of PKC\u03b1 from the cytosol to the membrane , but did not affect the translocation of PKC\u03b4 .\n\nThese results indicate that curcumin-mediated inhibition of TPA-induced MMP-9 expression and cell invasion involves the suppression of the PKC\u03b1 , MAPK and NF-\u03baB/AP-1 pathway in MCF-7 cells .\n\nCurcumin may have potential value in restricting breast cancer metastasis .", "output": "Activating invasion and metastasis" }, { "input": "Androgen receptor ( AR ) signals have been suggested to contribute to bladder tumorigenesis and cancer progression .\n\nActivation of epidermal growth factor receptor ( EGFR ) also leads to stimulation of bladder tumor growth .\n\nHowever , crosstalk between AR and EGFR pathways in bladder cancer remains uncharacterized .\n\nWe have recently shown that androgens activate the EGFR pathway in bladder cancer cells .\n\nThe purpose of this study was to investigate the effects of EGF on AR activity in bladder cancer .\n\nEGF increased AR transcriptional activity by 1.2- , 1.9- and 2.0-fold in UMUC3 , 5637-AR and J82-AR cell lines , respectively , over mock treatment and a specific EGFR inhibitor , PD168393 , antagonized the EGF effect .\n\nCombined treatment of EGF and dihydrotestosterone ( DHT ) further induced AR transactivation while an AR antagonist , hydroxyflutamide ( HF ) , abolished the effect of not only DHT but also EGF .\n\nIn growth assays , EGF alone/DHT alone/EGF+DHT increased cell numbers by 16/12/19% , 6/14/18% and 30/12/38% in UMUC3-control-shRNA , 5637-AR and J82-AR , respectively , whereas the effects of EGF were marginal or less significant in UMUC3-AR-shRNA ( 8% ) or AR-negative 5637-V ( <1% ) and J82-V ( 17% ) cells .\n\nHF treatment at least partially counteracted the EGF effect on the growth of AR-positive cells .\n\nWestern blotting demonstrated that EGF , especially in the presence of DHT , upregulated the expression of the p160 coactivator TIF2 and HF again blocked this stimulation .\n\nCo-immunoprecipitation revealed the association between AR and estrogen receptor ( ER)-\u03b2 or Src in UMUC3 cells and stronger associations with EGF treatment , implying the involvement of the AR/ER/Src complex in EGF-increased AR transactivation and cell growth .\n\nCurrent results , thus , suggest that EGF promotes bladder cancer cell proliferation via modulation of AR signals .\n\nTaken together with our previous findings , crosstalk between EGFR and AR pathways can play an important role in the progression of bladder cancer .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Angiogenesis plays an important role in the progression of colorectal cancer ( CRC ) .\n\nStudies have indicated vascular endothelial growth factor ( VEGF ) is the predominant angiogenic factor .\n\nCyclin D1 ( CCND1 ) induces production of VEGF and is required for migration of blood vessels .\n\nOur aim was to determine the roles of CCND1 and VEGF overexpression in CRC patients .\n\nMETHODS We analyzed clinicopathological features , VEGF and CCND1 expressions by immunohistochemical ( IHC ) staining in 100 stage I-III CRC patients ( 44 were postoperative relapsed ; 56 were postoperative non-relapsed ) to determine the correlation between clinicopathologic features and co-existence of CCND1 and VEGF .\n\nFurthermore , the clinical outcomes of co-existence of CCND1 and VEGF were investigated .\n\nRESULTS Multivariate analysis showed vascular invasion ( P = 0.019 ) , VEGF overexpression ( P = 0.033 ) , and high postoperative serum carcinoembryonic antigen ( CEA ) levels ( P = 0.022 ) were independent predictors of postoperative relapse .\n\nCo-existence of CCND1 and VEGF overexpression had significantly poorer disease-free survival rates ( P \u2009= 0.004 ) and overall survival rates ( P = 0.001 ) than other phenotypes .\n\nCONCLUSIONS Co-existence of CCND1 and VEGF overexpression would potentially assist in TNM staging systems to predict the prognosis of these patients who would benefit from intensive follow-up and therapeutic programs .", "output": "Inducing angiogenesis" }, { "input": "Cancer immunoediting is the process whereby immune cells protect against cancer formation by sculpting the immunogenicity of developing tumors .\n\nAlthough the full process depends on innate and adaptive immunity , it remains unclear whether innate immunity alone is capable of immunoediting .\n\nTo determine whether the innate immune system can edit tumor cells in the absence of adaptive immunity , we compared the incidence and immunogenicity of 3'methylcholanthrene-induced sarcomas in syngeneic wild-type , RAG2(-/-) , and RAG2(-/-)x \u03b3c(-/-) mice .\n\nWe found that innate immune cells could manifest cancer immunoediting activity in the absence of adaptive immunity .\n\nThis activity required natural killer ( NK ) cells and interferon \u03b3 ( IFN-\u03b3 ) , which mediated the induction of M1 macrophages .\n\nM1 macrophages could be elicited by administration of CD40 agonists , thereby restoring editing activity in RAG2(-/-)x \u03b3c(-/-) mice .\n\nOur results suggest that in the absence of adaptive immunity , NK cell production of IFN-\u03b3 induces M1 macrophages , which act as important effectors during cancer immunoediting .", "output": "Avoiding immune destruction" }, { "input": "Cellular senescence has emerged as a critical tumor suppressive mechanism in recent years , but relatively little is known about how senescence occurs .\n\nHere , we report that secreted Frizzled-related protein 1 ( SFRP1 ) , a secreted antagonist of Wnt signaling , is oversecreted upon cellular senescence caused by DNA damage or oxidative stress .\n\nSFRP1 is necessary for stress-induced senescence caused by these factors and is sufficient for the induction of senescence phenotypes .\n\nWe present evidence suggesting that SFRP1 functions as a secreted mediator of senescence through inhibition of Wnt signaling and activation of the retinoblastoma ( Rb ) pathway and that cancer-associated SFRP1 mutants are defective for senescence induction .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Multiple endocrine neoplasia type 2 ( MEN2 ) is an autosomal , dominantly inherited disease characterized by medullary thyroid carcinoma , pheochromocytoma , and primary hyperparathyroidism and is divided into types 2A and 2B .\n\nFamilial medullary thyroid carcinoma ( FMTC ) is characterized by the presence of medullary thyroid carcinoma alone in family members and is considered to be one of the subtypes of MEN2 .\n\nClinical and genetic data on 505 Japanese patients from 275 MEN2 or FMTC families registered at 54 medical institutions were collected by the MEN Consortium of Japan .\n\nThe diagnosis was MEN2A in 343 ( 67.9% ) patients , MEN2B in 29 ( 5.7% ) , FMTC in 103 ( 20.4% ) , and unclassified in 30 ( 5.9% ) .\n\nMedullary thyroid carcinoma was found in 91.2% of patients ( 437/479 ) , pheochromocytoma in 45.6% ( 212/465 ) , and primary hyperparathyroidism in 8.1% ( 37/457 ) .\n\nRET genetic testing was performed in 410 patients , and the germline RET mutation was found in 98.8% ( 397/402 ) .", "output": "Genomic instability and mutation" }, { "input": "In lung cancer , platelet-derived growth factor receptor \u03b1 ( PDGFR\u03b1 ) is expressed frequently by tumor-associated stromal cells and by cancer cells in a subset of tumors .\n\nWe sought to determine the effect of targeting stromal PDGFR\u03b1 in preclinical lung tumor xenograft models ( human tumor , mouse stroma ) .\n\nEffects of anti-human ( IMC-3G3 ) and anti-mouse ( 1E10 ) PDGFR\u03b1 monoclonal antibodies ( mAb ) on proliferation and PDGFR\u03b1 signaling were evaluated in lung cancer cell lines and mouse fibroblasts .\n\nTherapy studies were conducted using established PDGFR\u03b1-positive H1703 cells and PDGFR\u03b1-negative Calu-6 , H1993 , and A549 subcutaneous tumors in immunocompromised mice treated with vehicle , anti-PDGFR\u03b1 mAbs , chemotherapy , or combination therapy .\n\nTumors were analyzed for growth and levels of growth factors .\n\nIMC-3G3 inhibited PDGFR\u03b1 activation and the growth of H1703 cells in vitro and tumor growth in vivo , but had no effect on PDGFR\u03b1-negative cell lines or mouse fibroblasts. 1E10 inhibited growth and PDGFR\u03b1 activation of mouse fibroblasts , but had no effect on human cancer cell lines in vitro .\n\nIn vivo , 1E10-targeted inhibition of murine PDGFR\u03b1 reduced tumor growth as single-agent therapy in Calu-6 cells and enhanced the effect of chemotherapy in xenografts derived from A549 cells .\n\nWe also identified that low expression cancer cell expression of VEGF-A and elevated expression of PDGF-AA were associated with response to stromal PDGFR\u03b1 targeting .\n\nWe conclude that stromal PDGFR\u03b1 inhibition represents a means for enhancing control of lung cancer growth in some cases , independent of tumor cell PDGFR\u03b1 expression .", "output": "Sustaining proliferative signaling" }, { "input": "We show that sufficient concentrations of gold nanoparticles produced by an original synthesis method in EMT-6 and CT-26 cancer cells make it possible to detect the presence , necrosis and proliferation of such cells after inoculation in live mice .\n\nWe first demonstrated that the nanoparticles do not interfere with the proliferation process .\n\nThen , we observed significant differences in the tumor evolution and the angiogenesis process after shallow and deep inoculation .\n\nA direct comparison with pathology optical images illustrates the effectiveness of this approach .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "The development of targeted therapies and immunotherapies has markedly advanced the treatment of metastasized melanoma .\n\nWhile treatment with selective BRAF(V600E) inhibitors ( like vemurafenib or dabrafenib ) leads to high response rates but short response duration , CTLA-4 blocking therapies induce sustained responses , but only in a limited number of patients .\n\nThe combination of these diametric treatment approaches may further improve survival , but pre-clinical data concerning this approach is limited .\n\nWe investigated , using Tyr::CreER(T2)PTEN(F-/-)BRAF(F-V600E/+) inducible melanoma mice , whether BRAF(V600E) inhibition can synergize with anti-CTLA-4 mAb treatment , focusing on the interaction between the BRAF(V600E) inhibitor PLX4720 and the immune system .\n\nWhile PLX4720 treatment strongly decreased tumor growth , it did not induce cell death in BRAF(V600E)/PTEN(-/-) melanomas .\n\nMore strikingly , PLX4720 treatment led to a decreased frequency of tumor-resident T cells , NK-cells , MDSCs and macrophages , which could not be restored by the addition of anti-CTLA-4 mAb .\n\nAs this effect was not observed upon treatment of BRAF wild-type B16F10 tumors , we conclude that the decreased frequency of immune cells correlates to BRAF(V600E) inhibition in tumor cells and is not due to an off-target effect of PLX4720 on immune cells .\n\nFurthermore , anti-CTLA-4 mAb treatment of inducible melanoma mice treated with PLX4720 did not result in enhanced tumor control , while anti-CTLA-4 mAb treatment did improve the effect of tumor-vaccination in B16F10-inoculated mice .\n\nOur data suggest that vemurafenib may negatively affect the immune activity within the tumor .\n\nTherefore , the potential effect of targeted therapy on the tumor-microenvironment should be taken into consideration in the design of clinical trials combining targeted and immunotherapy .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND The presence of distant metastases from colorectal cancer ( CRC ) does not preclude curative treatment .\n\nEarly detection of pulmonary metastases at a potentially curable stage could improve survival .\n\nThe aim of the present study was to assess the prognostic significance of commonly reported clinicopathologic features to identify high-risk patients who would likely benefit from more intensive chest surveillance for pulmonary metastases .\n\nMATERIAL AND METHOD A total of 351 consecutive patients , with surgical stages I-III colorectal cancer , who underwent curative resection at Phramongkutklao hospital from 1999 to 2005 , were followed regularly according to the established guidelines with routine physical examination , serum carcinoembryonic antigen ( CEA ) and colonoscopic surveillance .\n\nImaging studies for detecting metastases were computed tomography ( CT ) , plain film radiography , and ultrasonograpy .\n\nClinical and pathologic features were analyzed for their association with pulmonary metastasis .\n\nRESULTS There were 145 patients who had been operated for longer than five years after curative intent surgery .\n\nOf these , nineteen patients were lost to follow-up or died from other causes that were unrelated to colorectal cancer .\n\nPulmonary metastases were detected in 26 patients by either CXR or CT scan .\n\nMedian time to pulmonary metastasis was 19 months ( 95 percent CI , 12-35 ) .\n\nAccording to an univariate analysis , with log-rank test , identified four factors associated with pulmonary metastasis : Tumor stage T4 , Nodal stage N2 , elevation of serum CEA > 3.4 ng/ml and presence of lymphovascular invasion(LVI) .\n\nAccording to a multivariate analysis , with Cox regression , found an elevation of serum CEA > 3.4 ng/ml which was an independent factor that was significantly associated with pulmonary metastasis ( Hazard ratio ( HR ) , 8.9 ; 95 percent CI , 3.6-22 ; p < 0.01 ) .\n\nThe present study revealed that 50 percent of patients who had more than one of these risk factors would eventually develop pulmonary metastases .\n\nCONCLUSION An elevation of serum CEA > or = 3.4 ng/ml was found as an independent factor that was significantly associated with pulmonary metastasis whereas tumor stage T4 , nodal stage N2 and presence of lymphovascular invasion ( LVI ) were not independent clinicopathologic features associated with subsequent pulmonary metastases .\n\nChest CT scan has greater sensitivity than chest radiography in detection of pulmonary metastasis and should be considered as an imaging study of choice for intensive chest surveillance for patients who had more than one of these risk factors .", "output": "Activating invasion and metastasis" }, { "input": "Depression is the most common psychiatric syndrome in cancer patients and adversely affects anti-cancer immune system and life quality of patients .\n\nAntidepressant desipramine ( DMI ) is clinically prescribed in the auxiliary treatment of cancer patients .\n\nIncreasing evidences suggest that DMI has a broad spectrum of target-off biological effects , such as anticancer properties .\n\nOur previous study revealed that DMI at the clinical relevant concentrations could induce CHOP-dependent apoptotic death in C6 glioma cells .\n\nIn this study , we further explored the pro-autophagic effect of DMI in C6 glioma cells and its underlying mechanism .\n\nTreatment with DMI could induce autophagic cell death characterized by the formation of autophagosome and the elevated level of autophagic protein Beclin-1 and cellular redistribution of marker LC3 .\n\nMeanwhile , DMI inhibited the activation of PI3K-AKT-mTOR pathway which is considered as a negative regulator of autophagy .\n\nFurthermore , DMI activated PERK-eIF2\u03b1 and ATF6 of endoplasmic reticulum ( ER ) stress pathway , while knockdown of PERK with the PERK-specific short interfering RNA ( siRNA ) could obviously attenuate the autophagy .\n\nThe results strongly suggested that DMI could induce autophagy through the PERK-ER stress pathway in C6 glioma cells .\n\nOur findings provided new insights into another beneficial potential of antidepressant DMI in the adjuvant therapy of cancer .", "output": "Resisting cell death" }, { "input": "Regulator of Cullins-1 ( ROC1 ) or RING box protein-1 ( RBX1 ) is an essential RING component of Cullin-RING ligase ( CRL ) .\n\nOur previous studies showed that ROC1 is required for the growth of several cancer cell lines while ROC1 siRNA silencing inactivates CRL , leading to cell cycle arrest , cell senescence and/or apoptosis .\n\nHowever , it is completely unknown whether ROC1 knockdown triggers autophagic response by inactivating CRL .\n\nMoreover , the role of ROC1 in liver cancer remains elusive .\n\nIn this study , we reported that ROC1 knockdown significantly inhibited the growth of liver cancer cells by sequentially and independently inducing autophagy and p21-dependent cell senescence .\n\nMechanism analysis revealed that ROC1 silencing triggered autophagy by inhibition of mammalian target of rapamycin ( mTOR ) activity due to accumulation of mTOR-inhibitory protein Deptor , a substrate of CRL .\n\nConsistently , Deptor knockdown significantly blocked autophagy response upon ROC1 silencing .\n\nBiologically , autophagy response upon ROC1 silencing was a survival signal , and blockage of autophagy pathway sensitized cancer cells to apoptosis .\n\nFinally , we demonstrated that ROC1 was overexpressed in hepatocellular carcinomas , which is associated with poor prognosis of liver cancer patients .\n\nThese findings suggest that ROC1 is an appealing drug target for liver cancer and provide a proof-of-concept evidence for a novel drug combination of ROC1 inhibitor and an autophagy inhibitor for effective treatment of liver cancer by enhancing apoptosis.Cell Death and Differentiation advance online publication , 31 August 2012 ; doi:10.1038/cdd.2012.113 .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "Therapy-induced senescence ( TIS ) , a cytostatic stress response in cancer cells , is induced inefficiently by current anticancer agents and radiation .\n\nThe mechanisms that mediate TIS in cancer cells are not well defined .\n\nHerein , we characterize a robust senescence response both in vitro and in vivo to the quinone diaziquone ( AZQ ) , previously identified in a high-throughput senescence-induction small-molecule screen .\n\nUsing AZQ and several other agents that induce senescence , we screened a series of cyclin-dependent kinase inhibitors and found that p27(Kip1) was induced in all investigated prostate cancer cell lines .\n\nThe ubiquitin-ligase Skp2 negatively regulates p27(Kip1) and , during TIS , is translocated to the cytoplasm before its expression is decreased in senescent cells .\n\nOverexpression of Skp2 blocks the effects of AZQ on senescence and p27(Kip1) induction .\n\nWe also find that stable long-term short hairpin RNA knockdown of Skp2 decreases proliferation but does not generate the complete senescence phenotype .\n\nWe conclude that Skp2 participates in regulating TIS but , alone , is insufficient to induce senescence in cancer cells .", "output": "Enabling replicative immortality" }, { "input": "Elderly patients with acute myeloid leukemia generally have a poor prognosis and a highly heterogeneous clinical outcome .\n\nPrognostic indicators are required for and aid in patient stratification .\n\nHowever , the prognostic value of genetic mutations and immunophenotypic features in elderly normal karyotype acute myeloid leukemia , the largest cytogenetic risk group , remains unclear .\n\nWe investigated the genetic mutations NPM1 , FLT3-ITD , and FLT3-TKD and expression of the membrane antigens CD7 , CD15 , CD34 , and CD56 in 144 elderly patients with de novo normal karyotype acute myeloid leukemia to retrospectively analyze the prognostic and clinical relevance of these parameters .\n\nCD7 , CD15 , CD34 , and CD56 were expressed in 24% , 47% , 52% , and 15% of patients , respectively .\n\nNPM1 and FLT3-ITD mutations were detected in 51% and 17% of patients , respectively .\n\nComplete remission was obtained in 94 patients ( 65% ) , and the median overall survival was 16.5 months .\n\nUnivariate analysis detected 5 markers with prognostic relevance : high leukocyte count , FLT3-ITD mutations , NPM1 mutations , CD34 expression , and CD56 expression in acute myeloid leukemia blasts .\n\nIn multivariate analysis , patients with NPM1 predicted a higher complete remission ( CR ) rate ( P = .016 ) , longer event-free survival ( P = .008 ) , and longer overall survival ( P = .049 ) .\n\nFLT3-ITD mutations predicted a shorter event-free survival ( P = .002 ) and shorter overall survival ( P < .001 ) .\n\nCD56 remained an independent predictor for lower CR rate ( P = .021 ) and shorter event-free survival ( P = .002 ) .\n\nOur data highlight the prognostic importance of both genetic and immunophenotypic characteristics in this population of elderly patients with newly diagnosed normal karyotype acute myeloid leukemia .\n\nBy combining genetic and immunophenotypic markers , we can divide patients into distinct prognostic groups with important implications for prognostic stratification and risk-adapted therapy .", "output": "Genomic instability and mutation" }, { "input": "Angiogenesis is defined as the formation of new blood vessels form existing vessels surrounding a tumor .\n\nThe process of angiogenesis is an important step for tumor growth and metastasis , as is inflammation .\n\nThus , angiogenesis inhibitors that suppress inflammation have been studied as an anticancer treatment .\n\nRecently , many research groups have investigated the anti-angiogenic activity of natural compounds since some have been demonstrated to have anticancer properties .\n\nAmong many natural compounds , we focused on betaine , which is known to suppress inflammation .\n\nBetaine , trimethylglycine ( TMG ) , was first discovered in the juice of sugar beets and was later shown to be present in wheat , shellfish and spinach .\n\nIn Southeast Asia , betaine is used in traditional oriental medicine for the treatment of hepatic disorders .\n\nHere , we report the anti-angiogenic action of betaine .\n\nBetaine inhibited invitro angiogenic cascade , tube formation , migration and invasion of human umbilical vein endothelial cells ( HUVECs ) .\n\nBetaine also inhibited invivo angiogenesis in the mouse Matrigel plug assay .\n\nThe mRNA expression levels of basic fibroblast growth factor ( bFGF ) , matrix metalloproteinase-2 ( MMP-2 ) and matrix metalloproteinase-9 ( MMP-9 ) in HUVECs were decreased by betaine treatment .\n\nIn addition , betaine suppressed NF-\u03baB and Akt activation .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Dendritic cells ( DCs ) and natural killer ( NK ) cells are central components of innate immunity for controlling tumor growth .\n\nThe therapeutic effects of certain anti-myeloma drugs are partially mediated by targeting the innate immune response .\n\nIn addition , novel types of natural compounds have been developed that efficiently modulate the activity of both the cellular and humoral compartments of immunity .\n\nMGN-3 is known as an activator of natural killer cells , inducer of apoptosis and cytokine production , and modulator of dendritic cell maturation and differentiation in vitro .\n\nWe have performed a randomized , placebo-controlled study to examine the effects of MGN-3 on innate immune system parameters in 48 multiple myeloma patients .\n\nWe performed immunophenotypic analysis of peripheral blood samples , determined NK cell activity , and assessed the cytokine profiles of plasma before and during 3months of treatment .\n\nThe results demonstrate a clear increase in NK activity in MGN-3-treated patients compared to the placebo group , an increased level of myeloid DCs in peripheral blood , and augmented concentrations of T helper cell type 1-related cytokines .\n\nThe present study suggests that MGN-3 may represent an immunologically relevant product for activating innate immunity in multiple myeloma patients and warrants further testing to demonstrate clinical efficacy .", "output": "Avoiding immune destruction" }, { "input": "AIM To elucidate the roles of receptor tyrosine kinases RET and VEGFR2 and the RAF/MEK/ERK signaling cascade in cancer treatment with sorafenib .\n\nMETHODS The cell lines A549 , HeLa , and HepG2 were tested .\n\nThe enzyme activity was examined under cell-free conditions using 384-well microplate assays .\n\nCell proliferation was evaluated using the Invitrogen Alarmar Blue assay .\n\nGene expression was analyzed using the Invitrogen SYBR Green expression assays with a sequence detection system .\n\nProtein expression analysis was performed using Western blotting .\n\nRESULTS Sorafenib potently suppressed the activities of cRAF , VEGFR2 , and RET with IC(50) values of 20.9 , 4 and 0.4 nmol/L , respectively .\n\nSorafenib inhibited cRAF , VEGFR2 , and RET via non-ATP-competitive , ATP-competitive and mixed-type modes , respectively .\n\nIn contrast , sorafenib exerted only moderate cytotoxic effects on the proliferation of the 3 cell lines .\n\nThe IC(50) values for inhibition of A549 , HeLa , and HepG2 cells were 8572 , 4163 , and 8338 nmol/L , respectively .\n\nIn the 3 cell lines , sorafenib suppressed the cell proliferation mainly by blocking the MEK/ERK downstream pathway at the posttranscriptional level , which in turn regulated related gene expression via a feed-back mechanism .\n\nCONCLUSION This study provides novel evidence that protein kinases RET and VEGFR2 play crucial roles in cancer treatment with sorafenib .", "output": "Sustaining proliferative signaling" }, { "input": "The cadherins are a family of cell surface glycoproteins responsible for cell adhesion which play an important role in cell morphology , contact inhibition and signal transduction during tumorigenesis .\n\nProtocadherin 8 ( PCDH8 ) , a member of the cadherin family , has been reported to act as a tumor suppressor involved in oncogenesis in breast cancer .\n\nIn this study , we aimed to investigate the epigenetic inactivation of PCDH8 and its tumor suppressor function in gastric cancer .\n\nThe expression of PCDH8 was markedly reduced or silenced in gastric cancer cell lines compared with normal gastric cells or tissues .\n\nMethylation of the PCDH8 gene promoter was observed in 100% ( 4/4 ) of cell lines and 55.38% ( 36/65 ) of the primary gastric cancer by methylation-specific PCR , but not in normal gastric mucosa ( 0/10 ) .\n\nMethylated PCDH8 was significantly associated with lymph node metastasis in a logistic regression analysis .\n\nThe demethylation reagent 5-aza-2'-deoxycytidine was able to restore or upregulate PCDH8 expression in gastric cancer cell lines .\n\nEctopic expression of PCDH8 in silenced gastric cancer cells significantly inhibited cell migration and induced apoptosis .\n\nFor the first time , our study demonstrates the epigenetic inactivation of PCDH8 by promoter methylation and its tumor suppressor function in human gastric cancer .\n\nThus , PCDH8 could be identified as a candidate tumor suppressor in human gastric cancer .", "output": "Resisting cell death, Activating invasion and metastasis" }, { "input": "Background/Aims : Cimetidine has been shown to play an important role in the treatment of cancer and the regulation of the immune system .\n\nTherefore , we aimed to observe the effects of cimetidine on the systematic immune response in the perioperative period .\n\nMethodology : Sixty patients with colorectal cancer were enrolled from Jan 2005 to Dec 2005 from Taizhou Hospital .\n\nThe patients were administrated with cimetidine ( 0.8g\ufffdd-1 or 1.2g\ufffdd-1 ) or saline from the day of admission to the 10th POD .\n\nVenous blood sample was collected and the T- , B- and NK-lymphocyte subsets were determined by flow cytometry .\n\nThe specimens were subjected to tumor-infiltrating lymphocytes ( TILs ) response examination .\n\nResults : The levels of CD3 and CD4 T-lymphocytes were increased significantly in both low and high dose cimetidine groups 10 days after operation .\n\nThe number of CD19 B cells was also elevated by cimetidine .\n\nHowever , no significant changes were observed in the CD8 , CD4/CD8 value .\n\nTIL responses in the cimetidine groups were also enhanced significantly .\n\nConclusions : Cimetidine can alleviate systematic immunosuppression and improve the local immune function of the colorectal cancer patients in the perioperative period .", "output": "Avoiding immune destruction" }, { "input": "Therapy-induced cellular senescence ( TCS ) , characterized by prolonged cell cycle arrest , is an in vivo response of human cancers to chemotherapy and radiation .\n\nUnfortunately , TCS is reversible for a subset of senescent cells , leading to cellular reproliferation and ultimately tumor progression .\n\nThis invariable consequence of TCS recapitulates the clinical treatment experience of patients with advanced cancer .\n\nWe report the findings of a clinicopathological study in patients with locally advanced non-small cell lung cancer demonstrating that marker of in vivo TCS following neoadjuvant therapy prognosticate adverse clinical outcome .\n\nIn our efforts to elucidate key molecular pathways underlying TCS and cell cycle escape , we have previously shown that the deregulation of mitotic kinase Cdk1 and its downstream effectors are important mediators of survival and cell cycle reentry .\n\nWe now report that aberrant expression of Cdk1 interferes with apoptosis and promotes the formation of polyploid senescent cells during TCS .\n\nThese polyploid senescent cells represent important transition states through which escape preferentially occurs .\n\nThe Cdk1 pathway is in part modulated differentially by p21 and p27 two members of the KIP cyclin-dependent kinase inhibitor family during TCS .\n\nAltogether , these studies underscore the importance of TCS in cancer therapeutics .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "The aberrant activation of sonic hedgehog ( SHH ) pathway contributes to initiation and progression of various malignancies .\n\nHowever , the roles and underlying mechanisms of SHH signaling pathway in invasion and metastasis of liver cancer have not been well understood .\n\nIn this study , we found that SHH signaling was activated and correlated with invasion and metastasis in hepatocellular carcinoma ( HCC ) .\n\nEnhanced SHH signaling by recombinant human SHH N-terminal peptide ( rSHH-N ) promoted hepatoma cell adhesion , migration and invasion , whereas blockade of SHH signaling with SHH neutralizing antibody or cyclopamine suppressed hepatoma cell adhesion , migration and invasion .\n\nFurthermore , matrix metalloproteinase ( MMP)-2 and MMP-9 expressions and activities were upregulated and downregulated by rSHH-N and SHH signaling inhibitor , respectively .\n\nThe rSHH-N-mediated hepatoma cell migration and invasion was blocked by MMP-specific inhibitors or neutralizing antibodies to MMP-2 and MMP-9 .\n\nIn addition , phosphorylations of AKT and focal adhesion kinase ( FAK ) were increased and decreased by rSHH-N and SHH signaling inhibitor , respectively .\n\nFurther investigations showed that activation of AKT and FAK were required for rSHH-N-mediated upregulation of MMP-2 and MMP-9 , cell migration and invasion .\n\nFinally , we found that SHH protein expression was positively correlated with phosphorylatd FAK Tyr397 , phosphorylatd AKT Ser473 , MMP-2 and MMP-9 protein expressions in HCC samples .\n\nTaken together , our findings suggest that SHH pathway induces cell migration and invasion through FAK/AKT signaling-mediated MMP-2 and MMP-9 production and activation in liver cancer .", "output": "Activating invasion and metastasis" }, { "input": "PURPOSE This study investigated the impact of specific mutations in codon 12 of the Kirsten-ras ( KRAS ) gene on treatment efficacy in patients with metastatic colorectal cancer ( mCRC ) .\n\nPATIENTS Overall , 119 patients bearing a KRAS mutation in codon 12 were evaluated .\n\nAll patients received cetuximab-based first-line chemotherapy within the Central European Cooperative Oncology Group ( CECOG ) , AIO KRK-0104 or AIO KRK-0306 trials .\n\nRESULTS Patients with KRAS codon 12 mutant mCRC showed a broad range of outcome when treated with cetuximab-based first-line regimens .\n\nPatients with tumors bearing a KRAS p.G12D mutation showed a strong trend to a more favorable outcome compared to other mutations ( overall survival 23.3 vs. 14-18 months ; hazard ratio 0.66 , range 0.43-1.03 ) .\n\nAn interaction model illustrated that KRAS p.G12C was associated with unfavorable outcome when treated with oxaliplatin plus cetuximab .\n\nCONCLUSION The present analysis suggests that KRAS codon 12 mutation may not represent a homogeneous entity in mCRC when treated with cetuximab-based first-line therapy .", "output": "Genomic instability and mutation" }, { "input": "Anal canal cancer is a rare tumor without clear treatment evidence in the metastatic setting .\n\nIn terms of the bad prognosis of patients with metastatic anal cancer , further therapeutic options are urgently needed .\n\nIn this paper we present the case of a 64-year-old man suffering from undifferentiated squamous cell carcinoma with liver metastases .\n\nAfter progression on cisplatin and fluorouracil , tumor tissue was analyzed with respect to anti-EGFR therapy with cetuximab .\n\nThere was no KRAS mutation and the EGFR expression level in the tumor tissue was 2+ ; ideal conditions for the immunotherapy .\n\nEncouraged by these results we started a therapy using FOLFIRI in combination with cetuximab .\n\nFortunately the patient showed a partial response after 6 cycles .\n\nOn patient's preference we did a therapy break of 6 weeks .\n\nWithin this time period the disease was progressive indicating its aggressiveness .\n\nHowever , the same immunotherapy was able to stabilize the disease for a further 3 months .\n\nThe patient died 21 months after diagnosis because of liver failure .\n\nNevertheless , from our perspective the combination of FOLFIRI and cetuximab is quite a promising therapeutic option for patients with metastatic anal cancer .\n\nPotential predictive factors of the immunochemotherapy are discussed in this paper .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "We examined how well the human papillomavirus ( HPV ) E6 oncogene can function in the absence of the E7 oncogene during the carcinogenic process in human keratinocytes using a common HPV variant strongly associated with cervical cancer : the Asian-American E6 variant ( AAE6 ) .\n\nThis E6 variant is 20 times more frequently detected in cervical cancer than the prototype European E6 variant , as evidenced by independent epidemiological data .\n\nUsing cell culture and cell-based functional assays , we assessed how this variant can perform crucial carcinogenesis steps compared to the prototype E6 variant .\n\nThe ability to immortalize and transform primary human foreskin keratinocytes ( PHFKs ) to acquire resilient phenotypes and the ability to promote cell migration were evaluated .\n\nThe immortalization capability was assayed based on population doublings , number of passages , surpassing mortality stages 1 and 2 , human telomerase reverse transcriptase ( hTERT ) expression , and the ability to overcome G(1) arrest via p53 degradation .\n\nTransformation and migration efficiency were analyzed using a combination of functional cell-based assays .\n\nWe observed that either AAE6 or prototype E6 proteins alone were sufficient to immortalize PHFKs , although AAE6 was more potent in doing so .\n\nThe AAE6 variant protein alone pushed PHFKs through transformation and significantly increased their migration ability over that of the E6 prototype .\n\nOur findings are in line with epidemiological data that the AA variant of HPV16 confers an increased risk over the European prototype for cervical cancer , as evidenced by a superior immortalization , transformation , and metastatic potential .", "output": "Enabling replicative immortality, Activating invasion and metastasis" }, { "input": "Biallelic mutations in MCPH1 cause primary microcephaly ( MCPH ) with the cellular phenotype of defective chromosome condensation .\n\nMCPH1 encodes a multifunctional protein that notably is involved in brain development , regulation of chromosome condensation , and DNA damage response .\n\nIn the present studies , we detected that MCPH1 encodes several distinct transcripts , including two major forms : full-length MCPH1 ( MCPH1-FL ) and a second transcript lacking the six 3 ' exons ( MCPH1\\u0394e9-14 ) .\n\nBoth variants show comparable tissue-specific expression patterns , demonstrate nuclear localization that is mediated independently via separate NLS motifs , and are more abundant in certain fetal than adult organs .\n\nIn addition , the expression of either isoform complements the chromosome condensation defect found in genetically MCPH1-deficient or MCPH1 siRNA-depleted cells , demonstrating a redundancy of both MCPH1 isoforms for the regulation of chromosome condensation .\n\nStrikingly however , both transcripts are regulated antagonistically during cell-cycle progression and there are functional differences between the isoforms with regard to the DNA damage response ; MCPH1-FL localizes to phosphorylated H2AX repair foci following ionizing irradiation , while MCPH1\\u0394e9-14 was evenly distributed in the nucleus .\n\nIn summary , our results demonstrate here that MCPH1 encodes different isoforms that are differentially regulated at the transcript level and have different functions at the protein level .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "BACKGROUND Chemokines are involved in multiple aspects of pathogenesis and cellular trafficking in tumorigenesis .\n\nIn this study , we report that the latest member of the C-X-C-type chemokines , CXCL17 ( DMC/VCC-1 ) , recruits immature myeloid-derived cells and enhances early tumor progression .\n\nMETHODOLOGY/PRINCIPAL FINDINGS CXCL17 was preferentially expressed in some aggressive types of gastrointestinal , breast , and lung cancer cells .\n\nCXCL17 expression did not impart NIH3T3 cells with oncogenic potential in vitro , but CXCL17-expressing NIH3T3 cells could form vasculature-rich tumors in immunodeficient mice .\n\nOur data showed that CXCL17-expressing tumor cells increased immature CD11b(+)Gr1(+) myeloid-derived cells at tumor sites in mice and promoted CD31(+) tumor angiogenesis .\n\nExtensive chemotactic assays proved that CXCL17-responding cells were CD11b(+)Gr1(high)F4/80(-) cells ( \u2248 90% ) with a neutrophil-like morphology in vitro .\n\nAlthough CXCL17 expression could not increase the number of CD11b(+)Gr1(+) cells in tumor-burdened SCID mice or promote metastases of low metastatic colon cancer cells , the existence of CXCL17-responding myeloid-derived cells caused a striking enhancement of xenograft tumor formation .\n\nCONCLUSIONS/SIGNIFICANCE These results suggest that aberrant expression of CXCL17 in tumor cells recruits immature myeloid-derived cells and promotes tumor progression through angiogenesis .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Activating K-RAS mutations occur at a frequency of 90% in pancreatic cancer , and to date no therapies exist targeting this oncogene .\n\nK-RAS signals via downstream effector pathways such as the MAPK and the PI3K signaling pathways , and much effort has been focused on developing drugs targeting components of these pathways .\n\nTo better understand the requirements for K-RAS and its downstream signaling pathways MAPK and PI3K in pancreatic tumor maintenance , we established an inducible K-RAS knock down system that allowed us to ablate K-RAS in established tumors .\n\nKnock down of K-RAS resulted in impaired tumor growth in all pancreatic xenograft models tested , demonstrating that K-RAS expression is indeed required for tumor maintenance of K-RAS mutant pancreatic tumors .\n\nWe further examined signaling downstream of K-RAS , and detected a robust reduction of pERK levels upon K-RAS knock down .\n\nIn contrast , no effect on pAKT levels could be observed due to almost undetectable basal expression levels .\n\nTo investigate the requirement of the MAPK and the PI3K pathways on tumor maintenance , three selected pancreatic xenograft models were tested for their response to MEK or PI3K inhibition .\n\nTumors of all three models regressed upon MEK inhibition , but showed less pronounced response to PI3K inhibition .\n\nThe effect of MEK inhibition on pancreatic xenografts could be enhanced further by combined application of a PI3K inhibitor .\n\nThese data provide further rationale for testing combinations of MEK and PI3K inhibitors in clinical trials comprising a patient population with pancreatic cancer harboring mutations in K-RAS .", "output": "Genomic instability and mutation" }, { "input": "UNLABELLED BACKGROUND We aimed to develop a mouse spontaneous liver metastasis model from an orthotopically implanted human colon cancer cell line stably expressing a human sodium/iodide symporter ( NIS ) reporter gene , which can be imaged with single-photon emission computed tomography ( SPECT ) using 99mTcO4- .\n\nMETHODS A recombinant plasmid containing a constitutively driven NIS gene ( pcDNA3-NIS ) was transfected into the human colon cancer cell line HCT116 , and stable cell lines were established .\n\nThe stable cells were subcutaneously injected into the nude mice .\n\nWhen the diameter reached 10\u2009mm , the xenografts were excised , cut into small fragments , and orthotopically implanted into the cecal walls of another nude mice. 99mTcO4- SPECT/CT imaging was initiated 8\u2009weeks later and repeated every 1 to 2\u2009weeks .\n\nRESULTS The production and function of NIS protein was confirmed in vitro by Western blotting and 99mTcO4- uptake assay .\n\nOn SPECT/CT imaging , focal 99mTcO4- uptake was detected in the liver .\n\nNecropsy revealed local growth of the orthotopic colon xenografts with extensive invasion , microscopic serosal metastasis , and metastatic foci in the corresponding hepatic regions showing focal 99mTcO4- uptake .\n\nImmunohistochemistry revealed high levels of NIS expression in cells forming liver tumor , indicating that the liver tumor cells originated from the orthotopic colon xenografts .\n\nCONCLUSIONS The present proof-of-concept study provided a rationale for employing a radionuclide reporter gene for the specific visualization of spontaneous liver metastasis in living mice .\n\nThis unique animal model of clinically relevant and externally detectable liver metastasis will be a powerful tool for investigating tumor biology and developing novel therapies for cancer metastasis .", "output": "Activating invasion and metastasis" }, { "input": "Cancer cells convert glucose preferentially to lactate even in the presence of oxygen ( aerobic glycolysis-Warburg effect ) .\n\nNew concepts in cancer treatment aim at inhibition of aerobic glycolysis .\n\nPyruvate dehydrogenase converts pyruvate to acetylCoA thus preventing lactate formation .\n\nTherefore , the aim of this study was to evaluate compounds that could activate pyruvate dehydrogenase in cancer cells .\n\nWe investigated the effects of ( R)-(+)-\u03b1-lipoic acid ( LPA ) and dichloroacetate ( DCA ) , possible activators of pyruvate dehydrogenase , on suppression of aerobic glycolysis and induction of cell death .\n\nThe neuroblastoma cell lines Kelly , SK-N-SH , Neuro-2a and the breast cancer cell line SkBr3 were incubated with different concentrations ( 0.1-30 mM ) of LPA and DCA .\n\nThe effects of both compounds on cell viability/proliferation ( WST-1 assay ) , [ 18F]-FDG uptake , lactate production and induction of apoptosis ( flow cytometric detection of caspase-3 ) were evaluated .\n\nFurthermore , NMRI nu/nu mice that had been inoculated s.c. with SkBr3 cells were treated daily for four weeks with LPA ( i.p , 18.5 mg/kg ) starting at day 7 p.i. .\n\nTumor development was measured with a sliding calliper and monitored via [ 18F]-FDG-PET .\n\nResidual tumors after therapy were examined histopathologically .\n\nThese data suggests that LPA can reduce ( 1 ) cell viability/proliferation , ( 2 ) uptake of [ 18F]-FDG and ( 3 ) lactate production and increase apoptosis in all investigated cell lines .\n\nIn contrast , DCA was almost ineffective .\n\nIn the mouse xenograft model with s.c .\n\nSkBr3 cells , daily treatment with LPA retarded tumor progression .\n\nTherefore , LPA seems to be a promising compound for cancer treatment .", "output": "Cellular energetics, Resisting cell death" }, { "input": "The epidermal growth factor receptor ( EGFR ) is a member of the HER family receptors and its activation induced by its natural ligand EGF results in colon cancer growth and progression .\n\nPanitumumab ( pmAb ) is a fully human IgG2 anti-EGFR antibody that blocks the EGFR actions .\n\nIn the present study , we evaluated the effects of pmAb on the EGF-mediated cellular responses in a panel of colon cancer cells ( HCT-8 , HT-29 , DLD-1 and HCT-116 ) .\n\nHCT-1116 and DLD-1 cells showed no significant EGF-dependent cell proliferation ; HT-29 and HCT-8 exhibited an EGF-dependent proliferation , with HCT-8 cells to be the most responsive with significant EGFR phosphorylation upon treatment with EGF .\n\nThe effects of pmAb were then evaluated in the most EGF-responsive cells , HCT-8 .\n\nIn that respect , pmAb impedes the signaling cascade mediated by EGFR intracellular phosphorylation and activity of focal adhesion kinase ( FAK ) as well as the EGF-induced invasive and migratory potential of colon cancer cells .\n\nAt the level of matrix effectors implicated in colon cancer progression we report that pmAb is a potent inhibitor of constitute and EGF-mediated gene expression of certain matrix effectors , such as membrane-type 1 metalloproteinase ( MT1-MMP ) , extracellular metalloproteinases inducer ( EMMPRIN ) , urokinase plasminogen activator ( uPA ) and syndecan-4 .\n\nThe obtained data demonstrated that pmAb is a specific blocker of EGF-mediated EGFR activation , resulting in a significant inhibition of colon cancer cell proliferation in early stages of growth , migration and invasiveness as well as of matrix effector implicated in cancer progression .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Hypoxia in the tumor microenvironment plays a central role in the evolution of immune escape mechanisms by tumor cells .\n\nIn this study , we report the definition of miR-210 as a miRNA regulated by hypoxia in lung cancer and melanoma , documenting its involvement in blunting the susceptibility of tumor cells to lysis by antigen-specific cytotoxic T lymphocytes ( CTL). miR-210 was induced in hypoxic zones of human tumor tissues .\n\nIts attenuation in hypoxic cells significantly restored susceptibility to autologous CTL-mediated lysis , independent of tumor cell recognition and CTL reactivity .\n\nA comprehensive approach using transcriptome analysis , argonaute protein immunoprecipitation , and luciferase reporter assay revealed that the genes PTPN1 , HOXA1 , and TP53I11 were miR-210 target genes regulated in hypoxic cells .\n\nIn support of their primary importance in mediating the immunosuppressive effects of miR-210 , coordinate silencing of PTPN1 , HOXA1 , and TP53I11 dramatically decreased tumor cell susceptibility to CTL-mediated lysis .\n\nOur findings show how miR-210 induction links hypoxia to immune escape from CTL-mediated lysis , by providing a mechanistic understanding of how this miRNA mediates immunosuppression in oxygen-deprived regions of tumors where cancer stem-like cells and metastatic cellular behaviors are known to evolve .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "BACKGROUND \u0393-Ionizing radiation ( IR ) therapy is one of major therapeutic tools in cancer treatment .\n\nNevertheless , \u03b3-IR therapy failed due to occurrence of metastasis , which constitutes a significant obstacle in cancer treatment .\n\nThe main aim of this investigation was to construct animal model which present metastasis during radiotherapy in a mouse system in vivo and establishes the molecular mechanisms involved .\n\nMATERIALS AND METHODS The C6L transfectant cell line expressing firefly luciferase ( fLuc ) was treated with \u03b3-IR , followed by immunoblotting , zymography and invasion assay in vitro .\n\nWe additionally employed the C6L transfectant cell line to construct xenografts in nude mice , which were irradiated with \u03b3-IR .\n\nIrradiated xenograft-containing mice were analyzed via survival curves , measurement of tumor size , and bioluminescence imaging in vivo and ex vivo .\n\nMetastatic lesions in organs of mice were further assessed using RT-PCR , H & E staining and immunohistochemistry .\n\nRESULTS \u03b3-IR treatment of C6L cells induced epithelial-mesenchymal transition ( EMT ) and increased cell invasion .\n\nIn irradiated xenograft-containing mice , tumor sizes were decreased dramatically and survival rates extended .\n\nAlmost all non-irradiated xenograft-containing control mice had died within 4 weeks .\n\nHowever , we also observed luminescence signals in about 22.5% of \u03b3-IR-treated mice .\n\nIntestines or lungs of mice displaying luminescence signals contained several lesions , which expressed the fLuc gene and presented histological features of cancer tissues as well as expression of EMT markers .\n\nCONCLUSIONS These findings collectively indicate that occurrences of metastases during \u03b3-IR treatment accompanied induction of EMT markers , including increased MMP activity .\n\nEstablishment of a murine metastasis model during \u03b3-IR treatment should aid in drug development against cancer metastasis and increase our understanding of the mechanisms underlying the metastatic process .", "output": "Activating invasion and metastasis" }, { "input": "Senescence , an inherent tumor suppressive mechanism , is a critical determinant for chemotherapy .\n\nIn the present study , we show that the monocarboxylate transporter 2 ( MCT2 ) protein was tumor-selectively expressed in human colorectal malignancies and knockdown of MCT2 induces mitochondrial dysfunction , cell-cycle arrest , and senescence without additional cellular stress in colorectal cancer cell lines .\n\nMoreover , the reactive oxygen species ( ROS ) scavenger , N-acetylcysteine , blocked MCT2 knockdown-induced growth arrest and cellular senescence , indicating a pivotal role of ROS in this pathway .\n\nDramatic induction of mitochondrial superoxide generation and decrease in ATP production was observed , indicating that mitochondrial dysfunction is the major mechanism underlying MCT2 knockdown-induced ROS generation .\n\nSenescence-associated DNA damage was also evident from the increase in promyelocytic leukemia bodies , \u03b3H2AX foci , and SAHF .\n\nConversely , overexpression of MCT2 prevented doxorubicin-induced ROS accumulation ( P = 0.0002 ) and cell growth inhibition ( P = 0.001 ) .\n\nMCT2 knockdown suppressed KRAS mutant colorectal tumor growth in vivo .\n\nIn addition , MCT2 knockdown and cytostatic drug combination further enhanced the antitumor effect .\n\nThese findings support the use of MCT2 as a promising target for inhibition of colorectal cancer .", "output": "Genomic instability and mutation, Tumor promoting inflammation, Enabling replicative immortality, Sustaining proliferative signaling" }, { "input": "OBJECTIVE To investigate the relationship between lymphangiogenesis and lymphatic metastasis in cervical squamous carcinoma .\n\nMETHODS Eighty cases of invasive cervical squamous cancer were selected as objects of our study .\n\nDouble immunohistochemical staining with antibodies against lymphatic vessel endothelial hyaluronan receptor 1 and Ki-67 was used to label the lymphatic vessels and mark the proliferative lymphatic vessels in cervical squamous cancer .\n\nThe peritumoral lymphatic vessel density and intratumoral lymphatic vessel density was assessed .\n\nThe lymphatic vessels proliferation index was evaluated by calculating Ki-67 proliferation index ( PI ) to reflect the lymphangiogenesis of cervical squamous cancer .\n\nThen the correlation between lymphangiogenesis and clinicopathologic features of cervical squamous cancer was analyzed .\n\nRESULTS The LVD of cervical cancer ( 15.23 \ufffd 3.6 ) was clearly higher than that of the adjacent normal cervical subepithelial tissues ( 9.9 \ufffd 2.5 , P < 0.001 ) .\n\nThe peritumoral lymphatic vessel density of cervical cancer ( 18.75 \ufffd 4.3 ) was significantly higher than the intratumoral lymphatic vessel density of cervical cancer ( 11.71 \ufffd 4.9 , P < 0.001 ) .\n\nLymphatic PI ( LPI ) of cervical cancer ( 0.258 \ufffd 0.07 ) was higher than that of the adjacent normal cervical subepithelial tissues ( 0.068 \ufffd 0.08 , P < 0.001 ) .\n\nThe peritumoral lymphatic vessel PI of cervical cancer ( 0.324 \ufffd 0.06 ) was notably higher than the intratumoral lymphatic vessel PI of cervical cancer ( 0.232 \ufffd 0.06 , P < 0.001 ) .\n\nPeritumoral lymphatic vessel density and peritumoral lymphatic vessel were clearly associated with the lymph node metastasis ( P = 0.001 and P = 0.002 , respectively ) and lymphovascular space invasion ( P = 0.024 and P = 0.01 , respectively ) .\n\nCONCLUSIONS The high density of peritumoral lymphatic vessels is a potential predictor of more aggressive phenotype of cervical squamous cancer .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Liver cancer ranks as the fifth most prevalent malignancy of all cancers worldwide .\n\nAccording to the principles of traditional Chinese medicine , liver Yin deficiency is a common clinical syndrome of liver cancer , and tonifying liver Yin is a common treatment method for liver cancer .\n\nHowever , no hepatocarcinoma-specific liver Yin tonifying formula has yet been established .\n\nIn the present study , we established a liver cancer-specific combination of herbs , which we term liver Yin tonifying formula ( LYTF ) .\n\nWe found that LYTF inhibits the proliferation of Bel-7402 cells in a dose- and time-dependent manner .\n\nLYTF induces apoptosis in Bel-7402 cells , which is accompanied by activation of caspases-8 , -9 and -3 .\n\nPan-caspase blocking completely abrogates LYTF-induced apoptosis and partially abrogates LYTF-induced proliferation inhibition .\n\nLYTF also induces cell senescence , as indicated by a large and flattened morphology , senescence-activated \u03b2-galactosidase-positive staining and G0/G1 cell cycle arrest , accompanied by the up-regulation of p16 and p21 and the down-regulation of retinoblastoma protein phosphorylation .\n\nThese findings suggest that LYTF is effective in inhibiting the growth and survival of hepatocarcinoma cells through the induction of apoptosis and cell senescence .\n\nOur study also provides insight into traditional Chinese medicine methods used for the treatment of liver cancer .", "output": "Enabling replicative immortality, Evading growth suppressors, Resisting cell death" }, { "input": "RASSF2 has recently been identified as a potential tumor suppressor that serves as a Ras effector in various types of human cancers .\n\nHowever , there have been few reports detailing this in gastric cancer .\n\nSamples of gastric adenocarcinoma from 276 Chinese patients with follow-up were analyzed for RASSF2 protein expression by immunohistochemistry .\n\nRASSF2 was expressed in up to 31.2% ( 86/276 ) of this group of gastric carcinoma .\n\nThe expression of RASSF2 was significantly lower in carcinomas than in normal mucosas ( P<0.05 ) .\n\nRASSF2 corresponded positively with patient age , histological differentiation , depth of tumor invasion , regional lymph node and distant metastasis , and TNM stage ( all P<0.05 ) .\n\nFurther multivariate analysis revealed that patient gender , depth of tumor invasion , distant metastasis , TNM stage and the expression of RASSF2 were independent prognostic factors for patients with gastric cancer .\n\nThe Kaplan-Meier plot showed that the overall mean survival time of the patients with RASSF2-negative expression was shorter than that of patients with positive expression ( \u03c7(2)=156.874 , P<0.0001 ) .\n\nMoreover , RASSF2-negative expression had a much more significant effect on the survival of those patients with early stage tumors ( \u03c7(2)=127.167 , P<0.0001 ) , highlighted by a >50.9% reduction in 3-year survival compared to that of patients with RASSF2-positive expression .\n\nIn late stages , the difference was also significant ( \u03c7(2)=6.246 , P=0.019 ) , with a 35.5% reduction in 3-year survival .\n\nIt is suggested that RASSF2 plays an important role in the evolution of gastric adenocarcinoma and should be considered as a potential marker for its prognosis .", "output": "Activating invasion and metastasis" }, { "input": "Cellular senescence can be a functional barrier to carcinogenesis .\n\nWe hypothesized that inflammation modulates carcinogenesis through senescence and DNA damage response ( DDR ) .\n\nWe examined the association between senescence and DDR with macrophage levels in inflammatory bowel disease ( IBD ) .\n\nIn vitro experiments tested the ability of macrophages to induce senescence in primary cells .\n\nInflammation modulating microRNAs were identified in senescence colon tissue for further investigation .", "output": "Enabling replicative immortality" }, { "input": "Despite insights into the molecular pathways regulating hypoxia-induced gene expression , it is not known which cell types accomplish oxygen sensing during neo-vasculogenesis .\n\nWe have developed a humanized mouse model of endothelial and mesenchymal progenitor co-transplantation to delineate the cellular compartments responsible for hypoxia response during vasculogenesis .\n\nMesenchymal stem/progenitor cells ( MSPCs ) accumulated nuclear hypoxia-inducible transcription factor ( HIF)-1\u03b1 earlier and more sensitively than endothelial colony forming progenitor cells ( ECFCs ) in vitro and in vivo .\n\nHypoxic ECFCs showed reduced function in vitro and underwent apoptosis within 24h in vivo when used without MSPCs .\n\nSurprisingly , only in MSPCs did pharmacologic or genetic inhibition of HIF-1\u03b1 abrogate neo-vasculogenesis .\n\nHIF deletion in ECFCs caused no effect .\n\nECFCs could be rescued from hypoxia-induced apoptosis by HIF-competent MSPCs resulting in the formation of patent perfused human vessels .\n\nSeveral angiogenic factors need to act in concert to partially substitute mesenchymal HIF-deficiency .\n\nResults demonstrate that ECFCs require HIF-competent vessel wall progenitors to initiate vasculogenesis in vivo and to bypass hypoxia-induced apoptosis .\n\nWe describe a novel mechanistic role of MSPCs as oxygen sensors promoting vasculogenesis thus underscoring their importance for the development of advanced cellular therapies .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "Background .\n\nAn important goal of personalized cancer therapy is to tailor specific therapies to the mutational profile of individual patients .\n\nHowever , whole genome sequencing studies have shown that the mutational profiles of cancers evolve over time and often differ between primary and metastatic sites .\n\nActivating point mutations in the PIK3CA gene are common in primary breast cancer tumors , but their presence in breast cancer bone metastases has not been assessed previously .\n\nResults .\n\nFourteen patients with breast cancer bone metastases were biopsied by three methods : CT-guided bone biopsies ; bone marrow trephine biopsies ; and bone marrow aspiration .\n\nSamples that were positive for cancer cells were obtained from six patients .\n\nThree of these patients had detectable PIK3CA mutations in bone marrow cancer cells .\n\nPrimary tumor samples were available for four of the six patients assessed for PIK3CA status in their bone metastases .\n\nFor each of these , the PIK3CA mutation status was the same in the primary and metastatic sites .\n\nConclusions .\n\nPIK3CA mutations occur frequently in breast cancer bone metastases .\n\nThe PIK3CA mutation status in bone metastases samples appears to reflect the PIK3CA mutation status in the primary tumour .\n\nBreast cancer patients with bone metastases may be candidates for treatment with selective PIK3CA inhibitors .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "Multiple myeloma ( MM ) is classically illustrated by a desynchronized cytokine system with rise in inflammatory cytokines .\n\nThere are recent reports which emphasized the potential role of angiogenesis in the development of MM. Role of cyclooxygenase 2 ( COX-2 ) is well documented in the pathogenesis of solid tumors , but little is known about its occurrence and function in hematologic neoplasms .\n\nInvolvement of neoangiogenesis is reported in the progression of MM , and angiopoietins probably contribute to this progression by enhancing neovascularization .\n\nCirculatory and mRNA levels of angiogenic factors and cyclooxygenase were determined in 125 subjects ( 75 MM patients and 50 healthy controls ) by using enzyme-linked immunosorbent assay and quantitative PCR .\n\nWe observed significant increase for angiogenic factors ( Ang-1 , Ang-2 , hepatocyte growth factor , and vascular endothelial growth factor ) and cyclooxygenase at circulatory level , as well as at mRNA level , as compared to healthy controls except insignificant increase for Ang-1 at circulatory level .\n\nWe have also observed the significant positive correlation of all angiogenic factors with cyclooxygenase .\n\nThe strong association found between angiogenic factors and COX-2 in this study may lead to the development of combination therapeutic strategy to treat MM. Therefore , targeting COX-2 by using its effective inhibitors demonstrating antiangiogenic and antitumor effects could be used as a new therapeutic approach for treatment of MM .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "MAP17 is a small 17 kDa non-glycosylated membrane protein previously identified as being overexpressed in carcinomas .\n\nBreast tumor cells that overexpress MAP17 show an increased tumoral phenotype with enhanced proliferative capabilities both in the presence or the absence of contact inhibition , decreased apoptotic sensitivity , and increased migration .\n\nMAP17-expressing clones also grow better in nude mice .\n\nThe increased malignant cell behavior induced by MAP17 is associated with an increase in reactive oxygen species ( ROS ) production , and the treatment of MAP17-expressing cells with antioxidants results in a reduction in the tumorigenic properties of these cells .\n\nThe MAP17-dependent increase in ROS and tumorigenesis relies on its PDZ-binding domain because disruption of this sequence by point mutations abolishes the ability of MAP17 to enhance ROS production and tumorigenesis .\n\nMAP17 is overexpressed in a great variety of human carcinomas , including breast tumors .\n\nImmunohistochemical analysis of MAP17 during cancer progression demonstrates that overexpression of the protein strongly correlates with tumoral progression .\n\nGeneralized MAP17 overexpression in human carcinomas indicates that MAP17 can be a good marker for tumorigenesis and , especially , for malignant progression .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "BACKGROUND Coronins are a family of highly evolutionary conserved proteins reportedly involved in the regulation of actin cytoskeletal dynamics , although only coronin 3 has been shown to be related to cancer cell migration .\n\nIn glioblastoma cells , the knockdown of coronin 3 inhibits cell proliferation and invasion .\n\nCoronin 3 is also associated with the aggression and metastasis of hepatocellular carcinoma .\n\nIn this paper , we analyze the migration , invasion and metastasis abilities of gastric cancer cells after up- or down-regulation of coronin 3 , and explore the mechanism of coronin 3 in the process of gastric cancer metastasis .\n\nRESULTS The expression of coronin 3 was higher in the highly metastatic sub-cell line MKN28-M , which we established in our laboratory .\n\nWe also demonstrated that the expression of coronin 3 was remarkably higher in lymph lode metastases than in primary gastric cancer tissues , and over-expression of coronin 3 was correlated with the increased clinical stage and lymph lode metastasis .\n\nRecombinant lentiviral vectors encoding shRNAs were designed to down-regulate coronin 3 expression in gastric cancer cell lines .\n\nStable knockdown of coronin 3 by this lentiviral vector could efficiently inhibit the migration and invasion of MKN45 gastric cancer cells .\n\nIn contrast , up-regulation of coronin 3 significantly enhanced migration and invasion of MKN28-NM cells .\n\nIn addition , knockdown of coronin 3 significantly reduced liver metastasis in mice after tail vein injection of gastric cancer cells .\n\nThe Human Tumor Metastasis PCR Array was used to screen the metastasis-associated genes identified by the down-regulation of coronin 3 , and the results suggested that , following the knockdown of coronin 3 , the tumor cell migration and invasion were inhibited by the reduced expression of MMP-9 and cathepsin K. CONCLUSION Coronin 3 is highly expressed in gastric cancer metastases and can promote the metastatic behaviors of gastric cancer cells , including their migration and invasion .", "output": "Activating invasion and metastasis" }, { "input": "Immunosenescence , the progressive decline of adaptive immunity and chronic inflammation with ageing has been demonstrated to be the main factor responsible for infections , cancer and autoimmune conditions in the elderly .\n\nSenescence-accelerated mouse ( SAM ) was used to study the protective effects of Pu-erh tea in the elderly .\n\nThe senile-prone sub-strain , SAM-P8 mice were administered individually with ripened or crude Pu-erh tea at 125 , 250 or 500mg/kg .\n\nThe results showed that Pu-erh tea significantly increased the fractions of na\ufffdve T lymphocytes , CD8(+)CD28(+) T lymphocytes and NK cells in the peripheral blood , but decreased the levels of IL-6 in aged mice .\n\nThese data suggested that the Pu-erh tea reversed the immunosenescence by restoring the immune deficiency and decreasing pro-inflammatory cytokine .\n\nThus , long term drinking of Pu-erh tea may be beneficial for the aged population in terms of increasing the body's resistance to infection and cancer .", "output": "Avoiding immune destruction" }, { "input": "The aim of this work was to characterize the antitumoral activity of the plant compound 7-epi-nemorosone in prostate carcinoma cell lines .\n\nProstate cancer is the most frequently diagnosed malignancy and the second-leading cause of cancer death in men .\n\nIn spite of the current therapeutic options for this cancer entity , many patients die due to metastases in distant organs and acquired chemotherapy resistance .\n\nThus , approaches to provide improvements in outcome and quality of life for such patients are urgently needed .\n\nRecently , the polyisoprenylated benzophenone 7-epi-nemorosone , originally collected by honeybees from Clusia rosea and Clusia grandiflora ( Clusiaceae ) , has been described to be a potent antitumoral agent .\n\nHere , its activity in prostate carcinoma is reported. 7-epi-nemorosone was isolated from Caribbean propolis employing RP-HPLC techniques .\n\nIts cytotoxicity was assessed using the MTT proliferation assay in human androgen-dependent prostate carcinoma LNCaP cells including an MDR1(+) sub-line .\n\nNo cross-resistance was detected .\n\nFACS-based cell cycle analysis revealed a significant increase in the sub-G0/G1 , G1 , and depletion in the S phase populations .\n\nA concomitant down-regulation of cyclins D1/D3 and CDK 4/6 in LNCaP cells was detected by Western blot .\n\nAnnexin-V-FITC labeling and caspase-3 cleavage assays showed that 7-epi-nemorosone induced apoptotic events .\n\nMajor signal transduction elements such as p38 MAPK and Akt/PKB as well as androgen receptor AR and PSA production were found to be down-regulated after exposure to the drug .\n\nERK1/2 protein levels and phosphorylation status were down-regulated accompanied by up-regulation but inhibition of the activity of their immediate upstream kinases MEK1/2 .\n\nAdditionally , Akt/PKB enzymatic activity was effectively inhibited at a similar concentration as for MEK1/2 .\n\nHere , we demonstrate for the first time that 7-epi-nemorosone exerts cytotoxicity in an androgen-dependent prostate carcinoma entity by targeting the MEK1/2 signal transducer .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "It is well established that aberrant production of inflammatory mediators has been associated with most the toxic manifestations and the genesis of different chronic diseases including cancer .\n\nThe basic aim of the present study is to investigate the effects of soy isoflavones ( SIF ) on 12-O-tetradecanoylphorbol-13-acetate ( TPA)-induced cutaneous inflammatory responses and to explore the underlying molecular mechanisms .\n\nWe have studied the protective effects of SIF against TPA induced oxidative stress , pro-inflammatory cytokines level , activation of NF-\u03baB , expression of COX-2 and ki-67 in mouse skin .\n\nAnimals were divided into five groups I-V ( n=6 ) .\n\nGroups II , III and IV received topical application of TPA at the dose of 10 nmol/0.2 ml of acetone/animal/day , for 2 days .\n\nAnimals of the groups III and IV were pre-treated with SIF 1.0 \u03bcg ( D1 ) and 2.0 \u03bcg ( D2 ) topically 30 min prior to each TPA administration , while groups I and V were given acetone ( 0.2 ml ) and SIF ( D2 ) , respectively .\n\nWe have found that SIF pretreatment significantly inhibited TPA induced oxidative stress , proinflammatory cytokines production and activation of NF-\u03baB .\n\nSIF also inhibited the expression of COX-2 and ki-67 .\n\nHistological findings further supported the protective effects of SIF against TPA-induced cutaneous damage .\n\nThus , our results suggest that inhibitory effect of SIF on TPA-induced cutaneous inflammation includes inhibition of proinflammatory cytokines , attenuation of oxidative stress , activation of NF-\u03baB and expression of COX-2 .", "output": "Tumor promoting inflammation" }, { "input": "Contact inhibition is a crucial mechanism regulating proliferation in vitro and in vivo .\n\nAlthough it is generally accepted that contact inhibition plays a pivotal role in maintaining tissue homeostasis , the molecular mechanisms of contact inhibition are still not fully understood .\n\nFoxM1 is known as a proliferation-associated transcription factor and is upregulated in many cancer types .\n\nVice versa , anti-proliferative signals , such as TGF-\\u03b2 and differentiation signals decrease FoxM1 expression .\n\nHere we investigated the role of FoxM1 in contact inhibition in fibroblasts .\n\nWe show that protein expression of FoxM1 is severely and rapidly downregulated upon contact inhibition , probably by inhibition of ERK activity , which then leads to decreased expression of cyclin A and polo-like kinase 1 .\n\nVice versa , ectopic expression of FoxM1 prevents the decrease in cyclin A and polo-like kinase 1 and causes a two-fold increase in saturation density indicating loss of contact inhibition .\n\nHence , we show that downregulation of FoxM1 is required for contact inhibition by regulating expression of cyclin A and polo-like kinase 1 .", "output": "Evading growth suppressors" }, { "input": "AIM To investigate whether luteolin , a highly prevalent flavonoid , reverses the effects of epithelial-mesenchymal transition ( EMT ) in vitro and in vivo and to determine the mechanisms underlying this reversal .\n\nMETHODS Murine malignant melanoma B16F10 cells were exposed to 1% O(2) for 24 h .\n\nCellular mobility and adhesion were assessed using Boyden chamber transwell assay and cell adhesion assay , respectively .\n\nEMT-related proteins , such as E-cadherin and N-cadherin , were examined using Western blotting .\n\nFemale C57BL/6 mice ( 6 to 8 weeks old ) were injected with B16F10 cells ( 1\ufffd10(6) cells in 0.2 mL per mouse ) via the lateral tail vein .\n\nThe mice were treated with luteolin ( 10 or 20 mg/kg , ip ) daily for 23 d .\n\nOn the 23rd day after tumor injection , the mice were sacrificed , and the lungs were collected , and metastatic foci in the lung surfaces were photographed .\n\nTissue sections were analyzed with immunohistochemistry and HE staining .\n\nRESULTS Hypoxia changed the morphology of B16F10 cells in vitro from the cobblestone-like to mesenchymal-like strips , which was accompanied by increased cellular adhesion and invasion .\n\nLuteolin ( 5-50 \u03bcmol/L ) suppressed the hypoxia-induced changes in the cells in a dose-dependent manner .\n\nHypoxia significantly decreased the expression of E-cadherin while increased the expression of N-cadherin in the cells ( indicating the occurrence of EMT-like transformation ) , which was reversed by luteolin ( 5 \u03bcmol/L ) .\n\nIn B16F10 cells , luteolin up-regulated E-cadherin at least partly via inhibiting the \u03b23 integrin/FAK signal pathway .\n\nIn experimental metastasis model mice , treatment with luteolin ( 10 or 20 mg/kg ) reduced metastatic colonization in the lungs by 50% .\n\nFurthermore , the treatment increased the expression of E-cadherin while reduced the expression of vimentin and \u03b23 integrin in the tumor tissues .\n\nCONCLUSION Luteolin inhibits the hypoxia-induced EMT in malignant melanoma cells both in vitro and in vivo via the regulation of \u03b23 integrin , suggesting that luteolin may be applied as a potential anticancer chemopreventative and chemotherapeutic agent .", "output": "Activating invasion and metastasis" }, { "input": "Exosomes are small-membrane vesicles secreted by hematopoietic and malignant epithelial cells as well as trophoblasts .\n\nThe composition of cancerous exosomes has been proven to play pivotal roles in the maintenance of the microenvironment that is beneficial for the progression of cancer , such as Fas-ligand-triggered lymphocyte apoptosis .\n\nWe supposed that the immunosuppressive effect of cancerous exosomes might be helpful in the treatment of diseases characterized by overactivation of the immune system and subsequent tissue injury .\n\nThe aim of this study was to evaluate the protective effect of tumor-derived exosomes in the mice model of lipopolysaccharide ( LPS)-induced inflammation .\n\nTetrazolium ( MTT ) and DNA electrophoresis were used to measure the cytotoxicity of exosomes on lymphocytes .\n\nPathologic observation of tissue sections , serologic analysis of aspartate aminotransferase/alanine aminotransferase ( AST/ALT ) , and urinary analysis of protein were used to assess the protection effect of exosomes in LPS-induced multiorgan damage .\n\nIn vitro outcomes of MTT and DNA electrophoresis showed the cytotoxicity of exosomes on lymphocytes .\n\nTogether with the alleviation of organ damages evaluated by urine protein , serum AST/ALT , and pathologic analysis , we confirmed the possibility that pretreatment of mice with exosomes , produced by H22 hepatic tumor cells , resulted in protection against LPS-induced tissue damage , which is caused by overactivation of the immune system and inflammation response .\n\nThis therapeutic strategy will raise an interesting way to search new therapeutics in pairs of diseases with complementarities in etiology and pathology , namely , a strategy of taking advantage of the mutual complementarities between diseases .", "output": "Tumor promoting inflammation" }, { "input": "p21 is a potent cyclin-dependent kinase inhibitor that plays a role in promoting G1 cell cycle arrest and cellular senescence .\n\nConsistent with this role , p21 is a downstream target of several tumour suppressors and oncogenes , and it is downregulated in the majority of tumours , including breast cancer .\n\nHere , we report that protein arginine methyltransferase 6 ( PRMT6 ) , a type I PRMT known to act as a transcriptional cofactor , directly represses the p21 promoter .\n\nPRMT6 knock-down ( KD ) results in a p21 derepression in breast cancer cells , which is p53-independent , and leads to cell cycle arrest , cellular senescence and reduced growth in soft agar assays and in severe combined immunodeficiency ( SCID ) mice for all the cancer lines examined .\n\nWe finally show that bypassing the p21-mediated arrest rescues PRMT6 KD cells from senescence , and it restores their ability to grow on soft agar .\n\nWe conclude that PRMT6 acts as an oncogene in breast cancer cells , promoting growth and preventing senescence , making it an attractive target for cancer therapy .", "output": "Sustaining proliferative signaling, Enabling replicative immortality" }, { "input": "The molecular mechanism mediating expression of senescent cell antigen-aggregated or cleaved band 3 and externalized phosphatidylserine ( PS ) on the surface of aged erythrocytes and their premature expression in certain anemias is not completely elucidated .\n\nThe erythrocytes with these surface modifications undergo macrophage-mediated phagocytosis .\n\nIn this study , the role of protein kinase C ( PKC ) isoforms in the expression of these surface modifications was investigated .\n\nInhibition of PKC \u03b1 by 30\u2009\u03bcM rottlerin ( R30 ) and 2.3\u2009nM G\ufffd 6976 caused expression of both the senescent cell marker-externalized PS measured by FACS analysis and aggregated band 3 detected by western blotting .\n\nIn contrast to this observation , but in keeping with literature , PKC activation by phorbol-12-myristate-13-acetate ( PMA ) also led to the expression of senescence markers .\n\nWe explain this antithesis by demonstrating that PMA-treated cells show reduction in the activity of PKC \u03b1 , thereby simulating inhibition .\n\nThe reduction in PKC \u03b1 activity may be attributed to the known downregulation of PMA-activated PKC \u03b1 , caused by its membrane translocation and proteolysis .\n\nWe demonstrate membrane translocation of PKC \u03b1 in PMA-treated cells to substantiate this inference .\n\nThus loss of PKC \u03b1 activity either by inhibition or downregulation can cause surface modifications which can trigger erythrophagocytosis .", "output": "Enabling replicative immortality" }, { "input": "Ovarian cancer-related angiogenesis is a complex process orchestrated by many positive and negative regulators .\n\nMany growth factors are involved in the development of the tumor-associated vasculature , and from these , endocrine gland-derived vascular endothelial growth factor ( EG-VEGF ) seems to play a crucial role .\n\nEG-VEGF is the first organ-specific angiogenic factor and its effects are restricted to the endothelial cells of the endocrine glands .\n\nAlthough EG-VEGF was detected in both normal and neoplastic ovaries , its clinical significance remains controversial .\n\nIn the present study , we analyzed 30 patients with epithelial ovarian cancer , and the immunohistochemical expression of EG-VEGF was compared with the conventional clinico-pathological parameters of prognosis .\n\nNeoplastic cells of the ovarian carcinoma expressed EG-VEGF in 73.33% of the cases , as a cytoplasmic granular product of reaction .\n\nWe found a strong correlation between the expression of EG-VEGF at protein level and tumor stage , grade , and microscopic type .\n\nThe expression of EG-VEGF was found in patients with stage III and IV , but not in stage II .\n\nThe majority of serous adenocarcinoma , half of the cases with clear cell carcinoma and two cases with endometrioid carcinoma showed definite expression in tumor cells .\n\nNo positive reaction was found in the cases with mucinous carcinoma .\n\nOur results showed that EG-VEGF expression is an indicator not only of the advanced stage , but also of ovarian cancer progression .\n\nBased on these data , we concluded that EG-VEGF expression in tumor cells of the epithelial ovarian cancer is a good marker of unfavorable prognosis and could be an attractive therapeutic target in patients with advanced-stage tumors , refractory conventional chemotherapy .", "output": "Inducing angiogenesis" }, { "input": "UNLABELLED Hepatocellular carcinoma ( HCC ) is a major liver malignancy .\n\nWe previously demonstrated that deregulation of epigenetic regulators is a common event in human HCC .\n\nSuppressor of variegation 3-9 homolog 1 ( SUV39H1 ) , the prototype of histone methyltransferase , is the major enzyme responsible for histone H3 lysine 9 trimethylation , which , essentially , is involved in heterochromatin formation , chromosome segregation , and mitotic progression .\n\nHowever , the implication of SUV39H1 in hepatocarcinogenesis remains elusive .\n\nIn this study , we found that SUV39H1 was frequently up-regulated in human HCCs and was significantly associated with increased Ki67 expression ( P < 0.001 ) and the presence of venous invasion ( P = 0.017 ) .\n\nTo investigate the role of SUV39H1 in HCC development , both gain- and loss-of-function models were established .\n\nSUV39H1 overexpression remarkably enhanced HCC cell clonogenicity , whereas knockdown of SUV39H1 substantially suppressed HCC cell proliferation and induced cell senescence .\n\nIn addition , ectopic expression of SUV39H1 increased the migratory ability of HCC cells , whereas a reduced migration rate was observed in SUV39H1 knockdown cells .\n\nThe significance of SUV39H1 in HCC was further demonstrated in a nude mice model ; SUV39H1 knockdown drastically inhibited in vivo tumorigenicity and abolished pulmonary metastasis of HCC cells .\n\nWe also identified microRNA-125b ( miR-125b ) as a post-transcriptional regulator of SUV39H1 .\n\nEctopic expression of miR-125b inhibited SUV39H1 3'-untranslated-region-coupled luciferase activity and suppressed endogenous SUV39H1 expression at both messenger RNA and protein levels .\n\nWe have previously reported frequent down-regulation of miR-125b in HCC .\n\nInterestingly , miR-125b level was found to be inversely correlated with SUV39H1 expression ( P = 0.001 ) in clinical specimens .\n\nOur observations suggested that miR-125b down-regulation may account for the aberrant SUV39H1 level in HCC .\n\nCONCLUSION Our study demonstrated that SUV39H1 up-regulation contributed to HCC development and metastasis .\n\nThe tumor-suppressive miR-125b served as a negative regulator of SUV39H1 .", "output": "Enabling replicative immortality, Activating invasion and metastasis" }, { "input": "Chronic inflammation is a risk factor for the development of colon cancer , providing genotoxic insults , growth and pro-angiogenic factors that can promote tumorigenesis and tumor growth .\n\nImmunomodulatory agents can interfere with the inflammation that feeds cancer , but their impact on the transformed cell is poorly understood .\n\nThe calcium/calcineurin signaling pathway , through activation of NFAT , is essential for effective immune responses , and its inhibitors cyclosporin A ( CsA ) and FK506 are used in the clinics to suppress immunity .\n\nMoreover , the kinases GSK3\u03b2 and mTOR , modulated by PI-3K/Akt , can inhibit NFAT activity , suggesting a cross-talk between the calcium and growth factor signaling pathways .\n\nBoth NFAT and mTOR activity have been associated with tumorigenesis .\n\nWe therefore investigated the impact of calcineurin and PI-3K/mTOR inhibition in growth of human colon carcinoma cells .\n\nWe show that despite the efficient inhibition of NFAT1 activity , FK506 promotes tumor growth , whereas CsA inhibits it due to a delay in cell cycle progression and induction of necroptosis .\n\nWe found NF\u03baB activation and mTORC1 activity not to be altered by CsA or FK506 .\n\nSimilarly , changes to mitochondrial homeostasis were equivalent upon treatment with these drugs .\n\nWe further show that , in our model , NFAT1 activation is not modulated by PI3K/mTOR .\n\nWe conclude that CsA slows cell cycle progression and induces necroptosis of human carcinoma cell lines in a TGF\u03b2- , NFAT- , NF\u03baB- and PI3K/mTOR-independent fashion .\n\nNevertheless , our data suggest that CsA , in addition to its anti-inflammatory capacity , may target transformed colon and esophagus carcinoma cells without affecting non-transformed cells , promoting beneficial tumoristatic effects .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Proline- , glutamic acid- and leucine-rich protein-1/modulator of non-genomic activity of estrogen receptor ( ER ) ( PELP1/MNAR ) is a novel nuclear receptor ( NR ) co-activator that plays an essential role in the actions of ER .\n\nEmerging findings suggest that PELP1/MNAR is a novel proto-oncogene , whose expression is deregulated in several hormone-responsive cancers , including endometrial cancer .\n\nIn this study , we demonstrate that PELP1/MNAR is widely expressed in endometrial carcinoma cell lines .\n\nTo investigate its possible role in endometrial carcinoma progression , we adopted an RNA interference technology to downregulate PELP1/MNAR expression in Ishikawa endometrial carcinoma cells .\n\nPELP1/MNAR downregulation substantially reduced cell proliferation , and the cells in which PELP1/MNAR expression was knocked down also exhibited a decreased migration and invasion ability , as shown by Boyden chamber and invasion assays .\n\nThe results showed that the expression of MMP-2 and MMP-9 was also decreased .\n\nThese results suggest that PELP1/MNAR plays a role in endometrial cancer progression and metastasis , and that PELP1/MNAR may be a potential therapeutic target for endometrial cancer .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "ABSTRACT : INTRODUCTION : Tumor cell migration and invasion are critical initiation steps in the process of breast cancer metastasis , the primary cause of breast cancer morbidity and death .\n\nHere we investigated the role of p21Cip1 ( p21 ) , a member of the core cell cycle machinery , in transforming growth factor-beta ( TGF\u03b2)-mediated breast cancer cell migration and invasion .\n\nMETHODS : A mammary fat pad xenograft mouse model was used to assess the mammary tumor growth and local invasion .\n\nThe triple negative human breast cancer cell lines MDA-MB231 and its sub-progenies SCP2 and SCP25 , SUM159PT , SUM149PT , SUM229PE and SUM1315MO2 were treated with 5 ng/ml TGF\u03b2 and the protein expression levels were measured by Western blot .\n\nCell migration and invasion were examined using the scratch/wound healing and Transwell assay .\n\nTGF\u03b2 transcriptional activity was measured by a TGF\u03b2/Smad reporter construct ( CAGA12-luc ) using luciferase assay. q-PCR was used for assessing TGF\u03b2 downstream target genes .\n\nThe interactions among p21 , p/CAF and Smad3 were performed by co-immunoprecipitation .\n\nIn addition , Smad3 on DNA binding ability was measured by DNA immunoprecipitation using biotinylated Smad binding element DNA probes .\n\nFinally , the association among active TGF\u03b2/Smad signaling , p21 and p/CAF with lymph node metastasis was examined by immunohistochemistry in tissue microarray containing 50 invasive ductal breast tumors , 25 of which are lymph node positive .\n\nRESULTS : We found p21 expression to correlate with poor overall and distant metastasis free survival in breast cancer patients .\n\nFurthermore , using xenograft animal models and in vitro studies , we found p21 to be essential for tumor cell invasion .\n\nThe invasive effects of p21 were found to correlate with Smad3 , and p/CAF interaction downstream of TGF\u03b2. p21 and p/CAF regulates TGF\u03b2-mediated transcription of pro-metastatic genes by controlling Smad3 acetylation , DNA binding and transcriptional activity .\n\nIn addition , we found that active TGF\u03b2/Smad signaling correlates with high p21 and p/CAF expression levels and lymph node involvement using tissue microarrays from breast cancer patients .\n\nCONCLUSIONS : Together these results highlight an important role for p21 and p/CAF in promoting breast cancer cell migration and invasion at the transcriptional level and may open new avenues for breast cancer therapy .", "output": "Activating invasion and metastasis" }, { "input": "OBJECTIVE To construct the eukaryotic expression vector for Max interacting protein 1 ( Mxi1 ) .\n\nMETHODS The full length cDNA of Mxi1 gene obtained from fetal lymphocyte and KG1 cells were inserted into plasmid pDs-red2-N1 respectively to generate pDs-red2-N1/Mri1 ( wild/mutation type ) .\n\nThen the recombinant vector was transfected into Cos-7 cells via liposome. 48 hours post transfection , mRNA of Mri1 gene was detected by RT-PCR and Mxi1 protein expression was detected by flow cytometry and fluorescence microscope in the Cos-7 cells .\n\nRESULTS The eukaryotic expression vector of Mxi1 was constructed and transfected into eukaryotic cells successfully .\n\nThe expression of red fluorescence protein in the transfected Cos-7 cells was observed under fluorescence microscope which implied the expression of Mxi1 .\n\nThe transfect efficiency of both wild and mutation type were in a high level in 3 days after transfected , which lasted to 6 d .\n\nRT-PCR amplified the total RNA extracted from the transfected Cos-7 cells could find increased mRNA level of Mxi1 gene .\n\nCONCLUSION We successfully constructed the eukaryote expression vector for Mri1 gene ; Cos-7 cells transfected with the vector via liposome could express Mxi1 protein .\n\nThese could be useful for the further study of the Myc gene modulation .", "output": "Genomic instability and mutation" }, { "input": "Macrophages are the prominent components of solid tumors and have complex dual functions in their interaction with cancer cells .\n\nStrong evidence suggests that TAM is a part of inflammatory circuits that promote tumor progression .\n\nB7-homologue 3 ( B7-H3 ) , a recently identified homologue of B7.1/2 ( CD80/86 ) , has been described to exert co-stimulatory and immune regulatory functions .\n\nHere , we showed that a fraction of macrophages in tumor stroma expressed surface B7-H3 molecule .\n\nNormal macrophages , which did not express B7-H3 , would be induced expressing B7-H3 molecule when culturing with tumor cell .\n\nAlthough a lung cancer cell line constitutively expressed B7-H3 mRNA and protein in plasma , primary tumor cell isolated from the transplanted lung carcinoma model expressed B7-H3 on the surface .\n\nInterestingly , in transplanted lung carcinoma model , the expression of membrane-bound B7-H3 in tumor cells was increased as prolonging of tumor transformation .\n\nIn support , IL-10 released from TAM could stimulate cancer cell expression of membrane bound B7-H3 .\n\nFurthermore , Lung cancer and TAM-related B7-H3 was identified as a strong inhibitor of T-cell effect and influenced the outcome of T cell immune response .\n\nIn conclusion , TAM-tumor cell interaction-induced membrane-bound B7-H3 represents a novel immune escape mechanism which links the pro-inflammatory response to immune tolerance in the tumor milieu .", "output": "Avoiding immune destruction" }, { "input": "TGF-\u03b2 derived from bone fuels melanoma bone metastases by inducing tumor secretion of prometastatic factors that act on bone cells to change the skeletal microenvironment .\n\nHalofuginone is a plant alkaloid derivative that blocks TGF-\u03b2 signaling with antiangiogenic and antiproliferative properties .\n\nHere , we show for the first time that halofuginone therapy decreases development and progression of bone metastasis caused by melanoma cells through the inhibition of TGF-\u03b2 signaling .\n\nHalofuginone treatment of human melanoma cells inhibited cell proliferation , phosphorylation of SMAD proteins in response to TGF-\u03b2 , and TGF-\u03b2-induced SMAD-driven transcription .\n\nIn addition , halofuginone reduced expression of TGF-\u03b2 target genes that enhance bone metastases , including PTHrP , CTGF , CXCR4 , and IL11 .\n\nAlso , cell apoptosis was increased in response to halofuginone .\n\nIn nude mice inoculated with 1205Lu melanoma cells , a preventive protocol with halofuginone inhibited bone metastasis .\n\nThe beneficial effects of halofuginone treatment were comparable with those observed with other anti-TGF-\u03b2 strategies , including systemic administration of SD208 , a small-molecule inhibitor of TGF-\u03b2 receptor I kinase , or forced overexpression of Smad7 , a negative regulator of TGF-\u03b2 signaling .\n\nFurthermore , mice with established bone metastases treated with halofuginone had significantly less osteolysis than mice receiving placebo assessed by radiography .\n\nThus , halofuginone is also effective in reducing the progression of melanoma bone metastases .\n\nMoreover , halofuginone treatment reduced melanoma metastasis to the brain , showing the potential of this novel treatment against cancer metastasis .\n\nCancer Res ; 72(23) ; 6247-56. \ufffd2012 AACR .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "AIM To assess the prognostic significance of nuclear factor-\u03baB ( NF-\u03baB ) and its target genes in gastric cancer .\n\nMETHODS The tumor tissues of 115 patients with gastric cancer were immunohistochemically evaluated using monoclonal antibodies against NF-\u03baB RelA .\n\nPreoperative serum levels of vascular endothelial growth factor ( VEGF ) , interleukin-6 ( IL-6 ) were assessed via enzyme-linked immuno-sorbent assay .\n\nC-reactive protein ( CRP ) and serum amyloid A ( SAA ) were measured via immunotrubidimetry .\n\nRESULTS Positive rate of NF-\u03baB RelA was 42.6% .\n\nNF-\u03baB RelA expression in tumor tissues was also related to serum levels of IL-6 ( P = 0.044 ) and CRP ( P = 0.010 ) .\n\nIL-6 , SAA , CRP were related to depth of invasion , VEGF and SAA were correlated with lymph node metastasis .\n\nIL-6 , VEGF , SAA and CRP were related to the stage .\n\nUnivariate analysis demonstrated that immunostaining of NF-\u03baB RelA , levels of IL-6 , VEGF , SAA were significantly related with both disease free survival and overall survival ( OS ) .\n\nMultivariate analysis verified that NF-\u03baB RelA [ hazard ratio ( HR ) : 3.40 , P = 0.024 ] and SAA ( HR : 3.39 , P = 0.045 ) were independently associated with OS .\n\nCONCLUSION Increased expression of NF-\u03baB RelA and high levels of serum SAA were associated with poor OS in gastric cancer patients .", "output": "Activating invasion and metastasis" }, { "input": "The suppression of MAPK and oxidative stress could be an important anti-inflammatory mechanism .\n\nFucoidan regulates MAPK activity in several cell lines .\n\nHowever , the mechanism for the anti-inflammatory and oxidative stress effect of low molecular weight fucoidan ( LMWF ) is poorly understood in RAW264.7 cells .\n\nThe objective of this study was to examine the critical role of LMWF during LPS-induced inflammation in RAW264.7 cells .\n\nTo determine the potential role of LMWF , we analysed pro-inflammatory cytokine , transcription factor , inflammation-related and oxidative stress related protein expression in vitro during LPS-induced inflammation in RAW264.7 cells .\n\nIn this study , we demonstrated the anti-inflammatory effects of LMWF on LPS-induced inflammation in macrophages through the regulation of signalling pathways , including its effect on the attenuation of inflammatory cytokines , such as IL-1\u03b2 , IL-1 and TNF-\u03b1 , and the degradation of phosphorylated p38 MAPK , ERK1/2 and JNK .\n\nThis study also demonstrates that LMWF might block NO as well as the expression of reactive oxygen species ( ROS ) , which subsequently inhibits the iNOS and COX-2 expression induced by LPS .\n\nBased on these findings , we suggest that LMWF might have great potential as an external pathogen prevention and intervention agent for inflammatory diseases .", "output": "Tumor promoting inflammation" }, { "input": "Nutrient deprivation based on the loss of essential amino acids by catabolic enzymes in the microenvironment is a critical means to control inflammatory responses and immune tolerance .\n\nHere we report the novel finding that Tph-1 ( tryptophan hydroxylase-1 ) , a synthase which catalyses the conversion of tryptophan to serotonin and exhausts tryptophan , is a potent regulator of immunity .\n\nIn models of skin allograft tolerance , tumor growth , and experimental autoimmune encephalomyelitis , Tph-1 deficiency breaks allograft tolerance , induces tumor remission , and intensifies neuroinflammation , respectively .\n\nAll of these effects of Tph-1 deficiency are independent of its downstream product serotonin .\n\nBecause mast cells ( MCs ) appear to be the major source of Tph-1 and restoration of Tph-1 in the MC compartment in vivo compensates for the defect , these experiments introduce a fundamentally new mechanism of MC-mediated immune suppression that broadly impacts multiple arms of immunity .", "output": "Avoiding immune destruction" }, { "input": "OBJECTIVES Human neutrophil proteins-1 , -2 , and -3 ( HNPs -1 , -2 , and -3 ) are expressed in several tumor types .\n\nHowever , the role of HNPs 1-3 in human bladder cancer has not yet been determined .\n\nWe investigated the association between the plasma levels of HNPs 1-3 and clinicopathological parameters in bladder cancer patients .\n\nDESIGN AND METHODS The plasma levels of HNPs 1-3 were measured in 60 patients with bladder cancer and in 58 healthy controls .\n\nThe plasma levels of HNPs 1-3 were determined by a solid-phase enzyme-linked immunosorbent assay ( ELISA ) .\n\nPlasma samples were obtained before surgery .\n\nPlasma samples were permitted to clot and were then stored at -80 \ufffdC until use .\n\nRESULTS The levels of the HNPs increased from grade 1 to 4 tumors and this difference was statistically significant ( p < 0.001 ) .\n\nAdditionally , plasma HNP levels were significantly higher in patients with metastatic bladder cancer and in patients with lymphovascular involvement , metastasis of the lymph nodes , and increased tumor burden ( p < 0.001 ) .\n\nCONCLUSIONS The preoperative plasma levels of HNPs 1-3 paralleled the progression and pathological stages of the malignancies .\n\nThis study suggests that HNPs 1-3 promote tumor invasion and are potential indicators of disease progression in patients with bladder cancer .", "output": "Activating invasion and metastasis" }, { "input": "This study is to investigate the effects of bortezomib on the angiogenesis of mesenchymal stem cells ( MSCs ) .\n\nWe examined the effects of bortezomib on the cellular proliferation , migration , and capillary network formation of HUVECs cocultured with CMs of MSCs .\n\nWe found that Bortezomib inhibited the cellular proliferation and tube formation of HUVECs cocultured with CMs in a dose-dependent fashion .\n\nBortezomib also prevented the migration of HUVECs cocultured with CMs .\n\nIn addition , bortezomib dose-dependently inhibited the growth of MSCs and prevented the production of angiogenic factors including VEGF ( vascular endothelial growth factor ) , HGF ( hepatocyte growth factor ) , and bFGF ( basic fibroblast growth factor ) in MSCs .\n\nIn conclusion , bortezomib prevented the angiogenesis mediated by MSCs .", "output": "Inducing angiogenesis" }, { "input": "Retinoic acid ( RA)-induced differentiation therapy is partially successful in neuroblastoma treatment .\n\nWe found that a novel combination of vanadium-based PTP inhibitors with RA induced extensive differentiation in neuroblastoma cells .\n\nIn contrast to RA alone , this led to either permanent differentiation or senescence after 14days of combined treatment followed by chemical removal .\n\nSenescence was dependent in part on synergistic AKT and ERK activation. p21 was also strongly induced , but in contrast to oncogene-induced senescence , p53 was not activated .\n\nVanadium-based inhibitors thus serve strongly to enhance RA's ability to drive differentiation and a novel form of senescence in neuroblastoma cells .", "output": "Enabling replicative immortality" }, { "input": "Formononetin is a novel herbal isoflavonoid isolated from Astragalus membranaceus , a medicinal plant that possesses antitumorigenic properties .\n\nOur previous findings demonstrated that formononetin initiates growth-inhibitory and pro-apoptotic activities in human colon cancer cells .\n\nIn the present study , we aimed to further examine the potential of formononetin in controlling angiogenesis and tumor cell invasiveness in human colon cancer cells and tumor xenografts .\n\nThe results showed that formononetin downregulated the expression of the key pro-angiogenic factors , including vascular endothelial growth factor ( VEGF ) and matrix metalloproteinases .\n\nWe also discovered that the invasiveness of metastatic colon cancer cells was alleviated following drug treatment .\n\nThe potential anti-angiogenic effect of formononetin was examined in nude mouse xenografts .\n\nThe tumor size and the number of proliferating cells were reduced in the tumor tissues obtained from the formononetin-treated group .\n\nThe serum VEGF level was also reduced in the drug-treated animals when compared to the controls .\n\nThese findings suggest that formononetin inhibits angiogenesis and tumor cell invasion , and thus support its use in the treatment of advanced and metastatic colon cancers .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Chronic inflammation is a critical component in breast cancer progression .\n\nPro-inflammatory mediators along with growth/survival factors within the tumor microenvironment potentiate the expression of pro-inflammatory cytokines ( IL-1 , IL-6 , TNF-\u03b1 ) , chemotactic cytokines and their receptors ( CXCR4 , CXCL12 , CXCL8 ) and angiogenic factors ( VEGF ) that often overcome the effect of anti-inflammatory molecules ( IL-4 , IL-10 ) thus evading the host's antitumor immunity .\n\nDetailed knowledge , therefore , of the regulatory mechanisms determining cytokine levels is essential to understand the pathogenesis of breast cancer .\n\nHIF-1\u03b1 and NF-\u03baB transcription factors are important players for the establishment of a pro-inflammatory and potentially oncogenic environment .\n\nHIF-1\u03b1 is the key mediator of the cellular response to oxygen deprivation and induces the expression of genes involved in survival and angiogenesis within solid hypoxic tumors .\n\nThe expression of these genes is often modulated by the p53 tumor suppressor protein that induces apoptosis or cell cycle arrest in neoplastic cells .\n\nFunctional crosstalk between HIF-1\u03b1 and p53 pathways mediated by modulators shared between the two transcription factors such as SRC-1 and SIRT-1 differentially regulate the expression of distinct subsets of their target genes under variable stress conditions .\n\nIn an attempt to shed light on the complex regulatory mechanisms involved in cancer-related inflammation , we investigated the role of the two common p53 and HIF-1\u03b1 co-regulators SRC-1 and SIRT-1 , in the expression of the highly potent metastatic chemokine receptor CXCR4 .\n\nBoth SRC-1 and SIRT-1 overexpression in DSFX-treated MCF-7 cells reduced CXCR4 cellular levels implying that both co-regulators are crucial factors in the determination of the metastatic potential of breast cancer cells .", "output": "Activating invasion and metastasis" }, { "input": "Given the important role of CXCR4 in cancer metastasis , microenvironmental factors that modulate CXCR4 may have an impact on the process of tumor expansion .\n\nHypoxia is a common feature of solid tumors and a significant microenvironmental factor that drives aggressive behavior .\n\nCXCR4 is upregulated in several cancer cells under hypoxic conditions , suggesting a relationship between tumor hypoxia and CXCR4 .\n\nHowever , the role of hypoxia in regulating CXCR4 in gastric cancer remains poorly understood .\n\nKATO III gastric cancer cells were exposed to hypoxia or normoxia .\n\nCXCR4 expression in cells transfected with shRNA specific for HIF-1\u03b1 was investigated by western blotting and flow cytometry .\n\nWound healing , migration and invasion assays were used to assess cell motility and the chemotactic response to CXCL12 , a major CXCR4 ligand .\n\nCXCR4 expression at the protein level and in the cell membrane was significantly increased in KATOIII cells following exposure to hypoxia .\n\nThis upregulation of CXCR4 was implicated in increased cell motility and enhanced chemotactic responses ( migration and invasion ) to CXCL12 treatment in vitro .\n\nThe increases in CXCR4 expression and metastatic potential in gastric cancer cells exposed to hypoxia were blocked by HIF-1\u03b1-specific shRNA .\n\nOur results indicate that hypoxia upregulates CXCR4 in gastric cancer cells in a HIF-1\u03b1-dependent manner , and that upregulation of CXCR4 plays a role in cancer cell migration and invasion .\n\nThus , disrupting the hypoxia-HIF-1\u03b1-CXCR4 axis could be an attractive therapeutic strategy for the treatment of gastric cancer .", "output": "Activating invasion and metastasis" }, { "input": "Advanced or metastatic prostate cancer is treated by androgen deprivation ; however , patients inevitably relapse with castration-resistant prostate cancer ( CRPC ) .\n\nCRPC remains dependent on androgen receptor ( AR ) signaling , which may include constitutive , ligand-independent action of naturally occurring AR splice variants .\n\nFor example , the AR splice variant AR3 ( also termed AR-V7 ) is expressed in CRPC and is linked to poor prognosis .\n\nVav3 , a Rho GTPase guanine nucleotide exchange factor , is an AR coactivator that is up-regulated in human prostate cancer compared with benign tissue and in preclinical models of CRPC .\n\nVav3 confers castration-resistant growth to androgen-dependent human prostate cancer cells .\n\nDespite the importance of AR coactivators in promoting CRPC , the potential role of these regulatory proteins in modulating AR splice variant activity is unknown .\n\nWe examined the contributions of Vav3 to AR activity in two CRPC cell lines that naturally express relatively high levels of Vav3 and AR3 .\n\nVav3 or AR3 knockdown greatly attenuated cell proliferation , soft agar growth , and ligand-independent AR activity .\n\nVav3 potently enhanced the transcriptional activity of AR3 and another clinically relevant AR splice variant , ARv567es .\n\nVav3 knockdown resulted in lowered nuclear AR3 levels , whereas total AR3 levels remained similar .\n\nConversely , overexpression of Vav3 resulted in increased nuclear AR3 .\n\nCoimmunoprecipitation revealed that AR3 and Vav3 interact .\n\nThese novel data demonstrating physical and functional interactions between Vav3 , a unique AR coactivator , and an AR splice variant provide insights into the mechanisms by which Vav3 exploits and enhances AR signaling in the progression to CRPC .", "output": "Sustaining proliferative signaling" }, { "input": "BACKGROUND Angiogenic factors following oncological surgery is important in tumor recurrence .\n\nVascular endothelial growth factor ( VEGF ) , angiopoietin 1 ( Ang-1 ) , Ang-2 , soluble VEGF-receptor 1 ( sVEGFR1 ) and sVEGFR2 may influence angiogenesis .\n\nThis prospective study examined the influence of open and video-assisted thoracic surgery ( VATS ) lung resections for early stage non-small cell lung cancer ( NSCLC ) on postoperative circulating angiogenic factors .\n\nMETHODS Forty-three consecutive patients underwent major lung resection through either VATS ( n = 23 ) or Open thoracotomy ( n = 20 ) over an 8-month period .\n\nBlood samples were collected preoperatively and postoperatively on days ( POD ) 1 and 3 for enzyme linked immunosorbent assay determination of angiogenic factors .\n\nRESULTS Patient demographics were comparable .\n\nFor all patients undergoing major lung resection , postoperative Ang-1 and sVEGFR2 levels were significantly decreased , while Ang-2 and sVEGFR1 levels markedly increased .\n\nNo significant peri-operative changes in VEGF levels were observed .\n\nCompared with open group , VATS had significantly lower plasma levels of VEGF ( VATS 170 \u00b1 93 pg/mL ; Open 486 \u00b1 641 pg/mL ; P = 0.04 ) and Ang-2 ( VATS 2484 \u00b1 1119 pg/mL ; Open 3379 \u00b1 1287 pg/mL ; P = 0.026 ) on POD3 .\n\nCONCLUSIONS Major lung resection for early stage NSCLC leads to a pro-angiogenic status , with increased Ang-2 and decreased Ang-1 productions .\n\nVATS is associated with an attenuated angiogenic response with lower circulating VEGF and Ang-2 levels compared with open .\n\nSuch differences in angiogenic factors may be important in lung cancer biology and recurrence following surgery .", "output": "Inducing angiogenesis" }, { "input": "Human breast tumors are infiltrated by memory CD4(+) T cells along with increased numbers of regulatory T cells ( Treg ) and plasmacytoid dendritic cells ( pDC ) that facilitate immune escape and correlate with poor prognosis .\n\nHere , we report that inducible costimulatory molecule ( ICOS ) , a T cell costimulatory molecule of the CTLA4/PD1/CD28 family , is expressed mostly by tumor-associated Treg in primary breast tumors .\n\nA large proportion of these ICOS(+) Treg were Ki67(+) and this evident proliferative expansion was found to rely on interactions with tumor-associated pDC .\n\nIndeed , tumor-associated Treg highly expanded in presence of pDC but failed to proliferate under CD3/CD28 signal .\n\nIn vitro experiments revealed that the addition of a neutralizing anti-ICOS antibody blocked pDC-induced Treg expansion and interleukin-10 secretion by memory CD4(+) T cells , establishing a pivotal role for ICOS in this process .\n\nSupporting these findings , the presence of ICOS(+) cells in clinical specimens of breast cancer correlated with a poor prognosis .\n\nTogether , our results highlight an important relationship between Treg and pDC in breast tumors , and show that ICOS/ICOS-L interaction is a central event in immunosuppression of tumor-associated memory CD4(+) T cells .\n\nThese findings strongly rationalize antibody-mediated ICOS blockade as a powerful clinical strategy to correct immune escape and promote therapeutic responses in breast cancer .\n\nCancer Res ; 72(23) ; 6130-41. \ufffd2012 AACR .", "output": "Avoiding immune destruction" }, { "input": "Tumor cells trigger angiogenesis through overexpression of various angiogenic factors including vascular endothelial growth factor ( VEGF ) and angiopoietin 1 ( Ang1 ) .\n\nTherefore , inhibition of the expression of both VEGF and Ang1 , the initial step of tumor angiogenesis , is a promising strategy for cancer chemoprevention and therapy .\n\nGrape seed proanthocyanidins ( GSPs ) are widely consumed dietary supplements that have antitumor activity .\n\nDue to their polymeric structure , GSPs are poorly absorbed along the gastrointestinal tract and can reach the colon at high concentrations , allowing these chemicals to act as chemopreventive agents for colon cancer .\n\nIn the present study , we found that GSPs inhibited colon tumor-induced angiogenesis and , thus , the growth of colon tumor xenografts on the chick chorioallantoic membranes .\n\nThe mechanisms of their action were related to inhibiting the expression of both VEGF and Ang1 through scavenging reactive oxygen species .\n\nPrevious studies have demonstrated that the chemopreventive effects of GSPs on colon cancer are associated with their growth inhibitory and apoptosis-inducing effects .\n\nOur results demonstrate another mechanism by which GSPs inhibit colon tumor growth , which will be helpful for developing GSPs as a pharmacologically safe angiopreventive agent against colorectal cancer .", "output": "Inducing angiogenesis, Tumor promoting inflammation, Resisting cell death" }, { "input": "Protein tyrosine kinase 6 ( PTK6 ) is a non-receptor tyrosine kinase expressed in epithelial cancers .\n\nDisruption of Ptk6 decreases azoxymethane-induced colon tumorigenesis in mice by preventing signal transducer and activator of transcription 3 activation .\n\nRelocalization of PTK6 in prostate cancers contributes to increased growth .\n\nAlthough not expressed in normal breast or ovary , PTK6 promotes anchorage-independent survival of breast and ovarian tumor cells .\n\nWe identified several potential PTK6 substrates in the human SW620 colon cancer cell line using mass spectrometry , including FAK ( focal adhesion kinase ) .\n\nWe show that FAK is a direct substrate of PTK6 in vitro and in vivo .\n\nExpression of membrane-targeted active PTK6 ( Palm-PTK6-YF ) induces constitutive activation of FAK and cell morphology changes , which are independent of SRC family kinases in Src-/- , Yes-/- , Fyn-/- ( SYF ) mouse embryonic fibroblasts ( MEFs ) .\n\nPalm-PTK6-YF expressing SYF cells are transformed and overcome contact inhibition , form colonies in transformation assays , proliferate in suspension and form tumors in a xenograft model .\n\nExpression of FAK and Palm-PTK6-YF in Fak-/- MEFs synergistically activates AKT and protects cells against anoikis .\n\nHowever , expression of Palm-PTK6-YF in Akt1/2-/- MEFs fails to protect cells from anoikis , indicating AKT is critical in PTK6 and FAK-mediated survival signaling .\n\nIn a conditional Pten knockout murine prostate cancer model , we identify prostate epithelial cells with enhanced activation of endogenous PTK6 and FAK at the plasma membrane .\n\nKnockdown of PTK6 in the PC3 human prostate cancer cell line disrupts FAK and AKT activation and promotes anoikis , which can be rescued by exogenous expression of FAK .\n\nOur data reveal important roles for a PTK6-FAK-AKT signaling axis in promoting anchorage-independent cell survival .", "output": "Evading growth suppressors" }, { "input": "The complement system contributes to various immune and inflammatory diseases , including cancer .\n\nIn this study , we investigated the capacity of lung cancer cells to activate complement and characterized the consequences of complement activation on tumor progression .\n\nWe focused our study on the production and role of the anaphylatoxin C5a , a potent immune mediator generated after complement activation .\n\nWe first measured the capacity of lung cancer cell lines to deposit C5 and release C5a .\n\nC5 deposition , after incubation with normal human serum , was higher in lung cancer cell lines than in nonmalignant bronchial epithelial cells .\n\nNotably , lung malignant cells produced complement C5a even in the absence of serum .\n\nWe also found a significant increase of C5a in plasma from patients with non-small cell lung cancer , suggesting that the local production of C5a is followed by its systemic diffusion .\n\nThe contribution of C5a to lung cancer growth in vivo was evaluated in the Lewis lung cancer model .\n\nSyngeneic tumors of 3LL cells grew slower in mice treated with an antagonist of the C5a receptor .\n\nC5a did not modify 3LL cell proliferation in vitro but induced endothelial cell chemotaxis and blood-vessels formation .\n\nC5a also contributed to the immunosuppressive microenvironment required for tumor growth .\n\nIn particular , blockade of C5a receptor significantly reduced myeloid-derived suppressor cells and immunomodulators ARG1 , CTLA-4 , IL-6 , IL-10 , LAG3 , and PDL1 ( B7H1 ) .\n\nIn conclusion , lung cancer cells have the capacity to generate C5a , a molecule that creates a favorable tumor microenvironment for lung cancer progression .", "output": "Inducing angiogenesis, Avoiding immune destruction" }, { "input": "Metastasis is a complex process utilizing both tumor-cell-autonomous properties and host-derived factors , including cellular immunity .\n\nWe have previously shown that germline polymorphisms can modify tumor cell metastatic capabilities through cell-autonomous mechanisms .\n\nHowever , how metastasis susceptibility genes interact with the tumor stroma is incompletely understood .\n\nHere , we employ a complex genetic screen to identify Cadm1 as a novel modifier of metastasis .\n\nWe demonstrate that Cadm1 can specifically suppress metastasis without affecting primary tumor growth .\n\nUnexpectedly , Cadm1 did not alter tumor-cell-autonomous properties such as proliferation or invasion , but required the host's adaptive immune system to affect metastasis .\n\nThe metastasis-suppressing effect of Cadm1 was lost in mice lacking T cell-mediated immunity , which was partially phenocopied by depleting CD8(+) T cells in immune-competent mice .\n\nOur data show a novel function for Cadm1 in suppressing metastasis by sensitizing tumor cells to immune surveillance mechanisms , and this is the first report of a heritable metastasis susceptibility gene engaging tumor non-autonomous factors .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "It is known that sperm samples from recurrent pregnancy loss ( RPL ) couples have an increase in their sperm DNA fragmentation ( SDF ) , but no studies have been performed in order to identify differences between single stranded SDF ( ssSDF ) and double stranded SDF ( dsSDF ) in these patients .\n\nThis could be relevant because the type of DNA damage could have different effects .\n\nSemen samples were classified attending their clinical status : 25 fertile donors and 20 RPL patients with at least two unexplained first trimester miscarriages .\n\nSDF was analysed using alkaline and neutral Comet assay , SCD test and pulsed-field gel electrophoresis ( PFGE ) , and ROC analysis including data from 105 more infertile patients ( n\\u200a=\\u200a150 ) was performed to establish predictive threshold values .\n\nSDF for alkaline and neutral Comet , and the SCD test was analysed in these categories of individuals .\n\nData revealed the presence of two subgroups within fertile donors .\n\nThe values obtained were 21.10\u00b19.13 , 23.35\u00b110.45 and 12.31\u00b14.31 , respectively , for fertile donors with low values for both ssSDF and dsSDF ; 27.86\u00b112.64 , 80.69\u00b112.67 and 12.43\u00b15.22 , for fertile donors with low ssSDF and high dsSDF ; and 33.61\u00b115.50 , 84.64\u00b111.28 and 19.28\u00b16.05 , for unexplained RPL patients , also showing a low ssSDF and high dsSDF profile .\n\nThis latter profile was seen in 85% of unexplained RPL and 33% of fertile donors , suggesting that it may be associated to a male risk factor for undergoing RPL .\n\nROC analysis regarding recurrent miscarriage set the cut-off value at 77.50% of dsDNA SDF .\n\nPFGE for low ssSDF and high dsSDF profile samples and positive controls treated with DNase , to induce dsDNA breaks , showed a more intense band of about 48 kb , which fits the toroid model of DNA compaction in sperm , pointing out that some nuclease activity may be affecting their sperm DNA in RPL patients .\n\nThis work identifies a very specific SDF profile related to the paternal risk of having RPL .", "output": "Genomic instability and mutation" }, { "input": "Perlecan Domain V ( DV ) promotes brain angiogenesis by inducing VEGF release from brain endothelial cells ( BECs ) following stroke .\n\nIn this study , we define the specific mechanism of DV interaction with the \u03b1(5)\u03b2(1) integrin , identify the downstream signal transduction pathway , and further investigate the functional significance of resultant VEGF release .\n\nInterestingly , we found that the LG3 portion of DV , which has been suggested to possess most of DV's angio-modulatory activity outside of the brain , binds poorly to \u03b1(5)\u03b2(1) and induces less BEC proliferation compared to full length DV .\n\nAdditionally , we implicate DV's DGR sequence as an important element for the interaction of DV with \u03b1(5)\u03b2(1) .\n\nFurthermore , we investigated the importance of AKT and ERK signaling in DV-induced VEGF expression and secretion .\n\nWe show that DV increases the phosphorylation of ERK , which leads to subsequent activation and stabilization of eIF4E and HIF-1\u03b1 .\n\nInhibition of ERK activity by U0126 suppressed DV-induced expression and secretion of VEGR in BECs .\n\nWhile DV was capable of phosphorylating AKT we show that AKT phosphorylation does not play a role in DV's induction of VEGF expression or secretion using two separate inhibitors , LY294002 and Akt IV .\n\nLastly , we demonstrate that VEGF activity is critical for DV increases in BEC proliferation , as well as angiogenesis in a BEC-neuronal co-culture system .\n\nCollectively , our findings expand our understanding of DV's mechanism of action on BECs , and further support its potential as a novel stroke therapy .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Although tumor-associated macrophages ( TAMs ) are involved in tumor growth and metastasis , the mechanisms controlling their pro-tumoral activities remain largely unknown .\n\nThe transcription factor c-MYC has been recently shown to regulate in vitro human macrophage polarization and be expressed in macrophages infiltrating human tumors .\n\nIn this study , we exploited the predominant expression of LysM in myeloid cells to generate c-Myc(fl/fl) LysM(cre/+) mice , which lack c-Myc in macrophages , to investigate the role of macrophage c-MYC expression in cancer .\n\nUnder steady-state conditions , immune system parameters in c-Myc(fl/fl) LysM(cre/+) mice appeared normal , including the abundance of different subsets of bone marrow hematopoietic stem cells , precursors and circulating cells , macrophage density , and immune organ structure .\n\nIn a model of melanoma , however , TAMs lacking c-Myc displayed a delay in maturation and showed an attenuation of pro-tumoral functions ( e.g. , reduced expression of VEGF , MMP9 , and HIF1\u03b1 ) that was associated with impaired tissue remodeling and angiogenesis and limited tumor growth in c-Myc(fl/fl) LysM(cre/+) mice .\n\nMacrophage c-Myc deletion also diminished fibrosarcoma growth .\n\nThese data identify c-Myc as a positive regulator of the pro-tumoral program of TAMs and suggest c-Myc inactivation as an attractive target for anti-cancer therapy .", "output": "Avoiding immune destruction" }, { "input": "BACKGROUND Cellular stress responses trigger signaling cascades that inhibit proliferation and protein translation to help alleviate the stress or if the stress cannot be overcome induce apoptosis .\n\nIn recent studies , we demonstrated the ability of lovastatin , an inhibitor of mevalonate synthesis , to induce the Integrated Stress Response as well as inhibiting epidermal growth factor receptor ( EGFR ) activation .\n\nMETHODOLOGY/PRINCIPAL FINDINGS In this study , we evaluated the effects of lovastatin on the activity of the LKB1/AMPK pathway that is activated upon cellular energy shortage and can interact with the above pathways .\n\nIn the squamous cell carcinoma ( SCC ) cell lines SCC9 and SCC25 , lovastatin treatment ( 1-25 \ufffdM , 24 hrs ) induced LKB1 and AMPK activation similar to metformin ( 1-10 mM , 24 hrs ) , a known inducer of this pathway .\n\nLovastatin treatment impaired mitochondrial function and also decreased cellular ADP/ATP ratios , common triggers of LKB1/AMPK activation .\n\nThe cytotoxic effects of lovastatin were attenuated in LKB1 null MEFs indicating a role for this pathway in regulating lovastatin-induced cytotoxicity .\n\nOf clinical relevance , lovastatin induces synergistic cytotoxicity in combination with the EGFR inhibitor gefitinib .\n\nIn LKB1 deficient ( A549 , HeLa ) and expressing ( SCC9 , SCC25 ) cell lines , metformin enhanced gefitinib cytotoxicity only in LKB1 expressing cell lines while both groups showed synergistic cytotoxic effects with lovastatin treatments .\n\nFurthermore , the combination of lovastatin with gefitinib induced a potent apoptotic response without significant induction of autophagy that is often induced during metabolic stress inhibiting cell death .\n\nCONCLUSION/SIGNIFICANCE Thus , targeting multiple metabolic stress pathways including the LKB1/AMPK pathway enhances lovastatin's ability to synergize with gefitinib in SCC cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Complex interactions between effector T cells and Foxp3(+) regulatory T cells ( Treg ) contribute to clinical outcomes in cancer , and autoimmune and infectious diseases .\n\nPrevious work showed that IL-12 reversed Treg-mediated suppression of CD4(+)Foxp3(-) T cell ( Tconv ) proliferation .\n\nWe and others have also shown that Tregs express T-bet and IFN-\u03b3 at sites of Th1 inflammation and that IL-12 induces IFN-\u03b3 production by Tregs in vitro .\n\nTo investigate whether loss of immunosuppression occurs when IFN-\u03b3 is expressed by Tregs we treated mouse lymphocyte cultures with IL-12 .\n\nIFN-\u03b3 expression did not decrease the ability of Tregs to suppress Tconv proliferation .\n\nRather , IL-12 treatment decreased Treg frequency and Foxp3 levels in Tregs .\n\nWe further showed that IL-12 increased IL-2R expression on Tconv and CD8 T cells , diminished its expression on Tregs and decreased IL-2 production by Tconv and CD8 T cells .\n\nTogether , these IL-12 mediated changes favored the outgrowth of non-Tregs .\n\nAdditionally , we showed that treatment with a second cytokine , IL-27 , decreased IL-2 expression without augmenting Tconv and CD8 T cell proliferation .\n\nNotably , IL-27 only slightly modified levels of IL-2R on non-Treg T cells .\n\nTogether , these results show that IL-12 has multiple effects that modify the balance between Tregs and non-Tregs and support an important role for relative levels of IL-2R but not for IFN-\u03b3 expression in IL-12-mediated reversal of Treg immunosuppression .", "output": "Avoiding immune destruction" }, { "input": "The SR/CR mouse phenotype , first described in 1999 in BALB/c and later bred into C57BL/6 mice , is resistant to cancer formation following high doses of cancer cells administered intraperitoneally .\n\nThe tumor cell targeting and destruction mechanisms have not been identified .\n\nBy fluorescence-activated cell sorting analysis , the immune response of SR/CR mice after intraperitoneal injection of cancer cells was investigated and compared with parent strain mice .\n\nA massive influx of leukocytes into the peritoneal cavity was found .\n\nA large fraction of these leukocytes were polymorphonuclear granulocytes , macrophages and natural killer cells .\n\nA relative decrease in influx of B-cells compared with controls was demonstrated .\n\nIncreased proportions of leukocytes belonging to the innate immune system were also demonstrated in splenocytes of SR/CR mice .\n\nCytospins of peritoneal fluid from SR/CR mice after cancer cell injection showed formations of immune cells morphologically resembling polymorphonuclear granulocytes and macrophages adjoining the cancer cells .\n\nThe results point to the potential involvement of innate immune cells in cancer immunology .\n\nOur data support migration of polymorphonuclear granulocytes , macrophages and NK cells into the peritoneum of the SR/CR mouse in response to intraperitoneal injection of S180 cancer cells .\n\nThe cell composition of spleens of SR/CR mice reflected the differential regulation of the innate immune cells in peritoneal exudates .\n\nBoth peritoneal exudates and the spleens of SR/CR mice contained decreased proportions of B-cells compared with BALB/c and C57BL/6 mice .\n\nWe reproduce important aspects of previous published data and further extend them by showing differentially regulated populations of splenocytes including B-lymphocytes in SR/CR mice compared with parent strain controls .\n\nImportantly , this differentially regulated immune response of SR/CR mice could not be found in response to challenge with the lymphoma cell line EL-4 .", "output": "Avoiding immune destruction" }, { "input": "In patients with advanced bladder cancer , glucocorticoids are frequently given to reduce acute toxicity , particularly hyperemesis , during chemotherapy , as well as to improve cachectic conditions .\n\nHowever , it remains unclear whether glucocorticoids directly affect the development and progression of bladder cancer through the glucocorticoid receptor pathway .\n\nGlucocorticoid receptor expression was first investigated in human bladder cancer lines and tissue microarrays .\n\nThen , the effects of dexamethasone on glucocorticoid receptor transcription , cell proliferation , apoptosis/cell cycle , and invasion were examined in bladder cancer lines .\n\nFinally , mouse xenograft models for bladder cancer were used to assess the efficacy of dexamethasone on tumor progression .\n\nAll the cell lines and tissues examined were found to express glucocorticoid receptor .\n\nDexamethasone increased glucocorticoid receptor-mediated reporter activity and cell proliferation , and inhibited apoptosis in the presence or absence of cisplatin .\n\nIn contrast , dexamethasone suppressed cell invasion , the expression of its related genes [ MMP-2/MMP-9 , interleukin ( IL)-6 , VEGF ] , and the activity of MMP-2/MMP-9 , and also induced mesenchymal-to-epithelial transition .\n\nIn addition , dexamethasone increased I\u03baB\u03b1 protein levels and cytosolic accumulation of NF-\u03baB .\n\nIn xenograft-bearing mice , dexamethasone slightly augmented the growth of the inoculated tumors but completely prevented the development of bloody ascites , suggestive of peritoneal dissemination of tumor cells , and actual metastasis .\n\nIn all these assays , dexamethasone effects were abolished by a glucocorticoid receptor antagonist or glucocorticoid receptor knockdown via RNA interference .\n\nThus , glucocorticoid receptor activation resulted in promotion of cell proliferation via inhibiting apoptosis yet repression of cell invasion and metastasis .\n\nThese results may provide a basis of developing improved chemotherapy regimens , including or excluding glucocorticoid receptor agonists/antagonists , for urothelial carcinoma .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Resisting cell death" }, { "input": "The goal of the present study was to examine hepatic differential gene expression patterns in Fisher-344 rats in response to dietary 2-aminoanthracene ( 2AA ) ingestion for 14 and 28 days .\n\nTwenty four post-weaning 3-4 week old F-344 male rats were exposed to 0 mgkg(-1)-diet ( control ) , 50 mgkg(-1)-diet ( low dose ) , 75 mgkg(-1)-diet ( medium dose ) and 100 mgkg(-1)-diet ( high dose ) 2AA for 14 and 28 days .\n\nThis was followed by analysis of the liver for global gene expression changes .\n\nIn both time points , the numbers of genes affected seem to correlate with the dose of 2AA .\n\nSixteen mRNAs were differentially expressed in all treatment groups for the short-term exposure group .\n\nSimilarly , 51 genes were commonly expressed in all 28-day exposure group .\n\nAlmost all the genes seem to have higher expression relative to the controls .\n\nIn contrast , cytochrome P450 family 4 , subfamily a , polypeptide 8 ( Cyp4a8 ) , and monocyte to macrophage differentiation-associated ( Mmd2 ) were down-regulated relative to controls .\n\nDifferentially expressed mRNAs were further analyzed for associations via DAVID .\n\nGO categories show the effect of 2AA to be linked with genes responsible for carbohydrate utilization and transport , lipid metabolic processes , stress responses such as inflammation and apoptosis processes , immune system response , DNA damage response , cancer processes and circadian rhythm .\n\nThe data from the current study identified altered hepatic gene expression profiles that may be associated with carcinoma , autoimmune response , and/or type 2 diabetes .\n\nPossible biomarkers due to 2AA toxicity in the liver for future study include Abcb1a , Nhej1 , Adam8 , Cdkn1a , Mgmt , and Nrcam .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Resisting cell death" }, { "input": "BACKGROUND Glioblastoma is the most common and most aggressive form of malignant glioma and is very difficult to treat .\n\nControlling tumour cell invasion and angiogenesis is essential to improve the prognosis of glioblastoma patients .\n\nSince constitutive activation of nuclear factor-\u03baB ( NF-\u03baB ) is necessary for tumour progression , NF-\u03baB may be an important pharmacological target for this disease .\n\nOur study aimed to evaluate the antitumour effects of parthenolide , a NF-\u03baB inhibitor , in two human glioblastoma cell lines ( U87MG and U373 ) and in glioblastoma xenografts .\n\nFurthermore , we aimed to investigate the molecular mechanisms underlying these effects .\n\nMETHODS The anti-invasive and anti-angiogenic effects of parthenolide were analysed using in vitro invasion and angiogenesis assays .\n\nParthenolide-induced growth inhibition of glioblastoma cells in vitro was determined using the MTT ( methyl thiazolyl tetrazolium ) assay .\n\nIn addition , the effect of parthenolide on orthotropic implantation in vivo was evaluated using an intracerebral human glioblastoma xenograft model .\n\nRESULTS We found that parthenolide suppresses proliferation , invasion , and tumour- induced angiogenesis of glioblastoma cells .\n\nMolecular studies demonstrated that parthenolide suppresses gene and protein expression of angiogenic factors .\n\nFurthermore , parthenolide reduced Akt phosphorylation and activated mitochondrial signalling , suggesting that the antitumour function of parthenolide may be mediated not only by the inhibition of NF-\u03baB but also by the inhibition of Akt signalling and the activation of apoptotic proteins .\n\nParthenolide suppressed neovascularity and tumour growth in glioblastoma xenografts .\n\nCONCLUSION The present study identified parthenolide as a new therapeutic agent for glioblastomas .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Overexpression of cyclin D1 is believed to endow mammary epithelial cells ( MEC ) with a proliferative advantage by virtue of its contribution to pRB inactivation .\n\nAccordingly , abrogation of the kinase-dependent function of cyclin D1 is sufficient to render mice resistant to breast cancer initiated by ErbB2 .\n\nHere , we report that mouse cyclin D1(KE/KE) MECs ( deficient in cyclin D1 activity ) upregulate an autophagy-like process but fail to implement ErbB2-induced senescence in vivo .\n\nIn addition , immortalized cyclin D1(KE/KE) MECs retain high rates of autophagy and reduced ErbB2-mediated transformation in vitro .\n\nHowever , highlighting its dual role during tumorigenesis , downregulation of autophagy led to an increase in senescence in cyclin D1(KE/KE) MECs .\n\nAutophagy upregulation was also confirmed in human mammary epithelial cells ( HMEC ) subjected to genetic and pharmacologic inhibition of cyclin D1 activity and , similar to our murine system , simultaneous inhibition of Cdk4/6 and autophagy in HMECs enhanced the senescence response .\n\nCollectively , our findings suggest a previously unrecognized function of cyclin D1 in suppressing autophagy in the mammary epithelium .\n\nCancer Res ; 72(24) ; 6477-89. \ufffd2012 AACR .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "The orphan receptor TR3 is an important regulator of cell proliferation and apoptosis .\n\nHowever , whether TR3 is involved in regulating the stem-like properties of cancer cells remains unknown .\n\nThe present study shows that TR3 expression is increased in gastric tumorsphere cells and is positively correlated with cancer stem cell ( CSC ) characteristics .\n\nKnocking down TR3 leads to the suppression of its stem-like properties in both gastric cancer cells and tumorsphere cells .\n\nThis process involves the decreased expression of the stemness-related genes Oct-4 and Nanog and the invasion-related gene MMP-9 .\n\nWe further identify Nanog as a new target for the transcription factor TR3 .\n\nTogether , these data demonstrate for the first time that TR3 is essential for the maintenance of stem-like properties in human gastric cancer cells and implicate TR3 as a new therapeutic target for gastric cancer .", "output": "Activating invasion and metastasis" }, { "input": "The recent approval of a prostate cancer vaccine has renewed hope for anticancer immunotherapies .\n\nHowever , the immunosuppressive tumor microenvironment may limit the effectiveness of current immunotherapies .\n\nAntiangiogenic agents have the potential to modulate the tumor microenvironment and improve immunotherapy , but they often are used at high doses in the clinic to prune tumor vessels and paradoxically may compromise various therapies .\n\nHere , we demonstrate that targeting tumor vasculature with lower vascular-normalizing doses , but not high antivascular/antiangiogenic doses , of an anti-VEGF receptor 2 ( VEGFR2 ) antibody results in a more homogeneous distribution of functional tumor vessels .\n\nFurthermore , lower doses are superior to the high doses in polarizing tumor-associated macrophages from an immune inhibitory M2-like phenotype toward an immune stimulatory M1-like phenotype and in facilitating CD4(+) and CD8(+) T-cell tumor infiltration .\n\nBased on this mechanism , scheduling lower-dose anti-VEGFR2 therapy with T-cell activation induced by a whole cancer cell vaccine therapy enhanced anticancer efficacy in a CD8(+) T-cell-dependent manner in both immune-tolerant and immunogenic murine breast cancer models .\n\nThese findings indicate that vascular-normalizing lower doses of anti-VEGFR2 antibody can reprogram the tumor microenvironment away from immunosuppression toward potentiation of cancer vaccine therapies .\n\nGiven that the combinations of high doses of bevacizumab with chemotherapy have not improved overall survival of breast cancer patients , our study suggests a strategy to use antiangiogenic agents in breast cancer more effectively with active immunotherapy and potentially other anticancer therapies .", "output": "Inducing angiogenesis, Avoiding immune destruction" }, { "input": "Neural stem cells ( NSCs ) are considered to be the cell of origin of glioblastoma multiforme ( GBM ) .\n\nHowever , the genetic alterations that transform NSCs into glioma-initiating cells remain elusive .\n\nUsing a unique transposon mutagenesis strategy that mutagenizes NSCs in culture , followed by additional rounds of mutagenesis to generate tumors in vivo , we have identified genes and signaling pathways that can transform NSCs into glioma-initiating cells .\n\nMobilization of Sleeping Beauty transposons in NSCs induced the immortalization of astroglial-like cells , which were then able to generate tumors with characteristics of the mesenchymal subtype of GBM on transplantation , consistent with a potential astroglial origin for mesenchymal GBM .\n\nSequence analysis of transposon insertion sites from tumors and immortalized cells identified more than 200 frequently mutated genes , including human GBM-associated genes , such as Met and Nf1 , and made it possible to discriminate between genes that function during astroglial immortalization vs. later stages of tumor development .\n\nWe also functionally validated five GBM candidate genes using a previously undescribed high-throughput method .\n\nFinally , we show that even clonally related tumors derived from the same immortalized line have acquired distinct combinations of genetic alterations during tumor development , suggesting that tumor formation in this model system involves competition among genetically variant cells , which is similar to the Darwinian evolutionary processes now thought to generate many human cancers .\n\nThis mutagenesis strategy is faster and simpler than conventional transposon screens and can potentially be applied to any tissue stem/progenitor cells that can be grown and differentiated in vitro .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Human podoplanin is a type-1 transmembrane sialomucin-like glycoprotein that is involved in cell migration , tumor cell invasion and metastasis .\n\nOur recent study of oral squamous cell carcinoma ( OSCC ) demonstrated that the degree of immunohistochemical expression of podoplanin was correlated with the severity of epithelial dysplasia and significantly associated with a poor pathologic grade of differentiation .\n\nFurthermore , it has been reported that Src directly associates with the epidermal growth factor receptor ( EGFR ) in OSCC cells upon stimulation with EGF and phosphorylates Crk-associated substrate ( Cas ) , podoplanin acting downstream of Src and Cas to promote cell migration .\n\nHowever , the molecular function of podoplanin remains unclear .\n\nIn this study we performed real-time RT-PCR , Western blotting and scratch assay using OSCC cell lines in order to clarify the molecular biological function of podoplanin expression associated with various growth factors including EGF and with the Src-Cas signaling pathway .\n\nPodoplanin was found to have a marked influence on cancer cell migration and the expression of matrix metalloprotease-9 ( MMP-9 ) in the oral cavity upon stimulation with EGF .\n\nPodoplanin promotes oral cancer cell migration , and the EGF-Src-Cas pathway is one of the possible mechanisms responsible for progression of cancer in the oral cavity .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "BACKGROUND : The immune system has been shown to play an important role in gastrointestinal stromal tumor ( GIST ) .\n\nThe neutrophil-to-lymphocyte ratio ( NLR ) in blood is an easily assessable parameter of systemic inflammatory response .\n\nThe aim of this study was to determine whether the NLR is prognostic in GIST .\n\nMETHODS : A total of 339 previously untreated patients with primary , localized GIST operated at our institution between 1995 and 2010 were identified from a prospectively collected sarcoma database .\n\nNLR was assessed preoperatively .\n\nPatients who received adjuvant imatinib treatment were excluded from the analysis ( n=64 ) .\n\nCox regression models were calculated and correlation analyses were performed .\n\nRESULTS : On univariate analysis , NLR was associated with recurrence-free survival ( RFS ) ( P=0.003 , hazard ratio 3.3 , 95% confidence interval 1.5-7.4 ) .\n\nPatients with a low NLR had a 1- and 5-year RFS of 98 and 91% , compared with 89 and 76% in those with a high NLR .\n\nThe median RFS was not reached .\n\nPositive correlations were found between NLR and mitotic rate ( Pearson correlation coefficient [ r]=0.15 , P=0.03 ) , and NLR and tumor size ( r=0.36 , P=0.0001 ) .\n\nRFS in patients with a GIST>5cm with low NLR was significantly longer compared to patients with high NLR ( P=0.002 ) .\n\nFlow cytometry analysis of freshly obtained GISTs revealed that neutrophils constituted a minimal percentage of intratumoral immune cells .\n\nCONCLUSIONS : NLR is a surrogate for high-risk tumor features .\n\nElevated blood NLR appears to represent systemic inflammation in patients with high-risk GIST .", "output": "Tumor promoting inflammation, Avoiding immune destruction" }, { "input": "The epithelial-mesenchymal transition ( EMT ) is a fundamental process governing morphogenesis in multicellular organisms and has recently been implicated in promoting carcinoma invasion and metastasis .\n\nBesides their therapeutic effects , accumulating evidences suggest that chemotherapeutic agents also induced EMT and enhanced the malignancy of treated cancer cells ; however , the mechanism(s) still remains unclear .\n\nHere , we investigated the role of \u03b2-catenin signaling in doxorubicin ( Dox)-induced EMT in human gastric cancer cell line BGC-823 .\n\nWe found that the transient treatment of Dox induced EMT and enhanced the in vitro migration ability of cancer cells .\n\nWe also found that \u03b2-catenin signaling was activated upon Dox treatment .\n\nInhibition of \u03b2-catenin by indomethacin ( Indo ) or siRNA suppressed Dox-induced EMT and decreased cancer cell migration ability .\n\nOur results showed that \u03b2-catenin signaling was critical to Dox-induced EMT .\n\nIndo and other \u03b2-catenin inhibitors may have a potential implication in prevention of gastric cancer metastasis .", "output": "Activating invasion and metastasis" }, { "input": "Certain mutations in BRCA1 and BRCA2 genes are frequent in the Ashkenazi Jewish population .\n\nSeveral factors contribute to this increased frequency , including consanguineous marriages and an event known as a \" bottleneck \" , which occurred in the past and caused a drastic reduction in the genetic variability of this population .\n\nSeveral studies were performed over the years in an attempt to elucidate the role of BRCA1 and BRCA2 genes in susceptibility to breast cancer .\n\nThe aim of this study was to estimate the carrier frequency of certain common mutations in the BRCA1 ( 185delAG and 5382insC ) and BRCA2 ( 6174delT ) genes in an Ashkenazi Jewish population from Porto Alegre , Brazil .\n\nMolecular analyses were done by PCR followed by RFLP ( ACRS ) .\n\nThe carrier frequencies for BRCA1 185delAG and 5382insC were 0.78 and 0 respectively , and 0.4 for the BRCA2 6174deT mutation .\n\nThese findings are similar to those of some prior studies but differ from others , possibly due to excluding individuals with a personal or family history of cancer .\n\nOur sample was drawn from the community group and included individuals with or without a family or personal history of cancer .\n\nFurthermore , increased dispersion among Ashkenazi subpopulations may be the result of strong genetic drift and/or admixture .\n\nIt is therefore necessary to consider the effects of local admixture on the mismatch distributions of various Jewish populations .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Growth Regulation by Estrogen in Breast cancer ( GREB1 ) was an estrogen receptor ( ER ) target gene , and GREB1 expression inversely correlated with HER2 status , possibly as a surrogate marker for ER status and a predictor for tamoxifen resistance in breast cancer patients .\n\nIn the present study , we examine the function and regulation of GREB1 in breast cancer , with the goal to develop GREB1 as a biomarker in breast cancer with de novo and acquired tamoxifen resistance .\n\nMETHODS We overexpressed GREB1 using adenovirus containing the full length GREB1 cDNA ( Ad-GREB1 ) in breast cancer cell lines .\n\nThe soft agar assay was used as a measure of anchorage independent growth .\n\nThe effects of GREB1 on cell proliferation in MCF-7 cells transduced with Ad-GREB1 were also measured by the me olic activity using AlamarBlue assay .\n\nWe tested whether there was interaction between STAT3 and ER , which could repress GREB1 expression by immunoprecipitation assay .\n\nThe effects of IL-6/JAK/STAT3 cascade activation on estrogen-induced GREB1 promoter activity were determined by luciferase assay and those on gene expression were measured by real time reverse transcription polymerase chain reaction ( qRT-PCR ) .\n\nRESULTS We found that the ability of breast cancer cells to grow in soft agar is enhanced following GREB1 transfection .\n\nIn MCF-7 cells transduced with Ad-GREB1 or transfected with siRNA GREB1 , the metabolic activity was increased or completely abolished , suggesting that GREB1 may function as a growth promoter in breast cancer .\n\nE2 treatment increased GREB1 promoter luciferase activity .\n\nIL-6 inhibited E2-induced GREB1 transcription activity and GREB1 mRNA expression .\n\nConstitutively expressing active STAT3 construct ( STAT3-C ) dramatically decreased GREB1 transcription .\n\nCONCLUSIONS These data indicate that overexpression of GREB1 promotes cell proliferation and increases the clonogenic ability in breast cancer cells .\n\nMoreover , Il6/STAT3 modulates estrogen-induced GREB1 transcriptional activity in breast cancer cells .", "output": "Sustaining proliferative signaling" }, { "input": "Vasohibin-1 ( VASH1 ) is isolated as an endothelial cell ( EC)-produced angiogenesis inhibitor .\n\nWe questioned whether VASH1 plays any role besides angiogenesis inhibition , knocked-down or overexpressed VASH1 in ECs , and examined the changes of EC property .\n\nKnock-down of VASH1 induced premature senescence of ECs , and those ECs were easily killed by cellular stresses .\n\nIn contrast , overexpression of VASH1 made ECs resistant to premature senescence and cell death caused by cellular stresses .\n\nThe synthesis of VASH1 was regulated by HuR-mediated post-transcriptional regulation .\n\nWe sought to define the underlying mechanism .\n\nVASH1 increased the expression of ( superoxide dismutase 2 ) SOD2 , an enzyme known to quench reactive oxygen species ( ROS ) .\n\nSimultaneously , VASH1 augmented the synthesis of sirtuin 1 ( SIRT1 ) , an anti-aging protein , which improved stress tolerance .\n\nParaquat generates ROS and causes organ damage when administered in vivo .\n\nMore VASH1 ( +/- ) mice died due to acute lung injury caused by paraquat .\n\nIntratracheal administration of an adenovirus vector encoding human VASH1 augmented SOD2 and SIRT1 expression in the lungs and prevented acute lung injury caused by paraquat .\n\nThus , VASH1 is a critical factor that improves the stress tolerance of ECs via the induction of SOD2 and SIRT1 .", "output": "Tumor promoting inflammation" }, { "input": "Widespread changes in gene expression drive tumorigenesis , yet our knowledge of how aberrant epigenomic and transcriptome profiles arise in cancer cells is poorly understood .\n\nHere , we demonstrate that metabolic transformation plays an important role .\n\nButyrate is the primary energy source of normal colonocytes and is metabolized to acetyl-CoA , which was shown to be important not only for energetics but also for HAT activity .\n\nDue to the Warburg effect , cancerous colonocytes rely on glucose as their primary energy source , so butyrate accumulated and functioned as an HDAC inhibitor .\n\nAlthough both mechanisms increased histone acetylation , different target genes were upregulated .\n\nConsequently , butyrate stimulated the proliferation of normal colonocytes and cancerous colonocytes when the Warburg effect was prevented from occurring , whereas it inhibited the proliferation of cancerous colonocytes undergoing the Warburg effect .\n\nThese findings link a common metabolite to epigenetic mechanisms that are differentially utilized by normal and cancerous cells because of their inherent metabolic differences .", "output": "Cellular energetics" }, { "input": "TNF , an inflammatory cytokine that is enriched in the tumor microenvironment , promotes tumor growth and subverts innate immune responses to cancer cells .\n\nWe previously reported that tumors implanted in TNF receptor-deficient ( Tnfr-/- ) mice are spontaneously rejected ; however , the molecular mechanisms underlying this rejection are unclear .\n\nHere we report that TNF signaling drives the peripheral accumulation of myeloid-derived suppressor cells ( MDSCs ) .\n\nMDSCs expand extensively during inflammation and tumor progression in mice and humans and can enhance tumor growth by repressing T cell-mediated antitumor responses .\n\nPeripheral accumulation of MDSCs was drastically impaired in Tnfr-/- mice .\n\nSignaling of TNFR-2 , but not TNFR-1 , promoted MDSC survival through upregulation of cellular FLICE-inhibitory protein ( c-FLIP ) and inhibition of caspase-8 activity .\n\nLoss of TNFRs impaired the induction of MDSCs from bone marrow cells , but this could be reversed by treatment with caspase inhibitors .\n\nThese results demonstrate that TNFR-2 signaling promotes MDSC survival and accumulation and helps tumor cells evade the immune system .", "output": "Tumor promoting inflammation" }, { "input": "This study intended to investigate the expression of the ZEB1 and E-cadherin proteins in lung squamous cell carcinoma ( LSCC ) tissues and to examine the clinicopathological correlation between protein levels and LSCC .\n\nRT-PCR and Western blot were used to examine the expression of ZEB1 and E-cadherin mRNAs and proteins in LSCC tissues as well as in adjacent normal tissues , and then analyze the relationship between the clinicopathological characteristics and the expression changes of ZEB1 and E-cadherin mRNAs in LSCC .\n\nIn addition , RNAi was used to knockdown the expression of the ZEB1 gene in Human HCC827 cells ; subsequently , changes in the invasive ability of the resultant cells were studied .\n\nThe positive rates of ZEB1 and E-cadherin mRNAs in LSCC tissues were 69.2 and 38.5% , respectively .\n\nThey differed significantly from the corresponding positive rates in the adjacent normal lung tissues ( 15.4 and 80.8% , p<0.05 ) .\n\nThere was a negative correlation between the protein levels of ZEB1 and E-cadherin in LSCC tissues ( r=-0.714 , p<0.001 ) ; in addition , it was found that ZEB1 protein expression in LSCC tissues was significantly higher than that in the neighboring normal lung tissues ( p<0.05 ) , and its expression was also significantly higher in patients with lymph node metastases and distant metastases compared to those patients without metastatic disease ( p<0.05 ) .\n\nOn the contrary , E-cadherin expression was significantly lower in LSCC tissues than that in the neighboring normal tissue ( p<0.05 ) .\n\nIt was lower in patients with lymph node metastasis and distant metastasis compared to patients without metastatic disease ( p<0.05 ) .\n\nHowever , the expression of ZEB1 and E-cadherin was independent of gender , age , tumor size , or tumor differentiation level ( p>0.05 ) .\n\nTransfection of ZEB1 siRNA into HCC827 cells significantly reduced the ZEB1 protein level ( p<0.01 ) and significantly elevated E-cadherin levels ( p<0.01 ) .\n\nMoreover , significantly less ZEB1 siRNA-transfected cells migrated through Transwell chambers in the LSCC tissue than that in the control groups ( untransfected or transfected with control siRNA , p<0.01 ) .\n\nThe expression of the ZEB1 gene in LSCC tissues is downregulated with the expression of E-cadherin .\n\nOn the other hand , the expression of siRNA against ZEB1 promotes E-cadherin expression and suppresses the invasive ability conferred by E-cadherin .\n\nIn conclusion , our data suggested that overexpression of the ZEB1 gene is possibly associated with the occurrence , development , invasion of LSCC .", "output": "Activating invasion and metastasis" }, { "input": "Although prostate cancer ( CaP ) is the most frequently diagnosed malignant tumor in American men , the mechanisms underlying the development and progression of CaP remain largely unknown .\n\nRecent studies have shown that downregulation of the microRNA miR-124 occurs in several types of human cancer , suggesting a tumor suppressive function of miR-124 .\n\nUntil now , however , it has been unclear whether miR-124 is associated with CaP .\n\nIn the present study , we completed a series of experiments to understand the functional role of miR-124 in CaP .\n\nWe detected the expression level of miR-124 in clinical CaP tissues , evaluated the influence of miR-124 on the growth of CaP cells and investigated the mechanism underlying the dysregulation of miR-124 .\n\nWe found that ( i ) miR-124 directly targets the androgen receptor ( AR ) and subsequently induces an upregulation of p53 ; ( ii ) miR-124 is significantly downregulated in malignant prostatic cells compared to benign cells , and DNA methylation causes the reduced expression of miR-124 ; and ( iii ) miR-124 can inhibit the growth of CaP cells in vitro and in vivo .\n\nData from this study revealed that loss of miR-124 expression is a common event in CaP , which may contribute to the pathogenesis of CaP .\n\nOur studies also suggest that miR-124 is a potential tumor suppressive gene in CaP , and restoration of miR-124 expression may represent a novel strategy for CaP therapy.Oncogene advance online publication , 15 October 2012 ; doi:10.1038/onc.2012.425 .", "output": "Sustaining proliferative signaling" }, { "input": "There is a complex interplay between the immune system and a developing tumor that is manifest in the way that the balance of Tcell subsets in the local tumor environment reflects clinical outcome .\n\nTumor infiltration by CD8(+) Tcells and regulatory Tcells ( Treg ) is associated with improved and reduced survival , respectively , in many cancer types .\n\nHowever , little is known of the prognostic value of immunological parameters measured in peripheral blood .\n\nIn this study , peripheral CD8(+) T cells and Treg from 43 patients with malignant mesothelioma or advanced non-small-cell lung cancer scheduled to commence palliative chemotherapy were assessed by flow cytometry and evaluated for association with patient survival .\n\nPatients had a higher proportion of peripheral Treg , proliferating CD8(+) Tcells and CD8(+) Tcells with an activated effector phenotype compared with age-matched healthy controls .\n\nHigher proportions of Treg and proliferating CD8(+) Tcells were both associated with poor survival in univariate analyses ( hazard ratio [ HR ] 3.81 , 95% CI 1.69-8.57 ; p<0.01 and HR 2.86 , 95% CI 1.26-6.50 ; p<0.05 , respectively ) .\n\nCD8(+) Tcell proliferation was independently predictive of reduced survival in multivariate analysis ( HR 2.58 , 95% CI 1.01-6.61 ; p<0.05 ) .\n\nThese findings suggest that peripheral CD8(+) T cell proliferation can be a useful prognostic marker in patients with thoracic malignancies planned for palliative chemotherapy .", "output": "Avoiding immune destruction" }, { "input": "This study examined the possible role of p120ctn in the pathogenesis and development of pancreatic cancer .\n\nPANC-1 cells , a kind of human pancreatic carcinoma cell line , were cultured in this study. p120ctn was immunocytochemically detected in PANC-1 cells .\n\nThe recombinant lentivirus vector was constructed to knock down the p120ctn expression of PANC-1 cells .\n\nReal-time quantitative PCR ( RQ-PCR ) and Western blotting were used to determine the expression of p120ctn and E-cadherin in PANC-1 cells after p120ctn knockdown .\n\nThe adhesion , invasion and migration capacity of PANC-1 cells after p120ctn knockdown was detected by cell adhesion , invasion and migration assays .\n\nCell growth was measured by the MTT method .\n\nCell cycle and apoptosis were analyzed by fluorescence-activated cell sorting .\n\nThe results showed that p120ctn knockdown led to significantly down-regulated E-cadherin and a reduced cell-to-cell adhesion ability in PANC-1 cells. shRNA-mediated knockdown of p120ctn reduced invasion and migration capacity of PANC-1 cells , inhibited cell growth , caused a significant decrease in the percentage of cells in G(1) , an increase in S , and promoted apoptosis of PANC-1 cells .\n\nIt was concluded that p120ctn plays a pivotal role in the proliferation and metastasis of pancreatic carcinoma , suggesting that p120ctn is a novel target for pancreatic carcinoma treatment .", "output": "Activating invasion and metastasis" }, { "input": "Liver metastasis from colorectal cancer is a leading cause of cancer mortality .\n\nMyeloid cells play pivotal roles in the metastatic process , but their prometastatic functions in liver metastasis remain incompletely understood .\n\nTo investigate their role , we simulated liver metastasis in C57BL/6 mice through intrasplenic inoculation of MC38 colon carcinoma cells .\n\nAmong the heterogeneous myeloid infiltrate , we identified a distinct population of CD11b/Gr1(mid) cells different from other myeloid populations previously associated with liver metastasis .\n\nThese cells increased in number dramatically during establishment of liver metastases and were recruited from bone marrow by tumor-derived CCL2 .\n\nLiver metastasis of Lewis lung carcinoma cells followed this pattern but this mechanism is not universal as liver colonization by B16F1 melanoma cells did not recruit similar subsets .\n\nInhibition of CCL2 signaling and absence of its cognate receptor CCR2 reduced CD11b/Gr1(mid) recruitment and decreased tumor burden .\n\nDepletion of the CD11b/Gr1(mid) subset in a transgenic CD11b-diphtheria toxin receptor mouse model markedly reduced tumor cell proliferation .\n\nThere was no evidence for involvement of an adaptive immune response in the prometastatic effects of CD11b/Gr1(mid) cells .\n\nAdditionally , an analogous myeloid subset was found in liver metastases of some colorectal cancer patients .\n\nConclusion : Collectively , our findings highlight the importance of myeloid cells-in this case a selective CD11b/Gr1(mid) subset-in sustaining development of colorectal cancer liver metastasis and identify a potential target for antimetastatic therapy .\n\n( HEPATOLOGY 2012 ) .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "Cancer can be envisioned as a metabolic disease driven by pressure selection and intercellular cooperativeness .\n\nTogether with anaerobic glycolysis , the Warburg effect , formally corresponding to uncoupling glycolysis from oxidative phosphorylation , directly participates in cancer aggressiveness , supporting both tumor progression and dissemination .\n\nThe transcription factor hypoxia-inducible factor-1 ( HIF-1 ) is a key contributor to glycolysis .\n\nIt stimulates the expression of glycolytic transporters and enzymes supporting high rate of glycolysis .\n\nIn this study , we addressed the reverse possibility of a metabolic control of HIF-1 in tumor cells .\n\nWe report that lactate , the end-product of glycolysis , inhibits prolylhydroxylase 2 activity and activates HIF-1 in normoxic oxidative tumor cells but not in Warburg-phenotype tumor cells which also expressed lower basal levels of HIF-1\\u03b1 .\n\nThese data were confirmed using genotypically matched oxidative and mitochondria-depleted glycolytic tumor cells as well as several different wild-type human tumor cell lines of either metabolic phenotype .\n\nLactate activates HIF-1 and triggers tumor angiogenesis and tumor growth in vivo , an activity that we found to be under the specific upstream control of the lactate transporter monocarboxylate transporter 1 ( MCT1 ) expressed in tumor cells .\n\nBecause MCT1 also gates lactate-fueled tumor cell respiration and mediates pro-angiogenic lactate signaling in endothelial cells , MCT1 inhibition is confirmed as an attractive anticancer strategy in which a single drug may target multiple tumor-promoting pathways .", "output": "Inducing angiogenesis, Cellular energetics" }, { "input": "A metaphase chromosome analysis method was used to evaluate the mutagenic activity of volatile petroleum fractions in chronically intoxicated rats .\n\nThe findings indicate that chronic inhalation exposure of the rats to volatile petroleum fractions at different concentrations results in a significant ( 2.5-fold ) increase , compared to the spontaneous level , in the occurrence of chromosomal aberrations in the bone marrow cells in a number of generations .\n\nIn addition to chromosomal structural abnormalities , there are genomic changes ( aneuploidy , polyploidy ) .\n\nThere is a tendency towards an increased rate of petroleum-induced chromosomal aberrations in a number of generations ( P , F1 , and F2 ) in the rat bone marrow cells .\n\nThe maximum mutagenic effect of volatile petroleum fractions was found when used at concentrations of 10 and 100 mg/l upon long-term chronic exposure during three generations ( P1 , F1 , and F2 ) .\n\nThe findings are indicative of the mutagenic activity of volatile petroleum fractions , which is likely to pose a potential risk from environmental pollution to biota and population health in the area of intensive oil extraction and refining .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Patients with invasive breast ductal carcinoma ( IBDC ) with metastasis have a very poor prognosis .\n\nLittle is known about the synergistic action of growth and inflammatory factors in IBDC metastases .\n\nMETHODS The expression of activated extracellular signal-regulated kinase1/2 ( phosphorylated or p-ERK1/2 ) was analyzed by immunohistochemistry in IBDC tissue samples from 80 cases .\n\nBT474 IBDC cell migration and invasion were quantified using the Transwell assay .\n\nMatrix metalloproteinase ( MMP)-9 expression and activity were analyzed by RT-PCR , Western blotting and zymography .\n\nActivator protein ( AP)-1 activity was measured with a luciferase reporter gene assay .\n\nThe Wilcoxon signed-rank test , Chi-square test , the partition of Chi-square test , independent t-test , and Spearman's method were used for the statistical analysis .\n\nRESULTS Phosphorylated ERK1/2 was detected in 58/80 ( 72.5% ) IBDC tissues , and was associated with higher TNM stage and lymph node metastasis , but not patient age or tumor size .\n\nIndividually , epidermal growth factor ( EGF ) , and interleukin ( IL)-1\u03b2 activated ERK1/2 , increased cell migration and invasion , MMP-9 expression and activity , AP-1 activation in vitro and the expression of p-ERK1/2 was positively correlated with EGF expression levels , as well as IL-1\u03b2 , MMP-9 and c-fos in IBDC tissue samples .\n\nCo-stimulation with EGF and IL-1\u03b2 synergistically increased ERK1/2 and AP-1 activation , cell migration and invasion , and MMP-9 expression and activity .\n\nInhibition of ERK1/2 using U0126 or siRNA abolished EGF and/or IL-1\u03b2-induced cell migration and invasion in a dose-dependent manner .\n\nCONCLUSION Activated ERK1/2 was associated with higher TNM stage and lymph node metastasis in IBDC .\n\nBoth in vitro and in vivo studies indicated that ERK-1/2 activation may increase the metastatic ability of IBDC cells .\n\nGrowth and inflammatory factors synergistically induced IBDC cell migration and invasion via ERK1/2 signaling , AP-1 activation and MMP-9 upregulation .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "PURPOSE Recently , a new renal cell cancer syndrome has been linked to germline mutation of multiple subunits ( SDHB/C/D ) of the Krebs cycle enzyme , succinate dehydrogenase .\n\nWe report our experience with the diagnosis , evaluation and treatment of this novel form of hereditary kidney cancer .\n\nMATERIALS AND METHODS Patients with suspected hereditary kidney cancer were enrolled on a National Cancer Institute institutional review board approved protocol to study inherited forms of kidney cancer .\n\nIndividuals from families with germline SDHB , SDHC and SDHD mutations , and kidney cancer underwent comprehensive clinical and genetic evaluation .\n\nRESULTS A total of 14 patients from 12 SDHB mutation families were evaluated .\n\nPatients presented with renal cell cancer at an early age ( 33 years , range 15 to 62 ) , metastatic kidney cancer developed in 4 and some families had no manifestation other than kidney tumors .\n\nAn additional family with 6 individuals found to have clear cell renal cell cancer that presented at a young average age ( 47 years , range 40 to 53 ) was identified with a germline SDHC mutation ( R133X ) Metastatic disease developed in 2 of these family members .\n\nA patient with a history of carotid body paragangliomas and an aggressive form of kidney cancer was evaluated from a family with a germline SDHD mutation .\n\nCONCLUSIONS SDH mutation associated renal cell carcinoma can be an aggressive type of kidney cancer , especially in younger individuals .\n\nAlthough detection and management of early tumors is most often associated with a good outcome , based on our initial experience with these patients and our long-term experience with hereditary leiomyomatosis and renal cell carcinoma , we recommend careful surveillance of patients at risk for SDH mutation associated renal cell carcinoma and wide surgical excision of renal tumors .", "output": "Genomic instability and mutation, Activating invasion and metastasis" }, { "input": "The aryl hydrocarbon receptor ( AhR ) contributes to the control of cell-to-cell communication , cell adhesion , migration or proliferation .\n\nIn the present study , we investigated the regulation of connexin43 ( Cx43 ) and Cx43-mediated gap junctional intercellular communication ( GJIC ) during the AhR-dependent disruption of contact inhibition in non-tumorigenic liver epithelial cells .\n\nThe contact inhibition of cell proliferation is a process restricting the cell division of confluent non-transformed cells , which is frequently abolished in cancer cells ; however , the mechanisms contributing to its disruption are still only partially understood .\n\nDisruption of contact inhibition , which was induced by toxic AhR ligands 2,3,7,8-tetrachlorodibenzo-p-dioxin ( TCDD ) or polycyclic aromatic hydrocarbons in epithelial WB-F344 cells , reduced Cx43 protein levels , possibly via enhanced proteasomal degradation , significantly decreased the amount of gap junction plaques and downregulated GJIC , in an AhR-dependent manner .\n\nAlthough both intracellular and membrane Cx43 pools were markedly reduced in cells released from contact inhibition by TCDD , siRNA-mediated Cx43 knock-down was not sufficient to stimulate proliferation in contact-inhibited cells .\n\nOur data suggest that downregulation of Cx43/GJIC in non-transformed epithelial cells is an inherent part of disruption of contact inhibition , which occurs at the post-transcriptional level .\n\nThis process runs in parallel with alterations of other forms of cell-to-cell communication , thus suggesting that toxic AhR agonists may simultaneously abrogate contact inhibition and reduce GJIC , two essential mechanisms linked to deregulation of cell-to-cell communication during tumor promotion and progression .", "output": "Evading growth suppressors" }, { "input": "Premature senescence , a key strategy used to suppress carcinogenesis , can be driven by p53/p21 proteins in response to various stresses .\n\nHere , we demonstrate that Wig1 plays a critical role in this process through regulation of p21 mRNA stability .\n\nWig1 controls the association of Argonaute2 ( Ago2 ) , a central component of the RNA-induced silencing complex ( RISC ) , with target p21 mRNA via binding of the stem-loop structure near the microRNA ( miRNA ) target site .\n\nDepletion of Wig1 prohibited miRNA-mediated p21 mRNA decay and resulted in premature senescence .\n\nWig1 plays an essential role in cell proliferation , as demonstrated in tumour xenografts in mice , and Wig1 and p21 mRNA levels are inversely correlated in human normal and cancer tissues .\n\nTogether , our data indicate a novel role of Wig1 in RISC target accessibility , which is a key step in RNA-mediated gene silencing .\n\nIn addition , these findings indicate that fine-tuning of p21 levels by Wig1 is essential for the prevention of cellular senescence .", "output": "Enabling replicative immortality" }, { "input": "Alteration of the oxidative stress of hepatocellular carcinoma ( HCC ) cells can influence the expressions of genes favored angiogenesis .\n\nQuinone reductase 2 which can activate quinones leading to reactive oxygen species production is a melatonin receptor known as MT3 .\n\nPrazosin prescribed for benign prostate hyperplasia and hypertension is a potent antagonist for MT3 .\n\nThis study was to investigate the influence of therapeutic concentrations of prazosin ( 0.01 and 0.1\u03bcM ) on cell proliferation and differential expressions of CCL2 , CCL20 , CXCL6 , CXCL10 , IL8 and IL6 genes related to inflammation and/or oxidative stress in human HCC cell lines .\n\nTwo HCC cell lines including one without susceptible to amphotericin B-induced oxidative stress ( cell line A ; HCC24/KMUH ) and one with this effect ( cell line B ; HCC38/KMUH ) were investigated by 0.01 and 0.1\u03bcM prazosin .\n\nThe premixed WST-1 cell proliferation reagent was applied for proliferation assay .\n\nDifferential expressions of genes were examined by quantitative reverse transcriptase-polymerase chain reaction .\n\nOur results showed that both 0.01 and 0.1\u03bcM prazosin did not influence cell proliferation in both cell lines .\n\nBoth 0.01 and 0.1\u03bcM prazosin in cell line A and 0.01\u03bcM prazosin in cell line B did not cause differential expressions of tested genes .\n\nHowever , 0.1\u03bcM prazosin caused remarkable up-regulation of IL6 gene and slightly up-regulation of CCL2 gene in cell line B. In conclusion , high therapeutic concentration of prazosin can up-regulate angiogenic IL6 and CCL2 genes in human HCC cells susceptible to amphotericin B-induced oxidative stress .\n\nClinical application of prazosin in patients with HCC should consider this possibility .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "Centrosome overduplication or amplification has been observed in many human cancers and in premalignant tissue , but the mechanisms leading to such centrosome aberrations are not fully understood .\n\nWe previously showed that abnormal mitotic cells with supernumerary centrosomes increase with replicative senescence in human fibroblasts , especially in a polyploid subpopulation .\n\nThis study examines localization of p53 protein at centrosomes in mitotic cells , which is often observed in association with DNA damage response , to investigate a possible association between p53 localization and numerical centrosome aberrations induced by cellular senescence .\n\nCultures at later passages or the 4th day after exposure to H(2)O(2) showed increased frequencies of mitotic cells with supernumerary centrosomes , especially in a polyploid subpopulation .\n\nImmunohistochemical analysis frequently showed p53-positive foci in mitotic cells , and some were localized at centrosomes .\n\nThe number of p53-positive foci in mitotic cells and its localization to centrosomes increased with replicative and premature senescence .\n\nSupernumerary centrosomes showed higher frequencies of p53 localization compared to normally duplicated centrosomes .\n\nCentrosome-associated p53 protein was phosphorylated at Ser15 .\n\nThese data suggest a possible association between localization of p53 protein and numerical centrosome aberrations in replicatively or prematurely senescent cells .", "output": "Enabling replicative immortality" }, { "input": "Purpose .\n\nThe purpose of this paper is to evaluate a new PET tracer ( 64)Cu-NODAGA-c(RGDyK ) for imaging of tumor angiogenesis using gene expression of angiogenesis markers as reference and to estimate radiation dosimetry for humans .\n\nProcedures .\n\nNude mice with human neuroendocrine tumor xenografts ( H727 ) were administered ( 64)Cu-NODAGA-c(RGDyK ) i.v. for study of biodistribution as well as for dynamic PET .\n\nGene expression of angiogenesis markers integrin \u03b1(V) , integrin \u03b2(3) , and VEGF-A were analyzed using QPCR and correlated to the tracer uptake in the tumors ( %ID/g ) .\n\nFrom biodistribution data human radiation-absorbed doses were estimated using OLINDA/EXM .\n\nResults .\n\nTumor uptake was 1.2%ID/g with strong correlations between gene expression and tracer uptake , for integrin \u03b1(V)\u2009R = 0.76 , integrin \u03b2(3)\u2009R = 0.75 and VEGF-A R = 0.81 ( all P < 0.05 ) .\n\nThe whole body effective dose for humans was estimated to be 0.038 and 0.029\u2009mSv/MBq for females and males , respectively , with highest absorbed dose in bladder wall .\n\nConclusion .\n\n( 64)Cu-NODAGA-c(RGDyK ) is a promising new angiogenesis PET tracer with potential for human use .", "output": "Inducing angiogenesis" }, { "input": "Cytokine-dependent cell lines have been used to analyze the cytokine-induced cellular signaling and the mechanism of oncogenesis .\n\nIn the current study , we analyzed MOTN-1 and PLT-2 cell lines established from different stages of a T-cell large granular lymphocyte leukemia patient ( Daibata et al. 2004 ) .\n\nMOTN-1 is IL-2-dependent derived from the chronic phase , whereas IL-2-independent PLT-2 is from the aggressive and terminal stage .\n\nThey shared considerable chromosome abnormalities and the pattern of T-cell receptor rearrangement , presuming that the cytokine independence of PLT-2 was due to the additive genetic abnormality .\n\nBesides IL-2 , IL-15 supported MOTN-1 cell growth , because these receptors share beta- and gamma-subunits .\n\nIL-2 activated ERK , AKT and STAT pathway of MOTN-1 .\n\nSTAT3 pathway of PLT-2 was also activated by IL-2 , suggesting intact IL-2 induces signal transduction of PLT-2 .\n\nHowever , ERK1/2 but not AKT , was continuously activated in PLT-2 , consistent with the increased Ras-activity of PLT-2 .\n\nSequence analysis revealed KRAS G12A mutation but not NRAS and HRAS mutation of PLT-2 but not MOTN-1 .\n\nAnother signaling molecule affecting Ras-signaling pathway , SHP2 , which has been frequently mutated in juvenile myelomonocytic leukemia ( JMML ) , did not show mutation .\n\nMoreover , MEK inhibitor , PD98059 , as well as farnesylation inhibitor inhibited PLT-2 cell growth .\n\nUsing NIH3T3 and MOTN-1 , ERK activation , increased cell proliferation and survival by KRAS G12A were shown , suggesting the important role of KRAS G12A in IL-2-independent growth of PLT-2 .\n\nTaken together , KRAS G12A is important for IL-2-independent growth of PLT-2 cells and suggests the possibility of involvement of KRAS mutation with disease progression .", "output": "Genomic instability and mutation" }, { "input": "Angiotensin II type 1 receptor ( AT1R ) promotes tumor invasion , migration , metastasis and angiogenesis .\n\nWe explored the potential antitumor effects of AT1R antagonists in breast cancer .\n\nWe found that angiotensin II promoted cell proliferation and upregulated the expression of vascular endothelial growth factor A ( VEGF-A ) in MCF-7 cells .\n\nLosartan downregulated the expression of VEGF-A in MCF-7 cells treated with angiotensin II .\n\nCandesartan downregulated the expression of VEGF-A in mice bearing MCF-7 xenografts and inhibited tumor growth and angiogenesis .\n\nAT1R and VEGF-A expression correlated with increased microvascular density in 102 breast cancer patients .\n\nOur data suggest that AT1R antagonists might be useful to suppress breast cancer by inhibiting the angiotensin II .", "output": "Inducing angiogenesis" }, { "input": "BACKGROUND Acrylamide is a common dietary exposure that crosses the human placenta .\n\nIt is classified as a probable human carcinogen , and developmental toxicity has been observed in rodents .\n\nOBJECTIVES We examined the associations between prenatal exposure to acrylamide and birth outcomes in a prospective European mother-child study .\n\nMETHODS Hemoglobin ( Hb ) adducts of acrylamide and its metabolite glycidamide were measured in cord blood ( reflecting cumulated exposure in the last months of pregnancy ) from 1,101 singleton pregnant women recruited in Denmark , England , Greece , Norway , and Spain during 2006-2010 .\n\nMaternal diet was estimated through food-frequency questionnaires .\n\nRESULTS Both acrylamide and glycidamide Hb adducts were associated with a statistically significant reduction in birth weight and head circumference .\n\nThe estimated difference in birth weight for infants in the highest versus lowest quartile of acrylamide Hb adduct levels after adjusting for gestational age and country was -132 g ( 95% CI : -207 , -56 ) ; the corresponding difference for head circumference was -0.33 cm ( 95% CI : -0.61 , -0.06 ) .\n\nFindings were similar in infants of nonsmokers , were consistent across countries , and remained after adjustment for factors associated with reduced birth weight .\n\nMaternal consumption of foods rich in acrylamide , such as fried potatoes , was associated with cord blood acrylamide adduct levels and with reduced birth weight .\n\nCONCLUSIONS Dietary exposure to acrylamide was associated with reduced birth weight and head circumference .\n\nConsumption of specific foods during pregnancy was associated with higher acrylamide exposure in utero .\n\nIf confirmed , these findings suggest that dietary intake of acrylamide should be reduced among pregnant women .", "output": "Genomic instability and mutation" }, { "input": "In cancer , glucose uptake and glycolysis are increased regardless of the oxygen concentration in the cell , a phenomenon known as the Warburg effect .\n\nSeveral ( but not all ) glycolytic enzymes have been investigated as potential therapeutic targets for cancer treatment using RNAi .\n\nHere , four previously untargeted glycolytic enzymes , aldolase A , glyceraldehyde 3-phosphate dehydrogenase , triose phosphate isomerase , and enolase 1 , are targeted using RNAi in Ras-transformed NIH-3T3 cells .\n\nOf these enzymes , knockdown of aldolase causes the greatest effect , inhibiting cell proliferation by 90% .\n\nThis defect is rescued by expression of exogenous aldolase .\n\nHowever , aldolase knockdown does not affect glycolytic flux or intracellular ATP concentration , indicating a non-metabolic cause for the cell proliferation defect .\n\nFurthermore , this defect could be rescued with an enzymatically dead aldolase variant that retains the known F-actin binding ability of aldolase .\n\nOne possible model for how aldolase knockdown may inhibit transformed cell proliferation is through its disruption of actin-cytoskeleton dynamics in cell division .\n\nConsistent with this hypothesis , aldolase knockdown cells show increased multinucleation .\n\nThese results are compared with other studies targeting glycolytic enzymes with RNAi in the context of cancer cell proliferation and suggest that aldolase may be a useful target in the treatment of cancer .", "output": "Cellular energetics" }, { "input": "Angiogenesis is a crucial step in the growth and metastasis of cancers , since it enables the growing tumor to receive oxygen and nutrients .\n\nCancer prevention using natural products has become an integral part of cancer control .\n\nWe studied the antiangiogenic activity of quercetin using ex vivo , in vivo and in vitro models .\n\nRat aortic ring assay showed that quercetin at non-toxic concentrations significantly inhibited microvessel sprouting and exhibited a significant inhibition in the proliferation , migration , invasion and tube formation of endothelial cells , which are key events in the process of angiogenesis .\n\nMost importantly , quercetin treatment inhibited ex vivo angiogenesis as revealed by chicken egg chorioallantoic membrane assay ( CAM ) and matrigel plug assay .\n\nWestern blot analysis showed that quercetin suppressed VEGF induced phosphorylation of VEGF receptor 2 and their downstream protein kinases AKT , mTOR , and ribosomal protein S6 kinase in HUVECs .\n\nQuercetin ( 20 mg/kg/d ) significantly reduced the volume and the weight of solid tumors in prostate xenograft mouse model , indicating that quercetin inhibited tumorigenesis by targeting angiogenesis .\n\nFurthermore , quercetin reduced the cell viability and induced apoptosis in prostate cancer cells , which were correlated with the downregulation of AKT , mTOR and P70S6K expressions .\n\nCollectively the findings in the present study suggest that quercetin inhibits tumor growth and angiogenesis by targeting VEGF-R2 regulated AKT/mTOR/P70S6K signaling pathway , and could be used as a potential drug candidate for cancer therapy .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Toll-like receptor 2 ( TLR2 ) is a target for immune system stimulation during cancer immunotherapy and a cell-surface marker for pancreatic cancer .\n\nTo develop targeted agents for cancer imaging and therapy , we designed , synthesized , and characterized 13 novel , fully synthetic high affinity TLR2 agonists .\n\nAnalogue 10 had the highest agonist activity ( NF-\u03baB functional assay , EC(50) = 20 nM ) and binding affinity ( competitive binding assay , K(i) = 25 nM ) .\n\nAs an immune adjuvant , compound 10 stimulated the immune system in vivo by generation and persistence of antigen-specific CD8+ T cells indicating its potential use in cancer immunotherapy .\n\nAfter conjugation of near-infrared dye to 10 , agonist activity ( EC(50) = 34 nM ) and binding affinity ( K(i) = 11 nM ) were retained in 13 .\n\nFluorescence signal was present in TLR2 expressing pancreatic tumor xenografts 24 h after injection of 13 , while an excess of unlabeled ligand blocked 13 from binding to the tumor , resulting in significantly decreased signal ( p < 0.001 ) demonstrating in vivo selectivity .", "output": "Avoiding immune destruction" }, { "input": "The CD4+CD25+ regulatory T cell ( Treg ) is a special kind of T cell subset .\n\nStudies have showed that Treg cells are involved in a number of physiological processes and pathologic conditions such as autoimmune diseases , transplantation tolerance and cancer .\n\nTregs with unique capacity for immune inhibition can impair anti-tumour immunity and help tumor cells to escape from immune surveillance .\n\nThe aim of our study was to investigate whether Tregs are involved in hepatocellular carcinoma ( HCC ) .\n\nA BABL/C mouse with HCC in situ model was established to evaluate the Treg existence in carcinoma tissues and the changes of Tregs in spleen using flow cytometry and immunohistochemistry methods .\n\nGranzyme B expression in carcinoma tissues was analyzed by immunohistochemistry to investigate the tumor local immune status .\n\nThe proportion of CD4+CD25+/CD4+ spleen lymphocytes of tumor bearing mice ( 18.8% \ufffd 1.26% ) was found to be significantly higher than that in normal mice ( 9.99% \ufffd 1.90% ) ( P<0.01 ) .\n\nImmunohistochemistry of spleen tissue also confirmed that there was an increase in Treg in tumor-bearing mice , while in carcinomas it showed Treg cells to be present in tumor infiltrating lymphocyte areas while Granzyme B was rarely observed .\n\nAnti-tumour immunity was suppressed , and this might be associated with the increase of Tregs .\n\nOur observations suggest that the CD4+CD25+Treg/ CD4+ proportion in spleen lymphocytes can be a sensitive index to evaluate the change of Tregs in hepatocellular carcinoma mice and the Treg may be a promising therapeutic target for cancer .", "output": "Avoiding immune destruction" }, { "input": "OBJECTIVE To evaluate the risk factors for sentinel lymph node metastasis and validate the value of the Memorial Sloan-Kettering Cancer Center nomogram for the prediction of sentinel lymph node metastasis in breast cancer patients .\n\nMETHODS A sentinel lymph node biopsy database containing 1227 consecutive breast cancer patients ( 416 patients with at least one positive sentinel lymph node ) was retrospectively analyzed .\n\nThe predictive value of the Memorial Sloan-Kettering Cancer Center nomogram was calculated by the trend line and the area under the receiver-operator characteristic curve .\n\nMeanwhile , predictors for sentinel lymph node metastasis were also evaluated .\n\nRESULTS Tumor size , histological grade , lymphovascular invasion , mulifocality , estrogen receptor and progesterone receptor status were significant independent predictors for sentinel lymph node metastasis ( all P<0.01 ) .\n\nThe Memorial Sloan-Kettering Cancer Center nomogram presented an area under the receiver-operator characteristic curve value of 0.730 .\n\nPatients with predictive value<16% had a frequency of sentinel lymph node metastasis of 0.9% .\n\nThose with values larger than 70% had a frequency of 96.2% .\n\nCONCLUSIONS The risk factors for sentinel lymph node metastasis in our study were consistent with those in the Memorial Sloan-Kettering Cancer Center nomogram .\n\nThe Memorial Sloan-Kettering Cancer Center nomogram is a useful tool that could accurately predict the probability of sentinel lymph node metastasis in our breast cancer patients .\n\nAxillary surgical staging might be avoided in patients with a predictive value of <16% and axillary lymph node dissection might be done directly in those with a predictive value >70% , while other patients should still accept sentinel lymph node biopsy .", "output": "Activating invasion and metastasis" }, { "input": "The topoisomerase II ( topo II ) DNA incision-and-ligation cycle can be poisoned ( for example following treatment with cancer chemotherapeutics ) to generate cytotoxic DNA double-strand breaks ( DSBs ) with topo II covalently conjugated to DNA .\n\nTyrosyl-DNA phosphodiesterase 2 ( Tdp2 ) protects genomic integrity by reversing 5'-phosphotyrosyl-linked topo II-DNA adducts .\n\nHere , X-ray structures of mouse Tdp2-DNA complexes reveal that Tdp2 \\u03b2-2-helix-\\u03b2 DNA damage-binding ' grasp ' , helical ' cap ' and DNA lesion-binding elements fuse to form an elongated protein-DNA conjugate substrate-interaction groove .\n\nThe Tdp2 DNA-binding surface is highly tailored for engagement of 5'-adducted single-stranded DNA ends and restricts nonspecific endonucleolytic or exonucleolytic processing .\n\nStructural , mutational and functional analyses support a single-metal ion catalytic mechanism for the exonuclease-endonuclease-phosphatase ( EEP ) nuclease superfamily and establish a molecular framework for targeted small-molecule blockade of Tdp2-mediated resistance to anticancer topoisomerase drugs .", "output": "Genomic instability and mutation" }, { "input": "The relevance of many BRCA2 variants of uncertain significance ( VUS ) to breast cancer has not been determined due to limited genetic information from families carrying these alterations .\n\nHere , we classified six new variants as pathogenic or nonpathogenic by analysis of genetic information from families carrying 64 individual BRCA2 DNA binding domain ( DBD ) missense mutations using a multifactorial likelihood model of cancer causality .\n\nNext , we evaluated the use of a homology-directed DNA break repair ( HDR ) functional assay as a method for inferring the clinical relevance of VUS in the DBD of BRCA2 using 18 established nonpathogenic missense variants and all 13 established pathogenic missense mutations from the BRCA2 DBD .\n\nCompared with the known status of these variants based on the multifactorial likelihood model , the sensitivity of the HDR assay for pathogenic mutations was estimated at 100% [ 95% confidence interval ( CI ) : 75.3%-100% ] and specificity was estimated at 100% ( 95% CI : 81.5%-100% ) .\n\nA statistical classifier for predicting the probability of pathogenicity of BRCA2 DBD variants was developed using these functional results .\n\nWhen applied to 33 additional VUS , the classifier identified eight with 99% or more probability of nonpathogenicity and 18 with 99% or more probability of pathogenicity .\n\nThus , in the absence of genetic evidence , a cell-based HDR assay can provide a probability of pathogenicity for all VUS in the BRCA2 DBD , suggesting that the assay can be used in combination with other information to determine the cancer relevance of BRCA2 VUS .", "output": "Genomic instability and mutation" }, { "input": "Dose-dense ( DD ) regimens of combination chemotherapy may produce superior clinical outcomes , but the basis for these effects are not completely clear .\n\nIn this study , we assessed whether a DD combinatorial regimen of low-dose cisplatin and paclitaxel produces superior immune-mediated efficacy when compared with a maximum tolerated dose ( MTD ) regimen in treating platinum-resistant ovarian cancer as modeled in mice .\n\nImmune responses generated by the DD regimen were identified with regard to the immune cell subset responsible for the antitumor effects observed .\n\nThe DD regimen was less toxic to the immune system , reduced immunosuppression by the tumor microenvironment , and triggered recruitment of macrophages and tumor-specific CD8(+) T-cell responses to tumors [ as determined by interleukin ( IL)-2 and IFN-\u03b3 secretion ] .\n\nIn this model , we found that the DD regimen exerted greater therapeutic effects than the MTD regimen , justifying its further clinical investigation .\n\nFourteen patients with platinum-resistant relapse of ovarian cancer received DD chemotherapy consisting of weekly carboplatin ( AUC2 ) and paclitaxel ( 60-80 mg/m(2) ) as the third- or fourth-line treatment .\n\nSerum was collected over the course of treatment , and serial IFN-\u03b3 and IL-2 levels were used to determine CD8(+) T-cell activation .\n\nOf the four patients with disease control , three had serum levels of IL-2 and IFN-\u03b3 associated with cytotoxic CD8(+) T-cell activity .\n\nThe therapeutic effect of the DD chemotherapy relied on the preservation of the immune system and the treatment-mediated promotion of tumor-specific immunity , especially the antitumor CD8(+) T-cell response .\n\nBecause the DD regimen controlled drug-resistant disease through a novel immune mechanism , it may offer a fine strategy for salvage treatment .\n\nCancer Res ; 73(1) ; 119-27. \ufffd2012 AACR .", "output": "Avoiding immune destruction" }, { "input": "Blockage of the metastasis process remains a significant clinical challenge , requiring innovative therapeutic approaches .\n\nFor this purpose , molecules that inhibit matrix metalloproteinases activity or induce the expression of their natural inhibitor , the tissue inhibitor of metalloproteinases ( TIMPs ) , are potentially interesting .\n\nIn a previous study , we have shown that synthetic ligands binding to cell surface nucleolin/nucleophosmin and known as HB 19 for the lead compound and NucAnt 6L ( N6L ) for the most potent analog , inhibit both tumor growth and angiogenesis .\n\nFurthermore , they prevent metastasis in a RET transgenic mice model which develops melanoma .\n\nHere , we investigated the effect of N6L on the invasion capacity of MDA-MB-435 melanoma cells .\n\nOur results show that the multivalent pseudopeptide N6L inhibited Matrigel invasion of MDA-MB-435 cells in a modified Boyden chamber model .\n\nThis was associated with an increase in TIMP-3 in the cell culture medium without a change in TIMP-3 mRNA expression suggesting its release from cell surface and/or extracellular matrix .\n\nThis may be explained by our demonstrated N6L interaction with sulfated glycosaminoglycans and consequently the controlled bioavailability of glycosaminoglycan-bound TIMP-3 .\n\nThe implication of TIMP-3 in N6L-induced inhibition of cell invasion was evidenced by siRNA silencing experiments showing that the loss of TIMP-3 expression abrogated the effect of N6L .\n\nThe inhibition of tumor cell invasion by N6L demonstrated in this study , in addition to its previously established inhibitory effect on tumor growth and angiogenesis , suggests that N6L represents a promising anticancer drug candidate warranting further investigation .", "output": "Inducing angiogenesis, Activating invasion and metastasis" }, { "input": "Melittin ( 1 ) is a major polypeptide in honey bee venom that has been used traditionally against chronic inflammation and cancer .\n\nHowever , its molecular mechanism has not been determined .\n\nIn this study , the antitumor effect of 1 was compared with that of NS398 , a cyclooxygenase-2 ( COX-2 ) inhibitor , in vivo and in vitro .\n\nSubcutaneous injection of 1 at 0.5 and 5 mg/kg suppressed significantly vascular endothelial growth factor ( VEGF)-A-transfected highly metastatic Lewis lung cancer ( VEGF-A-hm LLC ) tumor growth by 25% and 57% , respectively .\n\nAlso , 1 inhibited significantly the number of vessels around VEGF-A-hm LLC cells .\n\nThe results were superior to those obtained in the mice treated with NS398 .\n\nCompound 1 dose-dependently inhibited proliferation and tube formation in human umbilical vein endothelial cells ( VEGF-A-HUVECs ) , without affecting cell viability in native HUVECs .\n\nIn addition , 1 decreased the expression of VEGF receptor-2 ( VEGFR-2 ) , COX-2 , and prostaglandin E(2) ( PGE(2) ) in VEGF-A-transfected HUVECs .\n\nThese effects were accompanied by a reduction of the phosphorylation of extracellular signal-regulated kinase 1/2 and c-jun N-terminal kinase , whereas it increased the phosphorylation of p38 mitogen-activated protein kinase ( MAPK ) .\n\nSB203580 abolished the downregulation of COX-2 and VEGFR-2 and the inhibition of cell proliferation by 1 .\n\nThe antitumor activity of 1 may be associated with antiangiogenic actions via inhibiting VEGFR-2 and inflammatory mediators involved in the MAPK signaling pathway .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Although regulatory T cells ( T(regs) ) are known to suppress self-reactive autoimmune responses , their role during T cell responses to nonself antigens is not well understood .\n\nWe show that T(regs) play a critical role during the priming of immune responses in mice .\n\nT(reg) depletion induced the activation and expansion of a population of low-avidity CD8(+) T cells because of overproduction of CCL-3/4/5 chemokines , which stabilized the interactions between antigen-presenting dendritic cells and low-avidity T cells .\n\nIn the absence of T(regs) , the avidity of the primary immune response was impaired , which resulted in reduced memory to Listeria monocytogenes .\n\nThese results suggest that T(regs) are important regulators of the homeostasis of CD8(+) T cell priming and play a critical role in the induction of high-avidity primary responses and effective memory .", "output": "Avoiding immune destruction" }, { "input": "INTRODUCTION Characterized by the development of hundreds to thousands of colonic adenomas , classic familial adenomatous polyposis ( FAP ) is one of the most common hereditary syndromes associated with an increased risk of colorectal cancer .\n\nSeveral studies have attempted to correlate specific APC mutations with clinical phenotype.6 However , there is considerable variability in the expression of specific phenotypes within families and among individuals with identical mutations.7 CASE PRESENTATION A 30 year-old Hispanic female presented to the emergency department with a 2-week history of persistent , worsening , left lower quadrant abdominal pain .\n\nShe had no family history of malignancy .\n\nSigmoidoscopy revealed innumerable polyps in the rectum and sigmoid colon and a large mass in the sigmoid colon .\n\nBiopsy of the mass revealed a moderately differentiated adenocarcinoma invading the subserosa .\n\nEndoscopy revealed innumerable polyps .\n\nGenetic testing of the patient via southern blot revealed a germline APC mutation 3927del5 , resulting in a premature truncation of the APC protein at amino acid position 1312 .\n\nCONCLUSION Genetic information has only recently started being incorporated into clinical care .\n\nMore research and randomized clinical trials need to be conducted to definitively characterize random mutations .\n\nOnce these mutations are further understood , FAP patients may be able to be risk stratified and this may ultimately improve the screening , diagnosis , and treatment of this rare condition .", "output": "Genomic instability and mutation" }, { "input": "Trabecular meshwork ( TM ) cells have widely been used as an invitro model for glaucoma research .\n\nHowever , primary TM cells suffer the disadvantages of limited cell numbers and slow rates of proliferation .\n\nWe discovered a spontaneously transformed bovine TM ( BTM ) cell line , BTM-28T .\n\nThis cell line proliferated rapidly in low-glucose culture medium but also demonstrated contact inhibition in high-glucose culture medium .\n\nBTM-28T cells expressed key TM cell markers including \\u03b1-smooth muscle actin ( \\u03b1-SMA ) , laminin and collagen IV ( col IV ) .\n\nAlso , 100nM dexamethasone ( DEX ) enhanced the formation of cross-linked actin networks ( CLANs ) in confluent BTM-28T cell cultures .\n\nTransforming growth factor beta 2 ( TGF\\u03b22 ) induced the expression of fibronectin ( FN ) , plasminogen activator inhibitor-1 ( PAI-1 ) , and connective tissue growth factor ( CTGF ) in our cell cultures .\n\nThis cell line will be helpful to better understand the aqueous humor outflow pathway as related to the pathophysiology of glaucoma .", "output": "Evading growth suppressors" }, { "input": "BACKGROUND Investigations concerned the mechanism of HT-29 cells radiosensitization by cis-9,trans-11-conjugated linoleic acid ( c9,t11-CLA ) , a natural component of human diet with proven antitumor activity .\n\nMETHODS The cells were incubated for 24h with 70\\u03bcM c9,t11-CLA and then X-irradiated .\n\nThe following methods were used : gas chromatography ( incorporation of the CLA isomer ) , flow cytometry ( cell cycle ) , cloning ( survival ) , Western blotting ( protein distribution in membrane fractions ) , and pulse-field gel electrophoresis ( rejoining of DNA double-strand breaks ) .\n\nIn parallel , DNA-PK activity , \\u03b3-H2AX foci numbers and chromatid fragmentation were estimated .\n\nGene expression was analysed by RT-PCR and chromosomal aberrations by the mFISH method .\n\nNuclear accumulation of the EGF receptor ( EGFR ) was monitored by ELISA .\n\nRESULTS AND CONCLUSIONS C9,t11-CLA sensitized HT-29 cells to X-radiation .\n\nThis effect was not due to changes in cell cycle progression or DNA-repair-related gene expression .\n\nPost-irradiation DSB rejoining was delayed , corresponding with the insufficient DNA-PK activation , although chromosomal aberration frequencies did not increase .\n\nDistributions of cholesterol and caveolin-1 in cellular membrane fractions changed .\n\nThe nuclear EGFR translocation , necessary to increase the DNA-PK activity in response to oxidative stress , was blocked .\n\nWe suppose that c9,t11-CLA modified the membrane structure , thus disturbing the intracellular EGFR transport and the EGFR-dependent pro-survival signalling , both functionally associated with lipid raft properties .\n\nGENERAL SIGNIFICANCE The results point to the importance of the cell membrane interactions with the nucleus after injury inflicted by X -rays .\n\nCompounds like c9,t11-CLA , that specifically alter membrane properties , could be used to develop new anticancer strategies .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Lung cancer is primarily caused by exposure to tobacco smoke .\n\nTobacco smoke contains numerous carcinogens , including polycyclic aromatic hydrocarbons ( PAH ) .\n\nThe most common PAH studied is benzo[a]pyrene ( B[a]P ) .\n\nB[a]P is metabolically activated through multiple routes , one of which is catalyzed by aldo-keto reductase ( AKR ) to B[a]P-7,8-dione ( BPQ ) .\n\nBPQ undergoes a futile redox cycle in the presence of NADPH to generate reactive oxygen species ( ROS ) .\n\nROS , in turn , damages DNA .\n\nStudies with a yeast p53 mutagenesis system found that the generation of ROS by PAH o-quinones may contribute to lung carcinogenesis because of similarities between the patterns ( types of mutations ) and spectra ( location of mutations ) and those seen in lung cancer .\n\nThe patterns were dominated by G to T transversions , and the spectra in the experimental system have mutations at lung cancer hotspots .\n\nTo address repair mechanisms that are responsible for BPQ induced damage we observed the effect of mutating two DNA repair genes OGG1 and APE1 ( APN1 in yeast ) and tested them in a yeast reporter system for p53 mutagenesis .\n\nThere was an increase in both the mutant frequency and the number of G:C/T:A transversions in p53 treated with BPQ in ogg1 yeast but not in apn1 yeast .\n\nKnocking out APN2 increased mutagenesis in the apn1 cells .\n\nIn addition , we did not find a strand bias on p53 treated with BPQ in ogg1 yeast .\n\nThese studies suggest that Ogg1 is involved in repairing the oxidative damage caused by BPQ , Apn1 and Apn2 have redundant functions and that the stand bias seen in lung cancer may not be due to impaired repair of oxidative lesions .", "output": "Genomic instability and mutation" }, { "input": "The most potent tumorigen identified among the polycyclic aromatic hydrocarbons ( PAH ) is the nonplanar fjord region dibenzo[a,l]pyrene ( DB[a,l]P ) .\n\nIt is metabolically activated in vivo through the widely studied diol epoxide ( DE ) pathway to form covalent adducts with DNA bases , predominantly guanine and adenine .\n\nThe ( +)-11S,12R,13R,14S DE enantiomer forms adducts via its C14 position with the exocyclic amino group of guanine .\n\nHere , we present the first nuclear magnetic resonance solution structure of a DB[a,l]P-derived adduct , the 14R-(+)-trans-anti-DB[a,l]P-N(2)-dG ( DB[a,l]P-dG ) lesion in double-stranded DNA .\n\nIn contrast to the stereochemically identical benzo[a]pyrene-derived N(2)-dG adduct ( B[a]P-dG ) in which the B[a]P rings reside in the B-DNA minor groove on the 3'-side of the modifed deoxyguanosine , in the DB[a,l]P-derived adduct the DB[a,l]P rings intercalate into the duplex on the 3'-side of the modified base from the sterically crowded minor groove .\n\nWatson-Crick base pairing of the modified guanine with the partner cytosine is broken , but these bases retain some stacking with the bulky DB[a,l]P ring system .\n\nThis new theme in PAH DE-DNA adduct conformation differs from ( 1 ) the classical intercalation motif in which Watson-Crick base pairing is intact at the lesion site and ( 2 ) the base-displaced intercalation motif in which the damaged base and its partner are extruded from the helix .\n\nThe structural considerations that lead to the intercalated conformation of the DB[a,l]P-dG lesion in contrast to the minor groove alignment of the B[a]P-dG adduct , and the implications of the DB[a,l]P-dG conformational motif for the recognition of such DNA lesions by the human nucleotide excision repair apparatus , are discussed .", "output": "Genomic instability and mutation" }, { "input": "Molecular mechanisms involved in progression of clear-cell renal-cell carcinomas ( ccRCCs ) are poorly understood .\n\nA common genetic mutation found in ccRCC is the loss of the von Hippel-Lindau ( VHL ) gene , which contributes to cancer progression and metastasis .\n\nWe investigated VIP effects on metastatic and angiogenic factors in human VHL-null A498 ccRCC and HK2 renal cells .\n\nVIP increased adhesion but decreased expression of metalloproteinases , MMP2 and MMP9 , as well as cell migration and VEGF expression and secretion in A498 but not in HK2 cells .\n\nVIP enhanced ROS levels and decreased nuclear levels of \u03b2-catenin and NF\u03baB p50-subunit in A498 cells , suggesting neuropeptide involvement in the observed decrease of metastatic ability in clear-cell carcinoma .\n\nVIP effects in A498 cells were blocked by the VPAC(1/2)-receptor antagonist JV-1-53 .\n\nIn conclusion , present data point to a role of VIP in preventing invasion and metastasis in ccRCCs and support its potential therapeutic usefulness in this disease .", "output": "Inducing angiogenesis, Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "Angiogenesis is the process of new blood vessel formation from pre-existing ones .\n\nAngiogenic factors contribute to neovascularization that takes place in angiogenesis-dependent diseases , including cancer .\n\nInhibiting the activity of the angiogenic factors to block the angiogenesis pathways is the current strategy of cancer therapy .\n\nBasic fibroblast growth factor ( bFGF ) is regarded as one of the most important angiogenic factors .\n\nHerein , we selected polyoxometalates ( POMs ) with different structures to study the interactions between bFGF and POMs .\n\nThe results show that POMs could bind to the protein with high affinity , causing detectable changes in conformation and biophysical properties of protein .\n\nIn addition , POMs could effectively inhibit the cell proliferation induced by bFGF .\n\nSignificantly , we found that the structure , size and composition of POMs play a key role in the interactions between bFGF and POMs .\n\nThis study will be meaningful for future screening and design of polyoxometalate-based anticancer drugs .", "output": "Inducing angiogenesis" }, { "input": "Mitochondrial uncoupling protein 2 ( UCP2 ) can moderate oxidative stress by favoring the influx of protons into the mitochondrial matrix , thus reducing electron leakage from respiratory chain and mitochondrial superoxide production .\n\nHere , we demonstrate that UCP2 inhibition by genipin or UCP2 siRNA strongly increases reactive oxygen species ( ROS ) production inhibiting pancreatic adenocarcinoma cell growth .\n\nWe also show that UCP2 inhibition triggers ROS-dependent nuclear translocation of the glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase ( GAPDH ) , formation of autophagosomes , and the expression of the autophagy marker LC3-II .\n\nConsistently , UCP2 over-expression significantly reduces basal autophagy confirming the anti-autophagic role of UCP2 .\n\nFurthermore , we demonstrate that autophagy induced by UCP2 inhibition determines a ROS-dependent cell death , as indicated by the apoptosis decrease in the presence of the autophagy inhibitors chloroquine ( CQ ) or 3-methyladenine ( 3-MA ) , or the radical scavenger NAC .\n\nIntriguingly , the autophagy induced by genipin is able to potentiate the autophagic cell death triggered by gemcitabine , the standard chemotherapeutic drug for pancreatic adenocarcinoma , supporting the development of an anti-cancer therapy based on UCP2 inhibition associated to standard chemotherapy .\n\nOur results demonstrate for the first time that UCP2 plays a role in autophagy regulation bringing new insights into mitochondrial uncoupling protein field .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Hepatocellular carcinoma ( HCC ) is one of the most common malignancies worldwide ; however , the prognosis of HCC patients remains poor .\n\nThis poor prognosis is mainly attributed to the high rate of intrahepatic and distant metastasis .\n\nHCC often occurs in a hypoxic environment and hypoxia can activate metastatic programs , ultimately leading to tumor recurrence or metastasis .\n\nThus , the discovery and subsequent development of novel agents to block HCC invasion and migration are the primary objectives of hepatic cancer research .\n\nThe Notch1 signaling pathway might be involved in hypoxia-induced carcinoma metastasis .\n\nHowever , the mechanisms by which Notch1 mediates cell metastasis , particularly in hepatocellular carcinoma , are not yet entirely clear .\n\nThe results of the present study show that hypoxia increases the invasion and migration capacities of different HCC cells .\n\nActivation of the Notch1 signaling pathway contributes to hypoxia-induced invasion and migration in HCC cells .\n\nThe activated Notch1 signaling pathway can regulate Snail/E-cadherin through cyclooxygenase-2 ( COX-2 ) under hypoxic conditions .\n\nThe above results suggest that the Notch1/COX-2/Snail/E-cadherin pathway is possibly associated with hypoxia-induced invasion and migration in HCC cells .\n\nThus , targeting Notch1 may be useful for devising novel preventive and therapeutic strategies for HCC .", "output": "Activating invasion and metastasis" }, { "input": "Acrolein ( Acr ) is a ubiquitous environmental pollutant found in cigarette smoke and automobile exhaust .\n\nIt can also be produced endogenously by oxidation of polyunsaturated fatty acids .\n\nThe Acr-derived 1,N(2)-propanodeoxyguanosine ( Acr-dG ) adducts in DNA are mutagenic lesions that are potentially involved in human cancers .\n\nIn this study , monoclonal antibodies were raised against Acr-dG adducts and characterized using ELISA .\n\nThey showed strong reactivity and specificity toward Acr-dG , weaker reactivity toward crotonaldehyde- and trans-4-hydroxy-2-nonenal-derived 1,N(2)-propanodeoxyguanosines , and weak or no reactivity toward 1,N(6)-ethenodeoxyadenosine and 8-oxo-deoxyguanosine .\n\nUsing these antibodies , we developed assays to detect Acr-dG in vivo : first , a simple and quick FACS-based assay for detecting these adducts directly in cells ; second , a highly sensitive direct ELISA assay for measuring Acr-dG in cells and tissues using only 1 \\u03bcg of DNA without DNA digestion and sample enrichment ; and third , a competitive ELISA for better quantitative measurement of Acr-dG levels in DNA samples .\n\nThe assays were validated using Acr-treated HT29 cell DNA samples or calf thymus DNA , and the results were confirmed by LC-MS/MS-MRM .\n\nAn immunohistochemical assay was also developed to detect and visualize Acr-dG in HT29 cells as well as in human oral cells .\n\nThese antibody-based methods provide useful tools for the studies of Acr-dG as a cancer biomarker and of the molecular mechanisms by which cells respond to Acr-dG as a ubiquitous DNA lesion .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND/AIMS Analysis of cystic fluid may be useful in distinguishing between benign and malignant cysts which has significant impact on their management .\n\nThe aim of our study was to assess the diagnostic utility of carcinoembryonic antigen ( CEA ) and K-ras gene mutation in pancreatic cysts fluid .\n\nMETHODS The study included 56 patients with pancreatic cystic fluid collected for analysis .\n\nThe cysts were classified as benign ( simple cysts , pseudocysts , serous cystadenoma ) - 39 patients or premalignant/malignant ( mucinous cystadenoma , IPMN , cystadenocarcinoma ) - 17 patients .\n\nThe patients history , CEA fluid concentrations and presence of K-ras mutation were analyzed .\n\nRESULTS CEA were higher in patients with malignant cysts ( mean levels 238\u00b112.5ng/ml ; range 32.8-4985ng/ml ) compared to benign lesions ( mean levels 34.5\u00b13.7ng/ml ; range 3.9-693ng/ml ; p<0.001 ) .\n\nK-ras mutation correctly classified 11 of 17 patients with premalignant/malignant lesions .\n\nIt was also detected in 1 patient with final diagnosis of benign cyst ( the sensitivity 64.7% and the specificity 97.4% ; p<0.01 ) .\n\nIf CEA and molecular analysis were combined in that cysts with either CEA level>45ng/ml or presence of K-ras mutation , than 16 of 17 premalignant/malignant cysts were correctly identified ( 94.1% ) .\n\nCONCLUSION Molecular analysis of pancreatic cyst fluid adds diagnostic value to the preoperative diagnosis and should be considered when cyst cytologic examination is negative for malignancy .", "output": "Genomic instability and mutation" }, { "input": "The mammalian COP9 signalosome ( CSN ) complex is involved in cell transformation , but its molecular mechanism remains undetermined .\n\nHere we show that disruption of the fifth component ( CSN5 ) prevented the formation of tumors by p53-null cells transformed with an active form of Ras in subcutaneously injected mice .\n\nDepletion of CSN5 suppressed cell proliferation , and induced premature senescence characterized by upregulation of senescence-associated-\u03b2-galactosidase activity and increased expression of CDK inhibitors .\n\nCSN5-depleted cells exhibited enhanced activation of the PI3 kinase-Akt pathway , and chemical inhibition of this pathway reduced the level of senescence .\n\nThus , CSN5 is suggested to be a novel target in cancer therapy and for drugs against tumor cells harboring mutated p53 .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "Glioblastoma multiforme ( GBM ) and oxidative stress are closely linked .\n\nOxidative stress affects many signaling pathways and may cause the induction of autophagy .\n\nThe NF-E2-related factor 2 ( Nrf2)/Kelch-like ECH-associated protein 1 ( Keap1 ) signaling pathway is the main pathway responsible for cell defense against oxidative stress and Nrf2 is a critical transcription factor related with cancer multidrug resistance .\n\nHowever , the relation between Nrf2 and regulation of autophagy is not well understood .\n\nIn this study , we used temozolomide ( TMZ ) , which inhibited the viability of GBM cells mainly by inducing autophagic cell death and explored the role of Nrf2 downregulation on autophagy induced by TMZ in GBM cells .\n\nIn U251-Si-Nrf2 48h after transfection the protein levels of Nrf2 were significantly downregulated , while the protein levels of LC3B-II increased by western blot analysis .\n\nKnockdown of Nrf2 also led to a significant increase of autophagic vacuoles and acidic vesicular organelles ( AVOs ) , revealed by trans-mission electron microscopy ( TEM ) and acridine orange ( AO ) staining using flow cytometry .\n\nCollectively , these findings demonstrate that knockdown of Nrf2 can enhance the basal level of autophagy in the U251 glioma cell line .\n\nFurthermore , after the treatment with TMZ ( 100\u00b5M ) for 3 days , the U251-Si-Nrf2 transfected cells showed less viability rate by cell counting kit-8 ( CCK-8 ) assay and the levels of autophagy increased obviously through analysis of western blot and AO staining using flow cytometry .\n\nTaken together , our results suggest that knockdown of Nrf2 may enhance autophagy induced by TMZ in the U251 glioma cell line , which should be further evaluated for novel anticancer activity .", "output": "Resisting cell death" }, { "input": "LewisY ( LeY ) antigen is an oligosaccharide that is highly expressed at the cell surface in various human cancers .\n\nIncreased LeY expression activates epidermal growth factor receptor ( EGFR ) and human epidermal growth factor receptor 2 ( HER2 ) and promotes cell proliferation in EGFR-overexpressing cells .\n\nHowever , the effect of downregulation of LeY expression on cell proliferation in HER2-overexpressing cells remains unknown .\n\nFUT1 encodes \u03b11,2-fucosyltransferase , a key enzyme for LeY synthesis .\n\nWe knocked down FUT1 by short interfering RNA ( siRNA ) in four HER2-overexpressing human cancer cell lines , including NCI-N87 , MKN7 , SKBr3 and BT474 .\n\nWe investigated whether downregulation of LeY and alteration in the glycosylation status of these cells affect cell proliferation and HER2 activation .\n\nKnocking down FUT1 expression markedly inhibited proliferation of NCI-N87 , which highly expressed EGFR and was sensitive to EGFR deprivation .\n\nFurthermore , FUT1 siRNA downregulated the total amount of HER2 protein , phosphorylation of HER2 and EGFR , and phosphorylation of extracellular signal-regulated kinase ( ERK ) in this cell line .\n\nMoreover , the marked downregulation of phosphorylation of HER2 and ERK was observed following short-time EGF-stimulation .\n\nThese effects were not observed in the other three cell lines .\n\nOur results suggest that knockdown of FUT1 downregulates HER2 signaling via EGFR downregulation .\n\nFUT1 may serve as a new molecular target for HER2-overexpressing human cancers with activated EGFR signaling .", "output": "Sustaining proliferative signaling" }, { "input": "Blind mole rats Spalax ( BMR ) are small subterranean rodents common in the Middle East .\n\nBMR is distinguished by its adaptations to life underground , remarkable longevity ( with a maximum documented lifespan of 21 y ) , and resistance to cancer .\n\nSpontaneous tumors have never been observed in spalacids .\n\nTo understand the mechanisms responsible for this resistance , we examined the growth of BMR fibroblasts in vitro of the species Spalax judaei and Spalax golani .\n\nBMR cells proliferated actively for 7-20 population doublings , after which the cells began secreting IFN-\\u03b2 , and the cultures underwent massive necrotic cell death within 3 d .\n\nThe necrotic cell death phenomenon was independent of culture conditions or telomere shortening .\n\nInterestingly , this cell behavior was distinct from that observed in another long-lived and cancer-resistant African mole rat , Heterocephalus glaber , the naked mole rat in which cells display hypersensitivity to contact inhibition .\n\nSequestration of p53 and Rb proteins using SV40 large T antigen completely rescued necrotic cell death .\n\nOur results suggest that cancer resistance of BMR is conferred by massive necrotic response to overproliferation mediated by p53 and Rb pathways , and triggered by the release of IFN-\\u03b2 .\n\nThus , we have identified a unique mechanism that contributes to cancer resistance of this subterranean mammal extremely adapted to life underground .", "output": "Enabling replicative immortality, Evading growth suppressors, Resisting cell death" }, { "input": "Chromosomal translocations typically impair cell differentiation and often require secondary mutations for malignant transformation .\n\nHowever , the role of a primary translocation in the development of collaborating mutations is debatable .\n\nTo delineate the role of leukemic translocation NUP98-HOXD13 ( NHD13 ) in secondary mutagenesis , DNA break and repair mechanisms in stimulated mouse B lymphocytes expressing NHD13 were analyzed .\n\nOur results showed significantly reduced expression of non-homologous end joining ( NHEJ)-mediated DNA repair genes , DNA Pkcs , DNA ligase4 , and Xrcc4 leading to cell cycle arrest at G2/M phase .\n\nOur results showed that expression of NHD13 fusion gene resulted in impaired NHEJ-mediated DNA break repair .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "BACKGROUND Dickkopf-related protein 1 ( DKK1 ) has been reported involved in metastasis and invasion in several tumors .\n\nThis study sought to investigate the prognostic value of DKK1 in intrahepatic cholangiocarcinoma ( ICC ) and its role in promoting ICC metastasis .\n\nMETHODS Tissue microarrays of 138 ICC patient samples were employed to detect DKK1 , vascular endothelial growth factor C ( VEGF-C ) , and matrix metalloproteinase 9 ( MMP9 ) expression using immunohistochemistry .\n\nThe prognostic significances were assessed by Kaplan-Meier survival estimates .\n\nDKK1 expression was measured in an ICC cell line ( HCCC-9810 ) and ICC tissues by immunofluorescence assay , quantitative real-time polymerase chain reaction , and western blot .\n\nSerum levels of DKK1 from 37 ICC patients were tested by enzyme-linked immunosorbent assay .\n\nThe role of DKK1 in proliferation , migration , invasion , and gene expression regulation was assessed by DKK1 depletion using small interfering RNA .\n\nRESULTS Multivariate analyses revealed that DKK1 was an unfavorable predictor for overall survival and time to recurrence .\n\nThe prognostic significance was retained in ICC patients with low recurrence risk ( P < .05 ) .\n\nDKK1 expression was elevated in an ICC cell line , tumor samples , and patient sera .\n\nHigh levels of DKK1 in ICC tissues correlated with elevated MMP9 , VEGF-C , and metastasis of hepatic hilar lymph nodes .\n\nDKK1 depletion caused a decrease in cell migration and invasiveness , and down-regulation of MMP9 and VEGF-C expression .\n\nCONCLUSIONS DKK1 is a novel prognostic biomarker for ICC , and it enhances tumor cell invasion and promotes lymph node metastasis of ICC through the induction of MMP9 and VEGF-C .\n\nDKK1 may be a potential therapeutic target for ICC .", "output": "Activating invasion and metastasis" }, { "input": "A prominent feature of inflammatory diseases is endothelial dysfunction .\n\nFactors associated with endothelial dysfunction include proinflammatory cytokines , adhesion molecules , and matrix degrading enzymes .\n\nAt the transcriptional level , they are regulated by the histone deacetylase sirtuin ( SIRT ) 1 via its actions on the proinflammatory transcription factor nuclear factor-\u03baB ( NF-\u03baB ) .\n\nThe role of SIRT6 , also a histone deacetylase , in regulating inflammation in endothelial cells is not known .\n\nThe aim of this study was to determine the effect of SIRT6 knockdown on inflammatory markers in human umbilical vein endothelial cells ( HUVECs ) in the presence of lipopolysaccharide ( LPS ) .\n\nLPS decreased expression of SIRT6 in HUVECs .\n\nKnockdown of SIRT6 increased the expression of proinflammatory cytokines ( IL-1\u03b2 , IL-6 , IL-8 ) , COX-prostaglandin system , ECM remodelling enzymes ( MMP-2 , MMP-9 and PAI-1 ) , the adhesion molecule ICAM-1 , and proangiogenic growth factors VEGF and FGF-2 ; cell migration ; cell adhesion to leukocytes .\n\nLoss of SIRT6 increased the expression of NF-\u03baB , whereas overexpression of SIRT6 was associated with decreased NF-\u03baB transcriptional activity .\n\nTaken together , these results demonstrate that the loss of SIRT6 in endothelial cells is associated with upregulation of genes involved in inflammation , vascular remodelling , and angiogenesis .\n\nSIRT6 may be a potential pharmacological target for inflammatory vascular diseases .", "output": "Inducing angiogenesis, Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "New molecularly targeted therapies are needed for childhood ependymoma .\n\nAngiogenesis and the PDGFR pathway could be potential therapeutic targets .\n\nThis study aimed to screen ependymomas for the expression and clinicopathological correlates of angiogenic factors and potential therapeutic targets including VEGFR , endoglin ( CD105 ) , CD34 , CD31 , c-Kit , PDGFR-\u03b1 and PDGFR-\u03b2 .\n\nImmunohistochemistry for angiogenesis factors and PDGFR-\u03b1 and \u03b2 was performed in 24 archival tumor samples from children and adults treated for ependymoma at our institution .\n\nCD31 density , CD105 density and pericyte coverage index ( PCI ) were calculated .\n\nThese findings were correlated with clinical outcome .\n\nVEGFR2 was overexpressed in tumor cells in only one out of 24 cases , but was found overexpressed in the vessels in 6 cases .\n\nPDGFR-\u03b1 and \u03b2 were found to be over-expressed in the ependymoma tumor cells in seven out of 24 cases ( 29.2 % ) .\n\nCD31 density , CD105 density and PCI did not correlate with expression of PDGFRs .\n\nOverexpression of PDGFR-\u03b1 and \u03b2 in tumor cells and overexpression of PDGFR-\u03b1 in tumor endothelium had prognostic significance and this was maintained in multivariate analysis for overexpression of PDGFR-\u03b1 in tumor cells ( 2 year progression free survival was 16.7 \u00b1 15.2 for cases with overexpression of PDGFR-\u03b1 in the tumor vs. 74.5 \u00b1 15.2 for those with low/no expression , hazard ratio = 5.78 , p = 0.04 ) .\n\nA number of angiogenic factors are expressed in ependymoma tumor cells and tumor endothelium .\n\nPreliminary evidence suggests that the expression of PDGFRs could have a prognostic significance in ependymoma .\n\nThis data suggests that PDGFRs should be further evaluated as targets using novel PDGFR inhibitors .", "output": "Inducing angiogenesis" }, { "input": "BACKGROUND Engineered zinc-finger nucleases ( ZFN ) represented an innovative method for the genome manipulation in vertebrates .\n\nZFN introduced targeted DNA double strand breaks ( DSB ) and initiated non-homologous end joining ( NHEJ ) after pronuclear or cytoplasmatic microinjection into zygotes .\n\nResulting frame shift mutations led to functional gene ablations in zebra fish , mice , pigs and also in laboratory rats .\n\nTherefore , we targeted the rat Rag1 gene essential for the V(D)J recombination within the immunoglobulin production process and for the differentiation of mature B and T lymphocytes to generate an immunodeficient rat model in the LEW/Ztm strain .\n\nRESULTS After microinjection of Rag1 specific ZFN mRNAs in 623 zygotes of inbred LEW/Ztm rats 59 offspring were born from which one carried a 4 bp deletion .\n\nThis frame shift mutation led to a premature stop codon and a subsequently truncated Rag1 protein confirmed by the loss of the full-length protein in Western Blot analysis .\n\nTruncation of the Rag1 protein was characterized by the complete depletion of mature B cells .\n\nThe remaining T cell population contained mature CD4+/CD3+/TCR\\u03b1\\u03b2+ as well as CD8+/CD3+/TCR\\u03b1\\u03b2+ positive lymphocytes accompanied by a compensatory increase of natural killer cells in the peripheral blood .\n\nReduction of T cell development in Rag1 mutant rats was associated with a hypoplastic thymus that lacked follicular structures .\n\nHistological evaluation also revealed the near-complete absence of lymphocytes in spleen and lymph nodes in the immunodeficient Rag1 mutant rat .\n\nCONCLUSION The Rag1 mutant rat will serve as an important model for transplantation studies .\n\nFurthermore , it may be used as a model for reconstitution experiments related to the immune system , particularly with respect to different populations of human lymphocytes , natural killer cells and autoimmune phenomena .", "output": "Genomic instability and mutation, Avoiding immune destruction" }, { "input": "Inorganic arsenic in the drinking water is a multisite human carcinogen that potentially targets the kidney .\n\nRecent evidence also indicates that developmental arsenic exposure impacts renal carcinogenesis in humans and mice .\n\nEmerging theory indicates that cancer may be a disease of stem cells ( SCs ) and that there are abundant active SCs during early life .\n\nTherefore , we hypothesized that inorganic arsenic targets SCs , or partially differentiated progenitor cells ( PCs ) , for oncogenic transformation .\n\nThus , a rat kidney SC/PC cell line , RIMM-18 , was chronically exposed to low-level arsenite ( 500 nM ) for up to 28 weeks .\n\nMultiple markers of acquired cancer phenotype were assessed biweekly during arsenic exposure , including secreted matrix metalloproteinase ( MMP ) activity , proliferation rate , colony formation in soft agar , and cellular invasiveness .\n\nArsenic exposure by 10 weeks and after also induced marked and sustained increases in colony formation , indicative of the loss of contact inhibition , and increased invasiveness , both cancer cell characteristics .\n\nCompared to the passage-matched control , chronic arsenic exposure caused exposure-duration dependent increases in secreted MMP-2 and MMP-9 activity , Cox-2 expression , and more rapid proliferation ( all >2-fold ) , characteristics typical of cancer cells .\n\nDysregulation of SC maintenance genes and signaling pathways are common during oncogenesis .\n\nDuring arsenite exposure , expression of several genes associated with normal kidney development and SC regulation and differentiation ( i.e. , Wt-1 , Wnt-4 , Bmp-7 , etc. ) were aberrantly altered .\n\nArsenic-exposed renal SCs produced more nonadherent spheroid bodies that grew much more aggressively in Matrigel , typical of cancer SCs ( CSCs ) .\n\nThe transformed cells also showed gene overexpression typical of renal SCs/CSCs ( CD24 , Osr1 , Ncam ) and arsenic adaptation such as overexpression of Mt-1 , Mt2 , Sod-1 , and Abcc2 .\n\nThese data suggest that inorganic arsenic induced an acquired cancer phenotype in vitro in these rat kidney SCs potentially forming CSCs and , consistent with data in vivo , indicate that these multipotent SCs may be targets of arsenic during renal carcinogenesis .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling, Evading growth suppressors, Activating invasion and metastasis" }, { "input": "We previously demonstrated that downregulation of protein kinase CKII induces cellular senescence in human colon cancer HCT116 cells .\n\nTo investigate the role of microRNAs ( miRNAs ) in CKII downregulation during senescence , we employed computational algorithms .\n\nFour miRNAs ( miR-186 , miR-216b , miR-337-3p , and miR-760 ) were predicted to be miRNAs against CKII\u03b1 mRNA .\n\nMimics of all four miRNAs jointly downregulated CKII\u03b1 expression in HCT116 cells .\n\nReporter analysis and RT-PCR have suggested that these four miRNAs may stimulate degradation of CKII\u03b1 mRNA by targeting its 3 ' untranslated regions ( UTRs ) .\n\nThe four miRNA mimics increased senescent-associated \u03b2-galactosidase ( SA-\u03b2-gal ) staining , p53 and p21(Cip1/WAF1) expression , and reactive oxygen species ( ROS ) production .\n\nIn contrast , concomitant knockdown of the four miRNAs by antisense inhibitors increased the CKII\u03b1 protein level and suppressed CKII inhibition-mediated senescence .\n\nFinally , CKII\u03b1 overexpression antagonized senescence induced by the four miRNA mimics .\n\nTherefore , the present results show that miR-186 , miR-216b , miR-337-3p , and miR-760 cooperatively promote cellular senescence through the p53-p21(Cip1/WAF1) pathway by CKII downregulation-mediated ROS production in HCT116 cells .", "output": "Tumor promoting inflammation, Enabling replicative immortality" }, { "input": "Aberrant expression of mucins is likely associated with cancer biology as alterations in the expression and/or glycosylation patterns of various mucins have been noted .\n\nExpression of the mucin family in gastric cancers has been reported in numerous studies , but the results are conflicting .\n\nTherefore , we investigated the potential use of mucin ( MUC)1 and 4 as prognostic markers in gastric cancer according to histological subtype .\n\nThree-hundred and sixty-five gastric adenocarcinoma patients who underwent surgical resection were selected for this study .\n\nAmong the 365 gastric cancer samples tested here , 34% consisted of early gastric cancer and 66% were advanced .\n\nIn terms of location , 68.7% of the cohort had intestinal-type cancer and 30.7% had diffuse-type .\n\nWe constructed tissue microarrays with formalin-fixed paraffin-embedded blocks of gastric cancer and these micro-arrays were evaluated for phenotypic expression of MUC1/4 using monoclonal antibodies .\n\nTwo-hundred and ninety-two patients ( 92.7% ) were positive for MUC1 and 216 ( 60.5% ) were positive for MUC4 .\n\nMUC1 expression was not correlated with any other clinicopathological variables such as age , gender , depth of invasion , lymph node metastasis , Lauren classification or recurrence .\n\nHowever , loss of MUC4 expression was significantly correlated with recurrence ( p=0.033 ) .\n\nMUC4 expression was also significantly correlated with better disease-free survival ( p=0.049 ) and particularly in the intestinal-type ( p=0.018 ) .\n\nOur present findings demonstrated that loss of MUC4 expression can be used as a prognostic marker in gastric cancer .\n\nLoss of MUC4 expression is a prognostic indicator of increased recurrence and poor disease-free survival in patients with gastric cancer .", "output": "Activating invasion and metastasis" }, { "input": "To investigate whether altered energy metabolism induces the Warburg effect and results in tumor malignancy , the respiratory enzyme citrate synthase ( CS ) was examined , silenced , and the effects analyzed .\n\nIn human cervical carcinoma cells , RNAi-mediated CS knockdown induced morphological changes characteristic of the epithelial-mesenchymal transition ( EMT ) .\n\nThis switch accelerated cancer cell metastasis and proliferation in in vitro assays and in vivo tumor xenograft models .\n\nNotably , CS knockdown cells exhibited severe defects in respiratory activity and marked decreases in ATP production , but great increases in glycolytic metabolism .\n\nThis malignant progression was due to activation of EMT-related regulators ; altered energy metabolism resulted from deregulation of the p53/TIGAR and SCO2 pathways .\n\nThis phenotypic change was completely reversed by p53 reactivation via treatment with proteasome inhibitor MG132 or co-knockdown of E3 ligase HDM2 and partially suppressed by ATP treatment .\n\nThis study directly links the Warburg effect to tumor malignancy via induction of the EMT phenotype .", "output": "Cellular energetics, Activating invasion and metastasis" }, { "input": "OBJECT A considerable body of evidence indicates that inflammation and angiogenesis play a significant role in the development and progression of chronic subdural hematoma ( CSDH ) .\n\nWhile various experimental and clinical studies have implicated placental growth factor ( PlGF ) in the processes that underpin pathological angiogenesis , no study has thus far investigated its expression in CSDH .\n\nThe actions of PlGF and its related proangiogenic vascular endothelial growth factor ( VEGF ) are antagonized by a high-affinity soluble receptor , namely soluble VEGF receptor-1 ( sVEGFR-1 ) , and thus the ratio between sVEGFR-1 and angiogenic factors provides an index of angiogenic capacity .\n\nMETHODS In the present study , using an automated electrochemiluminescence assay , levels of PlGF and sVEGFR-1 were quantified in serum and hematoma fluid obtained in 16 patients with CSDH .\n\nRESULTS Levels of PlGF and sVEGFR-1 were significantly higher in hematoma fluid than in serum ( p < 0.0001 ) .\n\nIn serum , levels of sVEGFR-1 were higher than those of PlGF ( p < 0.0001 ) , whereas in hematoma fluid this difference was not apparent .\n\nFurthermore , the ratio of sVEGFR-1 to PlGF was significantly lower in hematoma fluid than in serum ( p < 0.0001 ) .\n\nCONCLUSIONS Given previous evidence indicating a role for PlGF in promoting angiogenesis , inflammatory cell chemotaxis , and stimulation , as well as its ability to amplify VEGF-driven signaling under conditions favoring pathological angiogenesis , enhanced expression of PlGF in hematoma fluid suggests the involvement of this factor in the mechanisms of inflammation and angiogenesis in CSDH .\n\nFurthermore , a reduced ratio of sVEGFR-1 to PlGF in hematoma fluid is consistent with the proangiogenic capacity of CSDH .\n\nFuture studies are warranted to clarify the precise role of PlGF and sVEGFR-1 in CSDH .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "When the prostate cancer cells become unresponsive to androgen therapy , resistance to chemotherapy becomes imminent , resulting in high mortality .\n\nTo combat this situation , lycopodine , a pharmacologically important bioactive component derived from Lycopodium clavatum spores , was tested against hormone sensitive ( LnCaP ) and refractory ( PC3 ) prostate cancer cells in vitro .\n\nThis study aims to check if lycopodine has demonstrable anti-cancer effects and if it has , to find out the possible mechanism of its action .\n\nThe MTT assay was performed to evaluate the cytotoxic effect .\n\nDepolarization of mitochondrial membrane potential , cell cycle , EGF receptor activity and apoptosis were recorded by FACS ; profiles of different anti- and pro-apoptotic genes and their products were studied by semi-quantitative RT-PCR , indirect-ELISA , western blotting .\n\nDrug-DNA interaction was determined by CD spectroscopy .\n\nAdministration of lycopodine down-regulated the expression of 5-lipoxygenase and the 5-oxo-ETE receptor ( OXE receptor1 ) and EGF receptor , and caused up-regulation of cytochrome c with depolarization of mitochondrial inner membrane potential , without palpable change in p53 activity , resulting in apoptosis , cell arrest at G0/G1 stage and ultimately reduced proliferation of cancer cells ; concomitantly , there was externalization of phosphotidyl serine residues .\n\nCD spectroscopic analysis revealed intercalating property of lycopodine with DNA molecule , implicating its ability to block cellular DNA synthesis .\n\nThe overall results suggest that lycopodine is a promising candidate suitable for therapeutic use as an anti-cancer drug .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND AND AIMS : Impairment of the immune system may contribute to the risk for having cancer as Toll-like receptors are important for innate immunity .\n\nWe examined the association between candidate disease-susceptibility polymorphisms in the single nucleotide polymorphism ( SNPs ) like TLR2 ( -196 to-174del ) , TLR3 ( C1377T ) , TLR4 ( Thr399Ile ) and TLR9 ( G2848A ) genes in patients with bladder cancer in a North Indian population .\n\nMETHODS : SNPs were comprised of TLR2 ( -196 to -174 Del ) , TLR3(C1377T) , TLR4 ( Thr399Ile ) and TLR9 ( G2848A ) genes .\n\nAllelic and genotypic frequencies of these TLRs SNP from histopathologically confirmed patients of bladder cancer ( n=200 ) and unrelated healthy controls of similar ethnicity ( n=200 ) were genotyped by polymerase chain reaction restriction fragment length polymorphism ( PCR-RFLP ) analysis .\n\nRESULTS : In TLR2 I/D gene polymorphism , the combination of ID+DD showed a significant 3-fold increased risk ( p=0.001 ) .\n\nTLR2 with heterozygous genotype ID showed a 3-fold risk and combination of heterozygous and variant genotype ( ID+DD ) also showed a 5-fold risk with tumor stage/grade of patients with bladder cancer .\n\nThe other genotypes of TLR3 , 4 and 9 did not exhibit any significant association with bladder cancer risk .\n\nCONCLUSIONS : Our results suggested the involvement of TLR2 ( -196 to-174 del ) in bladder cancer susceptibility ; however , TLR3 , 4 and 9 genes were not associated with risk of bladder cancer , implicating that polymorphisms in these tested TLRs genes are not likely to be associated with increased risk for developing bladder cancer .\n\nFunctional studies in ethnically diverse populations may provide a more comprehensive involvement of innate immunity in identifying the disease-associated variants for the etiology of bladder cancer .", "output": "Avoiding immune destruction" }, { "input": "Cellular senescence is a permanent out-of-cycle state regulated by molecular circuits acting during the G1 phase of the cell cycle .\n\nCdt1 is a central regulator of DNA replication licensing acting during the G1 phase and it is negatively controlled by Geminin .\n\nHere , we characterize the cell cycle expression pattern of Cdt1 and Geminin during successive passages of primary fibroblasts and compare it to tumour-derived cell lines .\n\nCdt1 and Geminin are strictly expressed in distinct subpopulations of young fibroblasts , similarly to cancer cells , with Geminin accumulating shortly after the onset of S phase .\n\nCdt1 and Geminin are down-regulated when primary human and mouse fibroblasts undergo replicative or stress-induced senescence .\n\nRNAi-mediated Geminin knock-down in human cells enhances the appearance of phenotypic and molecular features of senescence .\n\nMouse embryonic fibroblasts heterozygous for Geminin exhibit accelerated senescence compared to control fibroblasts .\n\nIn contrast , ectopic expression of Geminin in mouse embryonic fibroblasts delays the appearance of the senescent phenotype .\n\nTaken together , our data suggest that changes in Geminin expression levels affect the establishment of senescence pathways .", "output": "Enabling replicative immortality" }, { "input": "DNA mismatch repair ( MMR ) of simple base mismatches and small insertion-deletion loops in eukaryotes is initiated by the binding of the MutS homolog 2 ( MSH2)-MSH6 heterodimer to mismatched DNA .\n\nCadmium ( Cd ) is a genotoxic heavy metal that has been recognized as a human carcinogen .\n\nOxidant stress and inhibition of DNA repair have been proposed as major factors underlying Cd genotoxicity .\n\nOur previous studies indicated the ability of Cd to disturb the gene expression of MSH6 in zebrafish ( Danio rerio ) embryos .\n\nThis study was undertaken to explore if Cd-induced oxidative stress down-regulated MSH gene activities .\n\nFollowing the exposure of zebrafish embryos at 1 h post fertilization ( hpf ) to sublethal concentrations of Cd at 3-5 \\u03bcM for 4 or 9 h , a parallel down-regulation of MSH2 , MSH6 and Cu/Zn superoxide dismutase ( Cu/Zn-SOD ) gene expression was detected by real-time RT-PCR and the expression levels were 40-50% of control after a 9-h exposure .\n\nCd exposure also induced oxidative stress , yet no inhibition of catalase gene activity was observed .\n\nWhole mount in situ hybridization revealed a wide distribution of msh6 mRNA in the head regions of 10 hpf embryos and pretreatment of embryos with antioxidants butylhydroxytoluene ( BHT ) , d-mannitol or N-acetylcysteine ( NAC ) at 1-10 \\u03bcM restored Cd-suppressed msh6 expression .\n\nQPCR confirmed the protective effects of antioxidants on Cd-suppressed msh2/msh6 mRNA production .\n\nDown-regulated MSH gene activities reaching about 50% of control were also induced in embryos exposed to paraquat , a reactive oxygen species ( ROS)-generating herbicide , or hydrogen peroxide at 200 \\u03bcM .\n\nHence , Cd at sublethal levels down-regulates msh2/msh6 expression primarily via ROS as signaling molecules .\n\nThe transcriptional activation of human msh6 is known to be fully dependent on the specificity factor 1 ( Sp1 ) .\n\nCd failed to inhibit the DNA binding activity of zebrafish Sp1 unless at lethal concentrations based on band shift assay , therefore excluding the involvement of Sp1 inactivation in Cd-induced MSH gene inhibition in zebrafish embryos .", "output": "Tumor promoting inflammation" }, { "input": "Progression to malignancy requires that cells overcome senescence and switch to an immortal phenotype .\n\nThus , exploring the genetic and epigenetic changes that occur during senescence/immortalization may help elucidate crucial events that lead to cell transformation .\n\nIn the present study , we have globally profiled DNA methylation in relation to gene expression in primary , senescent and immortalized mouse embryonic fibroblasts .\n\nUsing a high-resolution genome-wide mapping technique , followed by extensive locus-specific validation assays , we have identified 24 CpG islands that display significantly higher levels of CpG methylation in immortalized cell lines as compared to primary murine fibroblasts .\n\nSeveral of these hypermethylated CpG islands are associated with genes involved in the MEK-ERK pathway , one of the most frequently disrupted pathways in cancer .\n\nApproximately half of the hypermethylated targets are developmental regulators , and bind to the repressive Polycomb group ( PcG ) proteins , often in the context of bivalent chromatin in mouse embryonic stem cells .\n\nBecause PcG-associated aberrant DNA methylation is a hallmark of several human malignancies , our methylation data suggest that epigenetic reprogramming of pluripotency genes may initiate cell immortalization .\n\nConsistent with methylome alterations , global gene expression analysis reveals that the vast majority of genes dysregulated during cell immortalization belongs to gene families that converge into the MEK-ERK pathway .\n\nAdditionally , several dysregulated members of the MAP kinase network show concomitant hypermethylation of CpG islands .\n\nUnlocking alternative epigenetic routes for cell immortalization will be paramount for understanding crucial events leading to cell transformation .\n\nUnlike genetic alterations , epigenetic changes are reversible events , and as such , can be amenable to pharmacological interventions , which makes them appealing targets for cancer therapy when genetic approaches prove inadequate .", "output": "Enabling replicative immortality" }, { "input": "Mps one binder 1a ( MOB1A ) and MOB1B are key components of the Hippo signaling pathway and are mutated or inactivated in many human cancers .\n\nHere we show that intact Mob1a or Mob1b is essential for murine embryogenesis and that loss of the remaining WT Mob1 allele in Mob1a(\\u0394/\\u0394)1b(tr/+) or Mob1a(\\u0394/+)1b(tr/tr) mice results in tumor development .\n\nBecause most of these cancers resembled trichilemmal carcinomas , we generated double-mutant mice bearing tamoxifen-inducible , keratinocyte-specific homozygous-null mutations of Mob1a and Mob1b ( kDKO mice). kDKO mice showed hyperplastic keratinocyte progenitors and defective keratinocyte terminal differentiation and soon died of malnutrition. kDKO keratinocytes exhibited hyperproliferation , apoptotic resistance , impaired contact inhibition , enhanced progenitor self renewal , and increased centrosomes .\n\nExamination of Hippo pathway signaling in kDKO keratinocytes revealed that loss of Mob1a/b altered the activities of the downstream Hippo mediators LATS and YAP1 .\n\nSimilarly , YAP1 was activated in some human trichilemmal carcinomas , and some of these also exhibited MOB1A/1B inactivation .\n\nOur results clearly demonstrate that MOB1A and MOB1B have overlapping functions in skin homeostasis , and exert their roles as tumor suppressors by regulating downstream elements of the Hippo pathway .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "We previously found that cancer metastasis is accelerated by immunosuppression during Snail-induced epithelial-to-mesenchymal transition ( EMT ) .\n\nHowever , the molecular mechanism still remained unclear .\n\nHere , we demonstrate that CCL2 is a critical determinant for both tumor metastasis and immunosuppression induced by Snail(+) tumor cells .\n\nCCL2 is significantly upregulated in various human tumor cells accompanied by Snail expression induced by snail transduction or TGF\u03b2 treatment .\n\nThe Snail(+) tumor-derived CCL2 amplifies EMT events in other cells including Snail(-) tumor cells and epithelial cells within tumor microenvironment .\n\nCCL2 secondarily induces Lipocalin 2 ( LCN2 ) in the Snail(+) tumor cells in an autocrine manner .\n\nCCL2 and LCN2 cooperatively generate immunoregulatory dendritic cells ( DCreg ) having suppressive activity accompanied by lowered expression of costimulatory molecules such as HLA-DR but increased expression of immunosuppressive molecules such as PD-L1 in human PBMCs .\n\nThe CCL2/LCN2-induced DCreg cells subsequently induce immunosuppressive CD4(+)FOXP3(+) Treg cells , and finally impair tumor-specific CTL induction .\n\nIn murine established tumor model , however , CCL2 blockade utilizing the specific siRNA or neutralizing mAb significantly inhibits Snail(+) tumor growth and metastasis following systemic induction of anti-tumor immune responses in host .\n\nThese results suggest that CCL2 is more than a chemoattractant factor that is the significant effector molecule responsible for immune evasion of Snail(+) tumor cells .\n\nCCL2 would be an attractive target for treatment to eliminate cancer cells via amelioration of tumor metastasis and immunosuppression .", "output": "Sustaining proliferative signaling, Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "Adoptive T cell therapy uses the specificity of the adaptive immune system to target cancer and virally infected cells .\n\nYet the mechanism and means by which to enhance T cell function are incompletely described , especially in the skin .\n\nIn this study , we use a murine model of immunotherapy to optimize cell-mediated immunity in the skin .\n\nWe show that in vitro-derived central but not effector memory-like T cells bring about rapid regression of skin-expressing cognate Ag as a transgene in keratinocytes .\n\nLocal inflammation induced by the TLR7 receptor agonist imiquimod subtly yet reproducibly decreases time to skin graft rejection elicited by central but not effector memory T cells in an immunodeficient mouse model .\n\nLocal CCL4 , a chemokine liberated by TLR7 agonism , similarly enhances central memory T cell function .\n\nIn this model , IL-2 facilitates the development in vivo of effector function from central memory but not effector memory T cells .\n\nIn a model of T cell tolerogenesis , we further show that adoptively transferred central but not effector memory T cells can give rise to successful cutaneous immunity , which is dependent on a local inflammatory cue in the target tissue at the time of adoptive T cell transfer .\n\nThus , adoptive T cell therapy efficacy can be enhanced if CD8(+) T cells with a central memory T cell phenotype are transferred , and IL-2 is present with contemporaneous local inflammation .", "output": "Tumor promoting inflammation" }, { "input": "Several germline single nucleotide polymorphisms ( SNPs ) have been identified in the POLB gene , but little is known about their cellular and biochemical impact .\n\nDNA Polymerase \\u03b2 ( Pol \\u03b2 ) , encoded by the POLB gene , is the main gap-filling polymerase involved in base excision repair ( BER ) , a pathway that protects the genome from the consequences of oxidative DNA damage .\n\nIn this study we tested the hypothesis that expression of the POLB germline coding SNP ( rs3136797 ) in mammalian cells could induce a cancerous phenotype .\n\nExpression of this SNP in both human and mouse cells induced double-strand breaks , chromosomal aberrations , and cellular transformation .\n\nFollowing treatment with an alkylating agent , cells expressing this coding SNP accumulated BER intermediate substrates , including single-strand and double-strand breaks .\n\nThe rs3136797 SNP encodes the P242R variant Pol \\u03b2 protein and biochemical analysis showed that P242R protein had a slower catalytic rate than WT , although P242R binds DNA similarly to WT .\n\nOur results suggest that people who carry the rs3136797 germline SNP may be at an increased risk for cancer susceptibility .", "output": "Genomic instability and mutation" }, { "input": "The number of renal cancers has increased over the last ten years and patient survival in advanced stages remains very poor .\n\nTherefore , new therapeutic approaches for renal cancer are essential .\n\nEnglerin A is a natural product with a very potent and selective cytotoxicity against renal cancer cells .\n\nThis makes it a promising drug candidate that may improve current treatment standards for patients with renal cancers in all stages .\n\nHowever , little is known about englerin A's mode of action in targeting specifically renal cancer cells .\n\nOur study is the first to investigate the biological mechanism of englerin A action in detail .\n\nWe report that englerin A is specific for renal tumor cells and does not affect normal kidney cells .\n\nWe find that englerin A treatment induces necrotic cell death in renal cancer cells but not in normal kidney cells .\n\nWe further show that autophagic and pyroptotic proteins are unaffected by the compound and that necrotic signaling in these cells coincided with production of reactive oxygen species and calcium influx into the cytoplasm .\n\nAs the first study to analyze the biological effects of englerin A , our work provides an important basis for the evaluation and validation of the compound's use as an anti-tumor drug .\n\nIt also provides a context in which to identify the specific target or targets of englerin A in renal cancer cells .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Regulation of poly(ADP-ribose) ( PAR ) synthesis and turnover is critical to determining cell fate after genotoxic stress .\n\nHyperactivation of PAR synthesis by poly(ADP-ribose) polymerase-1 ( PARP-1 ) occurs when cells deficient in DNA repair are exposed to genotoxic agents ; however , the function of this hyperactivation has not been adequately explained .\n\nHere , we examine PAR synthesis in mouse fibroblasts deficient in the base excision repair enzyme DNA polymerase \\u03b2 ( pol \\u03b2 ) .\n\nThe extent and duration of PARP-1 activation was measured after exposure to either the DNA alkylating agent , methyl methanesulfonate ( MMS ) , or to low energy laser-induced DNA damage .\n\nThere was strong DNA damage-induced hyperactivation of PARP-1 in pol \\u03b2 nullcells , but not in wild-type cells .\n\nIn the case of MMS treatment , PAR synthesis did not lead to cell death in the pol \\u03b2 null cells , but instead resulted in increased PARylation of the nonhomologous end-joining ( NHEJ ) protein Ku70 and increased association of Ku70 with PARP-1 .\n\nInhibition of the NHEJ factor DNA-PK , under conditions of MMS-induced PARP-1 hyperactivation , enhanced necrotic cell death .\n\nThese data suggest that PARP-1 hyperactivation is a protective mechanism triggering the classical-NHEJ DNA repair pathway when the primary alkylated base damage repair pathway is compromised .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Thrombospondin-1 is a matricellular protein with potent antitumour activities , the levels of which determine the fate of many different tumours , including renal carcinomas .\n\nHowever , the factors that regulate this protein remain unclear .\n\nIn renal carcinomas , hypoxic conditions enhance the expression of angiogenic factors that help adapt tumour cells to their hostile environment .\n\nTherefore , we hypothesized that anti-angiogenic factors should correspondingly be dampened .\n\nIndeed , we found that hypoxia decreased the thrombospondin-1 protein in several clear cell renal carcinoma cell lines ( ccRCC ) , although no transcriptional regulation was observed .\n\nFurthermore , we proved that hypoxia stimulates multiple signals that independently contribute to diminish thrombospondin-1 in ccRCC , which include a decrease in the activity of oxygen-dependent prolylhydroxylases ( PHDs ) and activation of the PI3K/Akt signalling pathway .\n\nIn addition , thrombospondin-1 regulation in hypoxia proved to be important for ccRCC cell migration and invasion .", "output": "Activating invasion and metastasis" }, { "input": "DNA damage induced by benzene and its metabolites is thought of as an important mechanism underlying benzene genotoxicity in chronic benzene poisoning ( CBP ) .\n\nTherefore , genetic variation in DNA repair genes may contribute to susceptibility to CBP in the exposed population .\n\nSince benzene-induced DNA damages include DNA adducts , we hypothesized that the polymorphisms of ERCC1 ( Excision repair cross complementation group 1 ) and ERCC2/XPD ( Excision repair cross complementation group 2/xeroderma pigmentosum group D ) are associated with the risk of CBP .\n\nA case-control study involving 102 benzene-poisoned patients and 204 none-benzene-poisoned controls occupationally exposed to benzene was carried out in the Northeast region of China .\n\nThe polymorphisms of codon 118 ( rs11615 ) and C8092A ( rs3212986 ) of ERCC1 , codon 751 ( rs13181 ) , 312 ( rs1799793 ) and 156 ( rs238406 ) of ERCC2/XPD were genotyped by TaqMan(\u00ae) Real-time PCR .\n\nThe results showed that individuals carrying the ERCC1 codon 118 TT genotype had an increased risk of CBP ( OR(adj)=3.390 ; 95%CI : 1.393-8.253 ; P=0.007 ) comparing with its CC genotype .\n\nAfter stratified by smoking , gender and exposure duration we found that the increased risk of CBP associated with the ERCC1 codon 118 TT genotype confined to nonsmokers ( OR=3.214 ; 95% CI : 1.359-7.601 ; P=0.006 ) , female ( OR=3.049 ; 95% CI : 1.235-7.529 ; P=0.013 ) and exposure duration> 12 years ( OR=3.750 ; 95% CI : 1.041-13.513 ; P=0.035 ) .\n\nSince ERCC1 and ERCC2/XPD are both located on chromosome 19q13.3 , haplotype analysis of all 5 SNPs was also conducted .\n\nHowever no correlations between the risks of CBP and other genotypes or haplotypes were found .\n\nTherefore , our findings suggest an important role of ERCC1 codon 118 polymorphisms for a biomarker to CBP in the Chinese occupational population .", "output": "Genomic instability and mutation" }, { "input": "The DNA repair protein Ku70 is a key player in chemoresistance to anticancer agents ( e.g. , etoposide ) or radioresistance .\n\nThe responses of different organs to radiation vary widely and likely depend on the cell population in the organs .\n\nPreviously , we established and characterized Ku70-deficient murine lung epithelial ( Ku70 -/- MLE ) cells and found that these cells are more sensitive than Ku70 +/- MLE cells ( control cells ) to X-irradiation , as determined by clonogenic survival assay ; however , the mechanism underlying this sensitivity remains unclear .\n\nIn this study , we examined the mechanism by which X-irradiation triggers the death of Ku70 -/- MLE cells .\n\nOur results showed that Ku70 -/- MLE cells were more sensitive to radiation-induced apoptosis than control cells , although X-irradiation activated caspase-3 and caspase-7 , and cleaved PARP in both cell lines .\n\nWe also examined the expression level of phosphorylated H2AX ( \\u03b3H2AX ) , which is a marker of DSB , and observed the phosphorylation of H2AX and the elimination of \\u03b3H2AX in both cell lines after X-irradiation .\n\nThe elimination in Ku70 -/- MLE cells was slower than that in control cells , suggesting that DSB repair activity in the Ku70 -/- MLE cells is lower than that in control cells .\n\nThese findings suggest that Ku70 might play a key role in the inhibition of apoptosis through the DSB repair pathway in lung epithelial cells .\n\nOur findings also suggest that these cell lines might be useful for the study of Ku70 functions and the Ku70-dependent DSB repair pathway in lung epithelial cells .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "The main feedback loop driving circadian rhythm in mice is controlled , in part , by the genes encoding the cryptochromes Cry1 and Cry2 .\n\nTargeted mutation of both Cry1 and Cry2 delay the early onset of tumor formation in p53-null mutant mice .\n\nFurthermore , Ras-transformed p53- and Cry-null mouse skin fibroblasts are more sensitive than p53 mutants to apoptotic cell death initiated by agents that activate either the intrinsic or the extrinsic apoptosis pathways .\n\nHere , we investigated the effect of Cry1 and Cry2 mutations on cell death by other genotoxic agents that generate alkylated bases , interstrand crosslinks , DNA-protein crosslinks , and double-strand breaks .\n\nBoth ultraviolet ( UV ) and the UV mimetic compound oxaliplatin and the radiomimetic compound doxorubicin promoted apoptosis by upregulating the tumor suppressor p73 .\n\nHowever , only the UV and oxaliplatin-induced upregulation of p73 mediated by the transcription factor Egr1 , but not the doxorubicin-induced upregulation mediated by the transcription factor E2F1 , was enhanced by Cry1/Cry2 double mutation .\n\nAccordingly , Egr1 downregulation reduced oxaliplatin-induced apoptosis , whereas E2F1 downregulation reduced doxorubicin-induced apoptosis .\n\nOur findings establish distinct roles for cryptochromes in intrinsic apoptosis induced by UV mimetic and radiomimetic agents .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "More effective treatments for metastatic lung cancer remain a pressing clinical need .\n\nIn this study , we identified migration inducting gene-7 ( MIG-7 ) protein as critical for COX-2/prostaglandin E2 ( PGE2)- and Akt/GSK-3\u03b2-dependent tumor invasion/metastasis .\n\nCOX-2/PGE2 activated EP4 to enhance Akt and GSK-3\u03b2 phosphorylation and \u03b2-catenin/T-cell factor/lymphoid enhancer factor signaling leading to MIG-7 upregulation .\n\nRNAi-mediated attenuation of MIG-7 blocked COX-2/PGE2- and Akt/GSK-3\u03b2-mediated migration/invasion effects .\n\nFurthermore , MIG-7 protein inhibited protein phosphatase 2A to sustain Akt/GSK-3\u03b2 phosphorylation and cancer-cell migration/invasion .\n\nCancer cells overexpressing MIG-7 exhibited increased expression of ZEB-1 and Twist in parallel with epithelial-mesenchymal transition , metastasis and cancer lethality .\n\nMIG-7 protein level positively correlated with advanced stages of human lung cancers .\n\nMIG-7 thus offers a theranostic target for cancer metastases arising from aberrant activation of the cellular COX-2/PGE2 and Akt/GSK-3\u03b2 signaling pathways .", "output": "Activating invasion and metastasis" }, { "input": "Ophiopogonin B ( OP-B ) is a bioactive component of Radix Ophiopogon Japonicus , which is often used in Chinese traditional medicine to treat pulmonary disease .\n\nHowever , whether or not OP-B has any potential antitumor activity has not been reported .\n\nHere , we show that the non-small cell lung cancer ( NSCLC ) cell lines NCI-H157 and NCI-H460 treated with OP-B grow more slowly and accumulate vacuoles in their cytoplasm compared to untreated control cells .\n\nFlow cytometric analysis showed that the cells were arrested in G0/G1 phase .\n\nNuclear morphology , Annexin-V/PI staining , and expression of cleaved caspase-3 all confirm that OP-B does not induce apoptosis .\n\nInstead , based on results from both transmission electron microscopy ( TEM ) and the expression of microtubule-associated protein 1 light chain 3-II ( LC3-II ) , we determined that OP-B treatment induced autophagy in both cell lines .\n\nNext , we examined the PI3K/Akt/mTOR signaling pathway and found that OP-B inhibited phosphorylation of Akt(Ser473 , Thr308 ) in NCI-H157 cells and also inhibited several key components of the pathway in NCI-H460 cells , such as p-Akt(Ser473 , Thr308 ) , p-p70S6K ( Thr389 ) .\n\nAdditionally , insulin-mediated activation of the PI3K/Akt/mTOR pathway provides evidence that activation of this pathway may correlate with induction of autophagy in H460 cells .\n\nTherefore , OP-B is a prospective inhibitor of PI3K/Akt and may be used as an alternative compound to treat NSCLC .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "It is well established that angiogenesis is the process of formation of new capillaries from pre-existing blood vessels .\n\nIt is a complex process , involving both pro- and anti-angiogenic factors , and plays a significant role in physiological and pathophysiological processes such as embryonic development , atherosclerosis , post-ischemic vascularization of the myocardium , tumor growth and metastasis , rheumatoid arthritis etc .\n\nThis is the first report of zinc oxide ( ZnO ) nanoflowers that show significant pro-angiogenic properties ( formation of new capillaries from pre-existing blood vessels ) , observed by in vitro and in vivo angiogenesis assays .\n\nThe egg yolk angiogenesis assay using ZnO nanoflowers indicates the presence of matured blood vessels formation .\n\nAdditionally , it helps to promote endothelial cell ( EA.hy926 cells ) migration in wound healing assays .\n\nFormation of reactive oxygen species ( ROS ) , especially hydrogen peroxide ( H(2)O(2))-a redox signaling molecule , might be the plausible mechanism for nanoflower-based angiogenesis .\n\nAngiogenesis by nanoflowers may provide the basis for the future development of new alternative therapeutic treatment strategies for cardiovascular and ischemic diseases , where angiogenesis plays a significant role .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "Tumor development requires angiogenesis and anti-angiogenic therapies have been introduced in the treatment of cancer .\n\nIn this context , heparan sulfate proteoglycans ( HSPGs ) emerge as interesting targets , owing to their function as co-receptors of major , pro-angiogenic factors .\n\nAccordingly , previous studies have suggested anti-tumor effects of heparin , i.e. over-sulfated HS , and various heparin mimetics ; however , a significant drawback is their unspecific mechanism of action and potentially serious side-effects related to their anticoagulant properties .\n\nHere , we have explored the use of human ScFv anti-HS antibodies ( \u03b1HS ) as a more rational approach to target HSPG function in endothelial cells ( ECs). \u03b1HS were initially selected for their recognition of HS epitopes localized preferentially to the vasculature of patient glioblastoma tumors , i.e. highly angiogenic brain tumors .\n\nUnexpectedly , we found that these \u03b1HS exhibited potent pro-angiogenic effects in primary human ECs. \u03b1HS were shown to stimulate EC differentiation , which was associated with increased EC tube formation and proliferation .\n\nMoreover , \u03b1HS supported EC survival under hypoxia and starvation , i.e. conditions typical of the tumor microenvironment .\n\nImportantly , \u03b1HS-mediated proliferation was efficiently counter-acted by heparin and was absent in HSPG-deficient mutant cells , confirming HS-specific effects .\n\nOn a mechanistic level , binding of \u03b1HS to HSPGs of ECs as well as glioblastoma cells was found to trigger p38 MAPK-dependent signaling resulting in increased proliferation .\n\nWe conclude that several \u03b1HS that recognize HS epitopes abundant in the tumor vasculature may elicit a pro-angiogenic response , which has implications for the development of antibody-based targeting of HSPGs in cancer .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Metastasis is a major cause of death of patients with malignant tumors .\n\nMatrix metalloproteinases ( MMPs ) are important for the migration and invasion of various types of cancer cell .\n\nPropofol is a known anesthetic agent , widely used for short-term anesthesia and for longer-term sedation .\n\nPropofol inhibits the proliferation of a variety of tumor cells , but there is no available information regarding propofol-inhibited migration and invasion of tumor cells in vitro .\n\nIn this study , we investigated the effects of propofol on the migration and invasion of human lung carcinoma A549 cells .\n\nWound healing assay and Boyden chamber assays indicated that propofol inhibited the migration and invasion of A549 cells in vitro .\n\nGelatin zymographic analysis showed the inhibitory effect of propofol on the activation of expression MMP-2 .\n\nWestern blot analysis also indicated that propofol suppressed the protein expiration of growth factor receptor-bound protein 2 ( GRB2 ) , Jun N-terminal kinases 1/2 ( p-JNK1/2 ) , p-p38 , MMP-2 and MMP-9 in A549 cells .\n\nResults from real-time PCR assay also showed that propofol inhibited the mRNA gene expression of MMP-2 , -7 and -9 , and enhanced that of tissue inhibitor of metalloproteinase 1 ( TIMP1 ) and TIMP2 in A549 cells .\n\nTaken together , these data show that propofol inhibits MMP-2 and -9 mRNA and protein expressions , resulting in suppression of lung cancer cell invasion and migration in vitro .", "output": "Activating invasion and metastasis" }, { "input": "OBJECTIVE To study the mechanism of interleukin 7/interleukin 7 receptor ( IL-7/IL-7R ) in promoting cell proliferation and inducing lymphangiogenesis of non-small cell lung cancer ( NSCLC ) in vivo and in vitro .\n\nMETHODS Immunohistochemical study for IL-7 , IL-7R , cyclin D1 and vascular endothelial growth factor-D ( VEGF-D ) was carried out in NSCLC tissues from 95 patients .\n\nThe relationship between IL-7/IL-7R expression and various parameters was analyzed .\n\nThe mechanism of IL-7/IL-7R in promoting cell proliferation and inducing lymphangiogenesis was studied by methylthiazolyldiphenyl-tetrazolium bromide , fluorescence-activated cell sorting , reverse transcriptase-PCR , Western blot , co-immunoprecipitation , chromatin immunoprecipitation and nude mice experiments with xenograft tumors .\n\nRESULTS IL-7 ( 63.2% , 60/95 ) , IL-7R ( 61.1% , 58/95 ) , cyclin D1 ( 52.6% , 50/95 ) and VEGF-D ( 58.9% , 56/95 ) showed that high level of expression in NSCLC .\n\nIL-7/IL-7R over-expression correlated with cyclin D1 expression ( P < 0.01 , P < 0.01 ) , VEGF-D expression ( P < 0.01 , P < 0.01 ) , increased lymphovascular density ( P = 0.005 , P = 0.013 ) , advanced clinical stage ( P = 0.008 , P = 0.005 ) and presence of lymph node metastasis ( P < 0.01 , P < 0.01 ) .\n\nIL-7/IL-7R could promote proliferation of A549 cell , increase cyclin D1 and VEGF-D expression , and enhance c-Fos/c-Jun expression and phosphorylation , resulting in formation of heterodimer .\n\nFurthermore , IL-7/IL-7R could induce binding of c-Fos/c-Jun to cyclin D1/VEGF-D promoters and regulate their transcription .\n\nIL-7/IL-7R could also promote proliferation and lymphangiogenesis of lung cancer xenograft tumors .\n\nCONCLUSIONS IL-7/IL-7R promotes c-Fos/c-Jun expression and activity in NSCLC .\n\nThis further facilitates cyclin D1 expression and accelerates proliferation of cells and VEGF-D-induced lymphovascular formation .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "OBJECTIVE To investigate the mutations of epidermal growth factor receptor ( EGFR ) in tumor tissue and pleural effusion in advanced non-small cell lung cancer ( NSCLC ) patients , and to analyze the relationship between EGFR mutations and the clinicopathologic characteristics .\n\nMETHODS Two-hundred and forty-one cases of formalin-fixed , paraffin-embedded tumor tissues and 14 paired pleural effusions from advanced NSCLC patients were collected .\n\nTwenty-nine different EGFR mutations in exons 18-21 were assessed by scorpions and amplification refractory mutation system ( scorpions ARMS ) using real time PCR .\n\nThe relationship between the EGFR mutations and clinical parameters was analyzed using statistical methods .\n\nEGFR mutation of 37 cases were detected with direct sequencing , and assessed the sensitivity , the specificity and the accuracy of scorpions ARMS .\n\nRESULTS EGFR somatic mutations were detected in 114 of 234 advanced NSCLC patients , with the mutation rate of 48.7% , including deletions in exon 19 in 65 patients and point mutation of L858R in exon 21 in 39 patients ; both accounting for 91.2% ( 104/114 ) of all types of EGFR mutations .\n\nThe test results of 14 paired pleural effusion specimens were entirely the same to the tissues .\n\nThe concordance rate of 2 different detection methods was 94.6% .\n\nMutation rate was higher in women ( 55.9% ) than in men ( 42.2% ) , and there was no difference in mutation rates between smokers and non-smokers ; patients in stage IIIB and stage IV ; adenocarcinoma and non-adenocarcinoma .\n\nCONCLUSIONS EGFR somatic mutations appear to occur frequently in Chinese .\n\nScorpions ARMS technology is a sensitive method to detect EGFR mutations and is suitable for screening patients who would likely respond to EGFR inhibitors therapy .", "output": "Genomic instability and mutation" }, { "input": "OBJECTIVE To explore the efficacy and side effects of icotinib hydrochloride in the treatment of patients with advanced non-small cell lung cancer ( NSCLC ) .\n\nMETHODS The efficacy and side effects of icotinib hydrochloride in treatment of 59 cases with stage IV NSCIC and followed-up from March 2009 to January 2012 were retrospectively analyzed .\n\nRESULTS Twenty seven patients ( 45.8% ) showed partial response ( PR ) , 17 patients ( 28.8% ) achieved SD , and 15 ( 25.4% ) had progressive disease .\n\nThe objective response rate ( ORR ) was 45.8% ( 27/59 ) , and disease control rate ( DCR ) was 74.6% ( 44/59 ) .\n\nAmong the 23 patients with EGFR mutation , ORR was 73.9% ( 17/23 ) , and DCR was 95.7% ( 22/23 ) .\n\nThirty six patients ( 61.0% ) achieved remission of symptoms to varying degrees .\n\nThe main symptoms relieved were cough , asthmatic suffocating , pain and hoarseness .\n\nThe major adverse events were mild skin rash ( 35.6% ) and diarrhea ( 15.3% ) .\n\nOthers were dry skin , nausea and stomach problems .\n\nThe efficacy of icotinib hydrochloride were related to the ECOG performance status , smoking history , EGFR mutation and rash significantly ( P < 0.05 ) .\n\nCONCLUSIONS Monotherapy with icotinib hydrochloride is effective and tolerable for patients with advanced NSCLC , especially with EGFR mutation .", "output": "Genomic instability and mutation" }, { "input": "Adhesion to the extracellular matrix ( ECM ) is critical for epithelial tissue homeostasis and function .\n\nECM detachment induces metabolic stress and programmed cell death via anoikis .\n\nECM-detached mammary epithelial cells are able to rapidly activate autophagy allowing for survival and an opportunity for re-attachment .\n\nHowever , the mechanisms controlling detachment-induced autophagy remain unclear .\n\nHere we uncover that the kinase PERK rapidly promotes autophagy in ECM-detached cells by activating AMP-activated protein kinase ( AMPK ) , resulting in downstream inhibition of mTORC1-p70(S6K) signaling .\n\nLKB1 and TSC2 , but not TSC1 , are required for PERK-mediated inhibition of mammalian target of rapamycinin MCF10A cells and mouse embryo fibroblast cells .\n\nImportantly , this pathway shows fast kinetics , is transcription-independent and is exclusively activated during ECM detachment , but not by canonical endoplasmic reticulum stressors .\n\nMoreover , enforced PERK or AMPK activation upregulates autophagy and causes luminal filling during acinar morphogenesis by perpetuating a population of surviving autophagic luminal cells that resist anoikis .\n\nHence , we identify a novel pathway in which suspension-activated PERK promotes the activation of LKB1 , AMPK and TSC2 , leading to the rapid induction of detachment-induced autophagy .\n\nWe propose that increased autophagy , secondary to persistent PERK and LKB1-AMPK signaling , can robustly protect cells from anoikis and promote luminal filling during early carcinoma progression.Oncogene advance online publication , 19 November 2012 ; doi:10.1038/onc.2012.512 .", "output": "Resisting cell death" }, { "input": "In response to genotoxic stress , a transient arrest in cell-cycle progression enforced by the DNA-damage checkpoint ( DDC ) signalling pathway positively contributes to genome maintenance .\n\nBecause hyperactivated DDC signalling can lead to a persistent and detrimental cell-cycle arrest , cells must tightly regulate the activity of the kinases involved in this pathway .\n\nDespite their importance , the mechanisms for monitoring and modulating DDC signalling are not fully understood .\n\nHere we show that the DNA-repair scaffolding proteins Slx4 and Rtt107 prevent the aberrant hyperactivation of DDC signalling by lesions that are generated during DNA replication in Saccharomyces cerevisiae .\n\nOn replication stress , cells lacking Slx4 or Rtt107 show hyperactivation of the downstream DDC kinase Rad53 , whereas activation of the upstream DDC kinase Mec1 remains normal .\n\nAn Slx4-Rtt107 complex counteracts the checkpoint adaptor Rad9 by physically interacting with Dpb11 and phosphorylated histone H2A , two positive regulators of Rad9-dependent Rad53 activation .\n\nA decrease in DDC signalling results from hypomorphic mutations in RAD53 and H2A and rescues the hypersensitivity to replication stress of cells lacking Slx4 or Rtt107 .\n\nWe propose that the Slx4-Rtt107 complex modulates Rad53 activation by a competition-based mechanism that balances the engagement of Rad9 at replication-induced lesions .\n\nOur findings show that DDC signalling is monitored and modulated through the direct action of DNA-repair factors .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Thymidine phosphorylase ( TYMP ) is an angiogenic factor that has potent chemotactic activity for endothelial cells and is involved in 5-fluorouracil ( 5-FU ) metabolism .\n\nIn colorectal cancer ( CRC ) , previous studies evaluating the relationship between TYMP expression and clinicopathological features have yielded inconsistent results .\n\nThe aim of this study was to investigate the prognostic value of TYMP , its association with other angiogenic factors , proliferation markers and , to our knowledge , for the first time its relationship with extracellular matrix components .\n\nMATERIALS AND METHODS Formalin-fixed , paraffin-embedded specimens from 97 patients with CRC were immunostained for TYMP , vascular endothelial growth factor ( VEGF ) , microvascular density ( CD34 ) , proliferation marker ( Ki-67 ) , proliferating cell nuclear antigen ( PCNA ) , p53 oncoprotein and extracellular matrix components ( collagen type IV , fibronectin , tenascin and laminin ) .\n\nSurvival curves were calculated with the Kaplan-Meier method .\n\nRESULTS Immunoreactivity was observed in the cytoplasm ( cyt ) and nucleus ( n ) of the tumor cells , as well in the stroma ( st ) , endothelium and tumor-associated macrophages .\n\nHigh TYMPcyt expression was observed in 7.2% of the cases , moderate in 22.7% and weak in 59.9% , while 10.3% were negative .\n\nHigh TYMPst expression was observed in 58.8% of the cases .\n\nTYMPcyt expression was correlated with the VEGF expression of tumor cells and VEGF expression of vessels ( p=0.014 and p=0.022 , respectively ) .\n\nTYMPst expression was correlated with VEGF expression and tenascin ( p=0.014 and p=0.011 , respectively ) .\n\nPatients with higher TYMPcyt expression had a more favorable overall survival ( p=0.041 ) in univariate analysis compared to patients without TYMP expression .\n\nCONCLUSION These findings suggest that TYMP plays an important role in angiogenesis , extracellular matrix remodeling and in the prognosis of patients with CRC , but further studies are needed to clearly define its role in CRC .", "output": "Inducing angiogenesis" }, { "input": "Olfactomedin 4 ( OLFM4 ) is highly expressed in gastrointestinal cancers and has an anti-apoptotic function .\n\nThe roles of OLFM4 in tumor growth and metastasis and how it functions in these processes remain elusive .\n\nWe investigated the function of OLFM4 in tumor growth and metastasis using B16F10 mouse melanoma cells as an experimental system .\n\nOur results showed that OLFM4 had no positive effect on cell viability or cell cycle progression in B16F10 cells .\n\nHowever , it significantly suppressed the tumorigenicity of B16F10 cells , i.e. , intradermal primary tumor growth and lung metastasis .\n\nOLFM4 also suppressed the migration and invasion of B16F10 cells in vitro .\n\nFor further insight into the mechanisms underlying OLFM4-mediated suppression of tumor progression , we examined the effect of OLFM4 on the expression of integrin and matrix metalloproteinase ( MMP ) , both of which are involved in tumor progression .\n\nOverexpression of OLFM4 clearly reduced the expression levels of integrin \u03b11 , integrin \u03b14 , integrin \u03b15 , integrin \u03b16 , and MMP9 .\n\nMoreover , forced expression of MMP9 attenuated the inhibitory activity of OLFM4 on migration and invasiveness .\n\nOur findings provide the experimental evidence that OLFM4 may function as a tumor suppressor and an anti-metastatic gene during tumor progression .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "Endothelial cells respond to hypoxic changes with resultant accumulation of several metabolites and switch over to angiogenic phenotype .\n\nAlthough certain intermediates of glycolytic and oxidative metabolic pathways have been known to affect angiogenesis , the effect of citrate , which accumulates in certain tumors , on angiogenesis is not known .\n\nTherefore , the effect of citrate on angiogenesis was studied using different model systems .\n\nIncreased vascularization in chorioallantoic membrane assay , increased endothelial sprouting in rat aortic rings , and increased expression of CD31 , E-selectin in endothelial cells suggested a possible proangiogenic effect of citrate .\n\nUpregulation of angiogenic factors such as vascular endothelial growth factor and fibroblast growth factor suggested that the effect of citrate involves modulation of expression of angiogenic growth factors .\n\nLY 294002 , an inhibitor of PI3K-Akt pathway , and wortmannin , an inhibitor of Akt pathway , reversed the effect of citrate in human umbilical vein endothelial cells .\n\nCitrate induced significant upregulation and activation of Akt in endothelial cells .\n\nRapamycin , an inhibitor of mTOR , also reversed the effect of citrate in human umbilical vein endothelial cells and sprouting of aortic rings suggesting that the angiogenic effect of citrate involves activation of PI3K-Akt-mTOR pathway .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "An epidemiological survey was conducted in the Seine estuary and in two smaller and relatively preserved estuaries on the French Atlantic coast in order to estimate the occurrence of liver lesions in European flounder , Platichthys flesus , and also to seek putative risk factors for the recorded pathologies .\n\nFour hundred and seventy-eight fish of both sexes and of different size ranges were sampled in the three studied areas , 338 of which in the Seine estuary .\n\nAll fish were examined for histopathological liver lesions , while DNA adducts and otoliths were analyzed on a subsample .\n\nFive categories of hepatic lesions were recorded with the following prevalence for the Seine estuary : 36.7 % inflammations , 8 % parasites ( mainly encysted nematodes ) , 6.5 % foci of cellular alteration ( FCA ) , 5.3 % foci of necrosis or regeneration ( FNR ) , and 1.5 % tumors .\n\nInflammation occurrence increased according to age , contrary to parasitic infestations and FCA which were more prevalent in young fish , notably those of <1 year old ( group 0 ) .\n\nTumors were only observed in females of more than two winters .\n\nFemales exhibited a higher prevalence of tumors ( 3.0 % ) and FCA ( 6.5 % ) than males ( 0 and 2.6 % , respectively ) .\n\nParasitic and infectious lesions and FNR were equally distributed in males and females .\n\nThe prevalence of FNR was also shown to vary according to sampling season , with significantly more occurrences of liver necrosis in the fish collected in summer than in spring .\n\nSpatial differences were observed with a higher occurrence of encysted parasites in flounders from the upper Seine estuary , while inflammations predominated in flounders living downstream .\n\nTemporal trends were also noted , with an increased prevalence of parasitic infestations , inflammations , and FCA in the 2002-2003 period in comparison to the 1996-1997 one .\n\nThe three flounder populations from the Seine estuary ( Normandy ) , Ster estuary ( Brittany ) , and Bay of Veys ( Normandy ) showed different spectra of hepatic lesions .\n\nFlounders from the Bay of Veys had relatively few liver lesions as compared to flounders from the two other estuaries .\n\nFlounders from the Ster estuary exhibited the highest prevalence of parasites ( 37.2 % ) and inflammations ( 51.1 % ) .\n\nFinally , FCA and liver tumors occurred at very similar levels in both flounder populations from the Seine and the Ster estuaries .\n\nGroup 0 flounders inhabiting the upper Seine estuary were more prone to parasitic and pre-neoplastic hepatic lesions and had higher levels of liver DNA adducts than the older ones living downstream .\n\nIt was postulated that group 0 European flounders may serve as valuable bioindicators for assessing the quality of estuarine waters and the health status of euryhaline fish populations .", "output": "Tumor promoting inflammation, Genomic instability and mutation" }, { "input": "Missense substitutions of uncertain clinical significance in the BRCA1 gene are a vexing problem in genetic counseling for women who have a family history of breast cancer .\n\nIn this study , we evaluated the functions of 29 missense substitutions of BRCA1 in two DNA repair pathways .\n\nRepair of double-strand breaks by homology-directed recombination ( HDR ) had been previously analyzed for 16 of these BRCA1 variants , and 13 more variants were analyzed in this study .\n\nAll 29 variants were also analyzed for function in double-strand break repair by the single-strand annealing ( SSA ) pathway .\n\nWe found that among the pathogenic mutations in BRCA1 , all were defective for DNA repair by either pathway .\n\nThe HDR assay was accurate because all pathogenic mutants were defective for HDR , and all nonpathogenic variants were fully functional for HDR .\n\nRepair by SSA accurately identified pathogenic mutants , but several nonpathogenic variants were scored as defective or partially defective .\n\nThese results indicated that specific amino acid residues of the BRCA1 protein have different effects in the two related DNA repair pathways , and these results validate the HDR assay as highly correlative with BRCA1-associated breast cancer .", "output": "Genomic instability and mutation" }, { "input": "A key requirement for the development of cancer immunotherapy is the identification of tumour-associated antigens that are differentially or exclusively expressed on the tumour and recognized by the host immune system .\n\nHowever , immune responses to such antigens are often muted or lacking due to the antigens being recognized as \" self \" , and further complicated by the tumour environment and regulation of immune cells within .\n\nIn an effort to circumvent the lack of immune responses to tumour antigens , we have devised a strategy to develop potential synthetic immunogens .\n\nThe strategy , termed mirror image phage display , is based on the concept of molecular mimicry as demonstrated by the idiotype/anti-idiotype paradigm in the immune system .\n\nHere as ' proof of principle ' we have selected molecular mimics of the well-characterised tumour associated antigen , the human mucin1 protein ( MUC1 ) from two different peptide phage display libraries .\n\nThe putative mimics were compared in structure and function to that of the native antigen .\n\nOur results demonstrate that several of the mimic peptides display T-cell stimulation activity in vitro when presented by matured dendritic cells .\n\nThe mimic peptides and the native MUC1 antigenic epitopes can cross-stimulate T-cells .\n\nThe data also indicate that sequence homology and/or chemical properties to the original epitope are not the sole determining factors for the observed immunostimulatory activity of the mimic peptides .", "output": "Avoiding immune destruction" }, { "input": "Colon cancer is the third most common malignant neoplasm in the world and it remains an important cause of death , especially in western countries .\n\nThe toxic environmental pollutant , 1 , 2-dimethylhydrazine ( DMH ) , is also a colon-specific carcinogen .\n\nTannic acid ( TA ) is reported to be effective against various types of chemically induced toxicity and also carcinogenesis .\n\nIn the present study , we evaluated the chemopreventive efficacy of TA against DMH induced colon toxicity in a rat model .\n\nEfficacy of TA against the colon toxicity was evaluated in terms of biochemical estimation of antioxidant enzyme activities , lipid peroxidation , histopathological changes and expression of early molecular markers of inflammation and tumor promotion .\n\nDMH treatment induced oxidative stress enzymes ( p<0.001 ) and an early inflammatory and tumor promotion response in the colons of Wistar rats .\n\nTA treatment prevented deteriorative effects induced by DMH through a protective mechanism that involved reduction of oxidative stress as well as COX-2 , i-NOS , PCNA protein expression levels and TNF-\u03b1(p<0.001) release .\n\nIt could be concluded from our results that TA markedly protects against chemically induced colon toxicity and acts plausibly by virtue of its antioxidant , anti-inflammatory and antiproliferative activities .", "output": "Tumor promoting inflammation" }, { "input": "Breast cancer causes death due to distant metastases in which tumor cells produce matrix metalloproteinase ( MMP ) enzymes which facilitate invasion .\n\nOleuropein , the main olive oil polyphenol , has anti-proliferative effects .\n\nThis study aimed to investigate the effect of oleuropein on the metastatic and anti-metastatic gene expression in the MDA human breast cancer cell line .\n\nWe evaluated the MMPs and TIMPs gene expression by semi-quantitative reverse transcriptase polymerase chain reaction ( RT-PCR ) in treated and untreated cells .\n\nThis study demonstrated that OL may induce anti-metastatic effects on human breast cancer cells .\n\nWe found that TIMP1,-3 , and -4 were over-expressed after all periods of incubation in treated cancer cells compared to untreated cells , while MMP2 and MMP9 genes were down-regulated , at least initially .\n\nTreatment of breast cancer cells with oleuropein could help in prevention of cancer metastasis by increasing the TIMPs and suppressing the MMPs gene expressions .", "output": "Activating invasion and metastasis" }, { "input": "Invasion and metastasis are the major causes of cancer-related death .\n\nPharmacological or therapeutic interventions such as chemoprevention of the progression stages of neoplastic development could result in substantial reduction in the incidence of cancer mortality .\n\n( -)-Epigallocatechin-3-gallate ( EGCG ) , a promising chemopreventive agent , has attracted extensive interest for cancer therapy utilizing its antioxidant , anti- proliferative and inhibitory effects on angiogenesis and tumor cell invasion .\n\nIn this study , we assessed the influence of EGCG on the proliferative potential of HeLa cells by cell viability assay and authenticated the results by nuclear morphological examination , DNA laddering assay and cell cycle analysis .\n\nFurther we analyzed the anti-invasive properties of EGCG by wound migration assay and gene expression of MMP-9 and TIMP-1 in HeLa cells .\n\nOur results indicated that EGCG induced growth inhibition of HeLa cells in a dose- and time- dependent manner .\n\nIt was observed that cell death mediated by EGCG was through apoptosis .\n\nInterestingly , EGCG effectively inhibited invasion and migration of HeLa cells and modulated the expression of related genes ( MMP-9 and TIMP-1 ) .\n\nThese results indicate that EGCG may effectively suppress promotion and progression stages of cervical cancer development .", "output": "Activating invasion and metastasis, Resisting cell death" }, { "input": "Senescence is a cellular response preventing tumorigenesis .\n\nThe Ras oncogene is frequently activated or mutated in human cancers , but Ras activation is insufficient to transform primary cells .\n\nIna search for cooperating oncogenes , we identify the lysine demethylase JMJD2A/KDM4A .\n\nWe show that JMJD2A functions as a negative regulator of Ras-induced senescence and collaborates with oncogenic Ras to promote cellular transformation by negatively regulating the p53 pathway .\n\nWe find CHD5 , a known tumor suppressor regulating p53 activity , as a target of JMJD2A .\n\nThe expression of JMJD2A inhibits Ras-mediated CHD5 induction leading to a reduced activity of the p53 pathway .\n\nIn addition , we show that JMJD2A is overexpressed inmouse and human lung cancers .\n\nDepletion of JMJD2A in the human lung cancer cell line A549 bearing an activated K-Ras allele triggers senescence .\n\nWe propose that JMJD2A is an oncogene that represents a target for Ras-expressing tumors .", "output": "Enabling replicative immortality" }, { "input": "The vacuolar H+-ATPase ( V-ATPase ) , a multisubunit proton pump , has come into focus as an attractive target in cancer invasion .\n\nHowever little is known about the role of V-ATPase in cell death and especially the underlying mechanisms remain mostly unknown .\n\nWe used the myxobacterial macrolide archazolid B , a potent inhibitor of the V-ATPase , as an experimental drug as well as a chemical tool to decipher V-ATPase related cell death signaling .\n\nWe found that archazolid induced apoptosis in highly invasive tumor cells at nanomolar concentrations which was executed by the mitochondrial pathway .\n\nPrior to apoptosis induction archazolid lead to the activation of a cellular stress response including activation of the hypoxia-inducible factor-1 alpha ( HIF1alpha ) and autophagy .\n\nAutophagy was induced at low concentrations of archazolid that do not alter pH in lysosomes and was shown by degradation of p62 or fusion of autophagosomes with lysosomes .\n\nHIF1alpha was induced due to energy stress shown by a decline of the ATP level and followed by a shut down of energy consuming processes .\n\nAs silencing HIF1alpha increases apoptosis , the cellular stress response was suggested to be a survival mechanism .\n\nWe conclude that archazolid leads to energy stress which activates adaptive mechanisms like autophagy mediated by HIF1alpha and finally leads to apoptosis .\n\nWe propose V-ATPase as a promising drugable target in cancer therapy caught up at the interplay of apoptosis , autophagy and cellular/metabolic stress .", "output": "Resisting cell death" }, { "input": "The omega-3 long chain polyunsaturated fatty acids , docosahexaenoic acid ( DHA ) and eicosapentaenoic acid ( EPA ) , elicit antiproliferative effects in cancer cell lines and in animal models .\n\nDietary DHA and EPA can be converted to their ethanolamine derivatives , docosahexaenoyl ethanolamine ( DHEA ) and eicosapentaenoyl ethanolamine ( EPEA ) , respectively ; however , few studies are reported on their anticancer activities .\n\nHere , we demonstrated that DHEA and EPEA were able to reduce cell viability in MCF-7 breast cancer cells whereas they did not elicit any effects in MCF-10A non-tumorigenic breast epithelial cells .\n\nSince DHA and EPA are ligands of Peroxisome Proliferator-Activated Receptor ( PPAR ) \u03b3 , we sought to determine whether PPAR\u03b3 may also mediate DHEA and EPEA actions .\n\nIn MCF-7 cells , both compounds enhanced PPAR\u03b3 expression , stimulated a PPAR response element-dependent transcription as confirmed by the increased expression of its target gene PTEN , resulting in the inhibition of AKT-mTOR pathways .\n\nBesides , DHEA and EPEA treatment induced phosphorylation of Bcl-2 promoting its dissociation from beclin-1 which resulted in autophagy induction .\n\nWe also observed an increase of beclin-1 and microtubule-associated protein 1 light chain 3 expression along with an enhanced autophagosomes formation as revealed by mono-dansyl-cadaverine staining .\n\nFinally , we demonstrated the involvement of PPAR\u03b3 in DHEA- and EPEA-induced autophagy by using siRNA technology and a selective inhibitor .\n\nIn summary , our data show that the two omega-3 ethanolamines exert antiproliferative effects by inducing autophagy in breast cancer cells highlighting their potential use as breast cancer preventive and/or therapeutic agents .\n\nJ. Cell .\n\nPhysiol. \u00a9 2012 Wiley Periodicals , Inc .", "output": "Resisting cell death" }, { "input": "Breast cancer is a heterogeneous disease at both the clinical and molecular levels .\n\nThis heterogeneity may give rise to different therapy responses .\n\nMolecular profiling has facilitated identification of signatures for stratifying patients who would potentially benefit from given therapies .\n\nPreviously , we reported on a subset of genes with the potential for predicting response of primary breast cancer to neoadjuvant chemotherapy .\n\nHerein , we report that patients with luminal ( estrogen receptor \u03b1 [ ER\u03b1]-expressing ) breast cancer were enriched for nonresponders .\n\nTo identify novel factors that contribute to the survival of breast cancer cells , a loss-of-function screen was performed with a subset of genes overexpressed in patients with disease resistant to chemotherapy .\n\nThis approach led us to identify protein phosphatase 1 , regulatory subunit 15B ( PPP1R15B ) as a factor with a potentially essential role in the survival of ER\u03b1-positive breast cancer cells .\n\nFunctional analyses showed that PPP1R15B depletion results in impaired proliferation due to unsuccessful transition of cells from G1 to S phase of the cell cycle , and apoptosis induction .\n\nMoreover , our data revealed a regulatory role for PPP1R15B in activating ER\u03b1 .\n\nFurthermore , a high level of PPP1R15B mRNA expression was associated with poor outcome following tamoxifen-based therapy .\n\nAccordingly , knockdown of PPP1R15B expression sensitized tamoxifen-resistant MCF-7 breast cancer cells to tamoxifen while reducing ER\u03b1 abundance in these cells .\n\nOur findings reveal a novel role for PPP1R15B in the survival and therapy response of ER\u03b1-positive breast cancer and may open new avenues for tumor subtype-specific therapeutic strategies in the era of personalized medicine .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Necrotic cells are known to activate the innate immune system and trigger inflammation by releasing damage associated molecular patterns ( DAMPs ) .\n\nHowever , how necrotic cells influence the induction of antigen-specific CD8(+) T cell-mediated adaptive immune responses under sterile conditions , in the absence of pathogen associated molecular patterns ( PAMPs ) , remains poorly understood .\n\nHere , we examined antigen-specific CD8(+) T-cell responses to primary sterile necrotic tumor cells both in vitro and in vivo .\n\nWe found that primary necrotic cells alone fail to generate CD8(+) T cell-dependent immune responses toward cell-associated antigens .\n\nWe show that necrotic cells trigger CD8(+) T-cell immunity only in the presence of PAMPs or analogs , such as p(dI-dC) and/or unmethylated CpG DNA .\n\nThe electroporation of tumor cells with these PAMPs prior to necrosis induction triggered antigen-specific CD8(+) T-cell responses through a TLR9/MyD88-dependent pathway .\n\nIn addition , we found that necrotic cells contain factors that can block the cross-priming of CD8(+) T cells even under non-sterile conditions and can serve as a possible mechanism of immunosuppression .\n\nThese results suggest that antigen-specific CD8(+) T-cell responses to primary necrotic tumor cells can be induced in the presence of PAMPs and thus have a substantial impact on the development of antitumor vaccination strategies .", "output": "Tumor promoting inflammation, Avoiding immune destruction, Resisting cell death" }, { "input": "The anthracycline doxorubicin is a widely used effective anti-cancer drug .\n\nHowever , its application and dosage are severely limited due to its cardiotoxicity .\n\nThe exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood .\n\nEven less is known about the impact of doxorubicin treatment on vascular damage .\n\nWe found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells ( VSMC ) .\n\nWe observed that expression of urokinase receptor ( uPAR ) was upregulated in response to doxorubicin .\n\nFurthermore , the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence .\n\nOn the contrary , uPAR overexpression promoted VSMC senescence .\n\nWe further found that proteasomal degradation of telomeric repeat binding factor 2 ( TRF2 ) mediates doxorubicin-induced VSMC senescence .\n\nOur results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism .\n\nTherefore , VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process .", "output": "Enabling replicative immortality" }, { "input": "Benzyl isothiocyanate ( BITC ) is a dietary chemopreventive agent that inhibits the growth of various human cancer cells by causing apoptotic cell death .\n\nIn this study , we demonstrate that BITC not only induces apoptosis but also induces autophagy in human hormone-sensitive ( Rv1 ) and -refractory ( PC3 ) prostate cancer cells .\n\nIn BITC-treated cells , the induction of autophagy was detected by monitoring the processing of an autophagy marker protein , microtubule-associated protein 1 light chain 3 ( LC3 ) , the aggregation of LC3 into granular structures and the formation of acidic organelles .\n\nInhibition of autophagy using 3-methyladenine increased BITC-induced apoptosis , whereas the administration of caspase inhibitor suppressed BITC-induced cell death .\n\nOur data also showed that BITC inhibits mammalian target of rapamycin ( mTOR ) kinase activity in a dose-dependent manner .\n\nThe expression of phospho-mTOR ( Ser2481 ) , an indicator of mTOR intrinsic catalytic activity , and phospho-UNC-51-like kinase 1 ( Ser757 ) , a direct substrate of mTOR , were decreased in BITC-treated cells .\n\nHowever , the increased expression of phospho-mTOR ( Ser2448 ) , phospho-AKT ( Ser473 ) and antiapoptotic Bcl-2 were detected only in PC3 cells at later stages of BITC treatment .\n\nCollectively , our results show that BITC induces a protective autophagy response in Rv1 and PC3 cells through inhibition of the mTOR signaling pathway .\n\nActivation of the AKT survival pathway was only observed in PC3 cells , representing a resistance mechanism of advanced prostate cancer upon BITC treatment .\n\nThese findings could potentially contribute to the beneficial effect of BITC in prostate cancer treatments .", "output": "Resisting cell death" }, { "input": "OBJECTIVE To explore the clinical significance of miRNA-216a expression in pancreatic cancer .\n\nMETHODS Fourteen patients with pancreatic cancer undergoing pancreaticoduodenectomy and 6 patients with benign pancreas lesions were examined for miR-216a expressions in the tumor or lesion tissues using Agilent Human miRNA Microarray ( V12.0 ) .\n\nThe relationship between miR-216a expressions and the clinicopathological features of the patients was analyzed .\n\nRESULTS The expression of miRNA-216a was significantly lower in pancreatic cancer than in benign pancreas lesions ( P=0.000 ) .\n\nThe expression of miRNA-216a was significantly correlated with the T stage of the tumor ( P=0.002 ) , but not with the patients ' age , gender , smoking status , tumor stage , lymph node metastases , distant metastasis , tumor differentiation , nerve invasion , vessel invasion or serum CA19-9 level ( P>0.05 ) .\n\nCONCLUSIONS The down-regulated expression of miR-216a in pancreatic cancer suggests the involvement of miR-216a in the tumorigenesis and development of pancreatic cancer. miR-216a may potentially serve as a novel tumor marker and also a prognostic factor for pancreatic cancer .", "output": "Activating invasion and metastasis" }, { "input": "PURPOSE We did this retrospective study to explore the association between epidermal growth factor receptor ( EGFR ) mutation and clinical features in postoperative recurrent female non-small-cell lung cancer ( NSCLC ) .\n\nMATERIALS AND METHODS We reviewed clinical data on 86 female patients who had postoperative recurrent disease between December 1992 and July 2007 .\n\nThe start of tyrosine kinase inhibitor therapy was treated as a censoring event .\n\nCorresponding surgical specimens of primary tumors were used to test for EGFR mutations .\n\nRESULTS Thirty patients presented with local recurrence and distant recurrence was identified in 56 .\n\nThirty-four of the 86 patients ( 40% ) harbored EGFR mutations .\n\nPatients with distant recurrence were more likely to have EGFR mutations than patients with local recurrence ( 48% versus 23% ; P = 0.024 ) .\n\nOn multivariate analysis , distant recurrence was associated with a high frequency of EGFR mutations ( OR , 3.3 ; P = 0.028 ) .\n\nSurvival analysis showed poor survival of patients with mutated EGFR ( HR , 2.3 ; P = 0.017 ) or with non-adenocarcinoma histology ( HR , 3.3 ; P = 0.001 ) .\n\nCONCLUSION The association between recurrence pattern and EGFR mutation status was suggested in recurrent female NSCLC patients .\n\nIn addition , our data indicate unfavorable disease process of EGFR mutated tumors .\n\nFurther studies need to be conducted to validate these findings .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "Polycyclic aromatic hydrocarbons ( PAHs ) likely play a role in many cancers even in never-smokers .\n\nWe tried to find a model to explain the relationship between variation in PAH-related DNA adduct levels among people with similar exposures , multiple genetic polymorphisms in genes related to metabolic and repair pathways , and nucleotide excision repair ( NER ) capacity .\n\nIn 111 randomly selected female never-smokers from the Golestan Cohort Study in Iran , we evaluated 21 SNPs in 14 genes related to xenobiotic metabolism and 12 SNPs in eight DNA repair genes .\n\nNER capacity was evaluated by a modified comet assay , and aromatic DNA adduct levels were measured in blood by32P-postlabeling .\n\nMultivariable regression models were compared by Akaike's information criterion ( AIC ) .\n\nAromatic DNA adduct levels ranged between 1.7 and 18.6 per 10(8) nucleotides ( mean : 5.8 \u00b1 3.1 ) .\n\nDNA adduct level was significantly lower in homozygotes for NAT2 slow alleles and ERCC5 non-risk-allele genotype , and was higher in the MPO homozygote risk-allele genotype .\n\nThe sum of risk alleles in these genes significantly correlated with the log-adduct level ( r = 0.4 , p < 0.001 ) .\n\nCompared with the environmental model , adding Phase I SNPs and NER capacity provided the best fit , and could explain 17% more of the variation in adduct levels .\n\nNER capacity was affected by polymorphisms in the MTHFR and ERCC1 genes .\n\nFemale non-smokers in this population had PAH-related DNA adduct levels three to four times higher than smokers and occupationally-exposed groups in previous studies , with large inter-individual variation which could best be explained by a combination of Phase I genes and NER capacity .", "output": "Genomic instability and mutation" }, { "input": "Metabolic reprogramming of cancer cells provides energy and multiple intermediates critical for cell growth .\n\nHypoxia in tumors represents a hostile environment that can encourage these transformations .\n\nWe report that glycogen metabolism is upregulated in tumors invivo and in cancer cells invitro in response to hypoxia .\n\nInvitro , hypoxia induced an early accumulation of glycogen , followed bya gradual decline .\n\nConcordantly , glycogen synthase ( GYS1 ) showed a rapid induction , followed by a later increase of glycogen phosphorylase ( PYGL ) .\n\nPYGL depletion and the consequent glycogen accumulation led to increased reactive oxygen species ( ROS ) levels that contributed to a p53-dependent induction of senescence and markedly impaired tumorigenesis invivo .\n\nMetabolic analyses indicated that glycogen degradation by PYGL is important for the optimal function of the pentose phosphate pathway .\n\nThus , glycogen metabolism is a key pathway induced by hypoxia , necessary for optimal glucose utilization , which represents a targetable mechanism of metabolic adaptation .", "output": "Tumor promoting inflammation, Enabling replicative immortality" }, { "input": "Pyruvate kinase M2 ( PKM2 ) is upregulated in multiple cancer types and contributes to the Warburg effect by unclear mechanisms .\n\nHere we demonstrate that EGFR-activated ERK2 binds directly to PKM2 Ile429/Leu431 through the ERK2 docking groove and phosphorylates PKM2 at Ser37 , but does not phosphorylate PKM1 .\n\nPhosphorylated PKM2 Ser37 recruits PIN1 for cis-trans isomerization of PKM2 , which promotes PKM2 binding to importin \\u03b15 and translocating to the nucleus .\n\nNuclear PKM2 acts as a coactivator of \\u03b2-catenin to induce c-Myc expression , resulting in the upregulation of GLUT1 , LDHA and , in a positive feedback loop , PTB-dependent PKM2 expression .\n\nReplacement of wild-type PKM2 with a nuclear translocation-deficient mutant ( S37A ) blocks the EGFR-promoted Warburg effect and brain tumour development in mice .\n\nIn addition , levels of PKM2 Ser37 phosphorylation correlate with EGFR and ERK1/2 activity in human glioblastoma specimens .\n\nOur findings highlight the importance of nuclear functions of PKM2 in the Warburg effect and tumorigenesis .", "output": "Sustaining proliferative signaling, Cellular energetics" }, { "input": "Telomerase is a ribonucleoprotein consisting of a catalytic subunit , the telomerase reverse transcriptase , TERT , and an integrally associated RNA , TR , which contains a template for the synthesis of short repetitive G-rich DNA sequences at the ends of telomeres .\n\nTelomerase can repetitively reverse transcribe its short RNA template , acting processively to add multiple telomeric repeats onto the same DNA substrate .\n\nThe contribution of enzyme processivity to telomere length regulation in human cells is not well characterized .\n\nIn cancer cells , under homeostatic telomere length-maintenance conditions , telomerase acts processively , while under nonequilibrium conditions , telomerase acts distributively on the shortest telomeres .\n\nTo investigate the role of increased telomerase processivity on telomere length regulation in human cells with limited lifespan that are dependent on human TERT ( hTERT ) for lifespan extension and immortalization , we mutated the leucine at position 866 in the reverse transcriptase C motif of hTERT to a tyrosine ( L866Y ) , which is the amino acid found at a similar position in HIV-1 reverse transcriptase .\n\nWe report that , similar to the previously reported ' gain of function ' Tetrahymena telomerase mutant ( L813Y ) , the human telomerase variant displays increased processivity. hTERT-L866Y , like wild-type hTERT can immortalize and extend the lifespan of limited lifespan cells .\n\nMoreover , hTERT-L866Y expressing cells display heterogenous telomere lengths , telomere elongation , multiple telomeric signals indicative of fragile sites and replicative stress , and an increase in short telomeres , which is accompanied by telomere trimming events .\n\nOur results suggest that telomere length and homeostasis in human cells may be regulated by telomerase enzyme processivity .", "output": "Enabling replicative immortality" }, { "input": "PURPOSE : Previous studies have shown that the novel microtubule poison , JG-03-14 , which binds to the colchicine binding site of tubulin , has the capacity to kill breast tumor cells primarily through the promotion of autophagy .\n\nThe current work was designed to determine whether autophagy was , in fact , the primary mode of action as well as susceptibility to JG-03-14 in two additional tumor cell models , the B16/F10 murine melanoma cell line and the HCT-116 human colon cancer cell line .\n\nMETHODS : Drug cytotoxicity was monitored based on viable cell number and clonogenic survival .\n\nApoptosis was assessed by DAPI staining , the TUNEL assay and/or FACS analysis .\n\nAutophagy was monitored based on staining with acridine orange , redistribution and punctuation of RFP-LC3 and electron microscopy as well as p62 degradation .\n\nSenescence was evaluated based on \u03b2-galactosidase staining and alterations in cell morphology .\n\nDrug effects were also evaluated in a murine model of B16/F10 cells that localizes to the lungs while peripheral neuropathy was assessed by three complementary behavioral assays .\n\nRESULTS : Both HCT-116 colon cancer cells and B16/F10 melanoma cells were sensitive to JG-03-14 in that the drug demonstrated tumor cell killing .\n\nHowever , there was minimal induction of apoptosis .\n\nIn contrast , there was clear evidence for autophagy and autophagic flux while the residual surviving cells appeared to be in a state of irreversible senescence .\n\nInhibition of drug-induced autophagy in either the melanoma cells or the colon carcinoma cells was only slightly protective as the cells instead died by apoptosis .\n\nJG-03-14 reduced the size of tumor nodules in mice lungs ; furthermore , the drug did not promote peripheral neuropathy .\n\nCONCLUSIONS : Taken together with evidence for its actions as a vascular disrupting agent , these observations support the potential utility of JG-03-14 to effectively treat malignancies that might be resistant to conventional chemotherapy through evasion of apoptosis .", "output": "Enabling replicative immortality, Resisting cell death" }, { "input": "Arginine deprivation is a promising strategy for treating ASS-negative malignant tumors including melanoma .\n\nHowever , autophagy can potentially counteract the effectiveness of this treatment by acting as a pro-survival pathway .\n\nBy combining tumor necrosis factor-related apoptosis-inducing ligand ( TRAIL ) with arginine deprivation using ADI-PEG20 ( pegylated arginine deiminase ) , we achieved enhanced apoptosis and accelerated cell death in melanoma cell lines .\n\nThis implies a switch from autophagy to apoptosis .\n\nIn our current investigation , we found that TRAIL could induce the cleavage of two key autophagic proteins , Beclin-1 and Atg5 , in the combination treatment .\n\nUsing specific inhibitors for individual caspases , we found that caspase-8 inhibitor could completely abolish the cleavage .\n\nFurthermore , caspase-8 inhibitor was able to fully reverse the enhanced cytotoxicity induced by TRAIL .\n\nInhibitors for caspase-3 , 6 , 9 , and 10 were able to block the cleavage of these two autophagic proteins to some extent and correspondingly rescue cells from the cytotoxicity of the combination of TRAIL and arginine deprivation .\n\nIn contrast , calpain inhibitor could not prevent the cleavage of either Beclin-1 or Atg5 , and was unable to prevent cell death .\n\nOverall , our data indicate that the cleavage of Beclin-1 and Atg5 by TRAIL-initiated caspase activation is one of the mechanisms that lead to the enhancement of the cytotoxicity in the combination treatment .", "output": "Resisting cell death" }, { "input": "Oncogene-induced senescence can provide a protective mechanism against tumour progression .\n\nHowever , production of cytokines and growth factors by senescent cells may contribute to tumour development .\n\nThus , it is unclear whether induction of senescence represents a viable therapeutic approach .\n\nHere , using a mouse model with orthotopic implantation of metastatic melanoma tumours taken from 19 patients , we observed that targeting aurora kinases with MLN8054/MLN8237 impaired mitosis , induced senescence and markedly blocked proliferation in patient tumour implants .\n\nImportantly , when a subset of tumour-bearing mice were monitored for tumour progression after pausing MLN8054 treatment , 50% of the tumours did not progress over a 12-month period .\n\nMechanistic analyses revealed that inhibition of aurora kinases induced polyploidy and the ATM/Chk2 DNA damage response , which mediated senescence and a NF-\u03baB-related , senescence-associated secretory phenotype ( SASP ) .\n\nBlockade of IKK\u03b2/NF-\u03baB led to reversal of MLN8237-induced senescence and SASP .\n\nResults demonstrate that removal of senescent tumour cells by infiltrating myeloid cells is crucial for inhibition of tumour re-growth .\n\nAltogether , these data demonstrate that induction of senescence , coupled with immune surveillance , can limit melanoma growth .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "The environmental arylamine mutagens are implicated in the etiology of various sporadic human cancers .\n\nArylamine-modified dG lesions were studied in two fully paired 11-mer duplexes with a -G*CN- sequence context , in which G* is a C8-substituted dG adduct derived from fluorinated analogs of 4-aminobiphenyl ( FABP ) , 2-aminofluorene ( FAF ) or 2-acetylaminofluorene ( FAAF ) , and N is either dA or dT .\n\nThe FABP and FAF lesions exist in a simple mixture of ' stacked ' ( S ) and ' B-type ' ( B ) conformers , whereas the N-acetylated FAAF also samples a ' wedge ' ( W ) conformer .\n\nFAAF is repaired three to four times more efficiently than FABP and FAF .\n\nA simple A- to -T polarity swap in the G*CA/G*CT transition produced a dramatic increase in syn-conformation and resulted in 2- to 3-fold lower nucleotide excision repair ( NER ) efficiencies in Escherichia coli .\n\nThese results indicate that lesion-induced DNA bending/thermodynamic destabilization is an important DNA damage recognition factor , more so than the local S/B-conformational heterogeneity that was observed previously for FAF and FAAF in certain sequence contexts .\n\nThis work represents a novel 3'-next flanking sequence effect as a unique NER factor for bulky arylamine lesions in E. coli .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Phosphorylation of the H2AX histone is an early indicator of DNA double-strand breaks and of the resulting DNA damage response .\n\nIn the present study , we assessed the expression and prognostic significance of \\u03b3-H2AX in a cohort of 96 patients with operable non-small cell lung carcinoma .\n\nMETHODS Ninety-six paraffin-embedded specimens of non-small cell lung cancer patients were examined .\n\nAll patients underwent radical thoracic surgery of primary tumor ( lobectomy or pneumonectomy ) and regional lymph node dissection. \\u03b3-H2AX expression was assessed by standard immunohistochemistry .\n\nFollow-up was available for all patients ; mean duration of follow-up was 27.50 \u00b1 14.07 months ( range 0.2-57 months , median 24 months ) .\n\nRESULTS Sixty-three patients ( 65.2% ) died during the follow-up period .\n\nThe mean survival time was 32.2 \u00b1 1.9 months ( 95% confidence interval [ CI ] : 28.5-35.8 months ; median 30.0 months ) ; 1- , 2- and 3-year survival rates were 86.5% \u00b1 3.5% , 57.3% \u00b1 5.1% , and 37.1% \u00b1 5.4% , respectively .\n\nLow \\u03b3-H2AX expression was associated with a significantly better survival as compared with those having high \\u03b3-H2AX expression ( 35.3 months for low \\u03b3-H2AX expression versus 23.2 months for high \\u03b3-H2AX expression , P = 0.009 ; hazard ratio [ HR ] 1.95 , 95% CI : 1.15-3.30 ) .\n\nFurther investigation with multivariate Cox proportional hazards regression analysis revealed that high expression of \\u03b3-H2AX remained an independent prognostic factor of shorter overall survival ( HR 2.15 , 95% CI : 1.22-3.79 , P = 0.026 ) .\n\nA combined p53/\\u03b3-H2AX analysis was performed , and we found that the p53 low/\\u03b3-H2AX low phenotype was associated with significantly better survival compared with all other phenotypes .\n\nCONCLUSION Our study is the first to demonstrate that expression of \\u03b3-H2AX detected by immunohistochemistry may represent an independent prognostic indicator of overall survival in patients with non-small cell lung cancer .\n\nFurther studies are needed to confirm our results .", "output": "Genomic instability and mutation" }, { "input": "Identifying prognostic factors for osteosarcoma ( OS ) aids in the selection of patients who require more aggressive management .\n\nCD133 has been found to be a prognostic factor of certain tumor types .\n\nHowever , the association between CD133 expression and the prognosis of OS remains unknown .\n\nIn this study , we analyzed the association of CD133 expression in OS with clinical factors and overall survival , and further investigated its potential role in metastasis in vitro .\n\nWe found CD133 expression in 65.7% ( 46/70 ) of OS samples using immunohistochemistry , and it was positively correlated with lung metastasis analyzed by Chi-square test ( P=0.002 ) and shorter overall survival time using the Kaplan-Meier method compared by log-rank test ( P=0.000 ) .\n\nMultivariate analysis showed that CD133 expression was an independent prognostic factor of patients with OS .\n\nTo test for direct participation of CD133 , we separated CD133(+) and CD133(-) cells in the MG63 cell line using magnetic-activated cell sorting and found that CD133(+) cells were more active in migration by scratch wound-healing assay and invasion by Matrigel invasion assay compared with CD133(-) cells .\n\nElevated mRNA expression of the stemness gene octamer-binding transcription factor 4 ( Oct-4 ) and NANOG , and the metastasis-related receptor C-X-C chemokine receptor type 4 ( CXCR4 ) were also found in CD133(+) cells by reverse transcription-polymerase chain reaction .\n\nThus , expression of CD133 was correlated with lung metastasis and poor prognosis in OS patients .\n\nCD133(+) cells may be a type of cancer stem cell with high expression of self-renewal capacity and metastasis-related genes .", "output": "Activating invasion and metastasis" }, { "input": "Ursolic acid ( UA ) is a pentacyclic triterpenoid with promising cancer chemopreventive properties .\n\nA better understanding of the mechanisms underlying anticancer activity of UA is needed for further development as a clinically useful chemopreventive agent .\n\nHere , we found that both endoplasmic reticulum ( ER ) stress and autophagy were induced by UA in MCF-7 human breast cancer cells .\n\nSurprisingly , ER stress was identified as an effect rather than a cause of UA-induced autophagy .\n\nAutophagy-dependent ER stress protected the cells from UA-induced apoptosis through EIF2AK3-mediated upregulation of MCL1 .\n\nActivation of MAPK1/3 but not inhibition of MTOR pathway contributed to UA-induced cytoprotective autophagy in MCF-7 cells .\n\nOur findings uncovered a novel cellular mechanism involved in the anticancer activity of UA , and also provided a useful model to study biological significance and mechanisms of autophagy-mediated ER stress .", "output": "Resisting cell death" }, { "input": "Endoplasmic reticulum ( ER ) stress induces both autophagy and apoptosis yet the molecular mechanisms and pathways underlying the regulation of these two cellular processes in cells undergoing ER stress remain less clear .\n\nWe report here that eukaryotic elongation factor-2 kinase ( EEF2K ) is a critical controller of the ER stress-induced autophagy and apoptosis in tumor cells .\n\nDDIT4 , a stress-induced protein , was required for transducing the signal for activation of EEF2K under ER stress .\n\nWe further showed that phosphorylation of EEF2K at Ser398 was essential for induction of autophagy , while phosphorylation of the kinase at Ser366 and Ser78 exerted an inhibitory effect on autophagy .\n\nSuppression of the ER stress-activated autophagy via silencing of EEF2K aggravated ER stress and promoted apoptotic cell death in tumor cells .\n\nMoreover , inhibiting EEF2K by either RNAi or NH125 , a small molecule inhibitor of the enzyme , rendered tumor cells more sensitive to curcumin and velcade , two anticancer agents that possess ER stress-inducing action .\n\nOur study indicated that the DDIT4-EEF2K pathway was essential for inducing autophagy and for determining the fate of tumor cells under ER stress , and suggested that inhibiting the EEF2K-mediated autophagy can deteriorate ER stress and lead to a greater apoptotic response , thereby potentiating the efficacy of the ER stress-inducing agents against cancer .", "output": "Resisting cell death" }, { "input": "That a knock-in mouse harboring a dominant-negative thyroid hormone receptor ( TR)-\u03b2 ( Thrb ) mutation develops metastatic thyroid cancer strongly suggests the involvement of TR\u03b2 in carcinogenesis .\n\nEpigenetic silencing of the THRB gene is common in human cancers .\n\nThe aim of the present study was to determine how DNA methylation affected the expression of the THRB gene in differentiated thyroid cancer ( DTC ) and how reexpression of the THRB gene attenuated the cancer phenotypes .\n\nWe used methylation-specific PCR to examine the expression and promoter methylation of the THRB gene in DTC tissues .\n\nThyroid cancer cells with hypermethylated THRB were treated with the demethylating agents 5'-aza-2'-deoxycytidine ( 5'-aza-CdR ) and zebularine to evaluate their impact on the cancer cell phenotypes .\n\nTHRB mRNA expression in DTC was 90% lower than in normal controls , and this decrease was associated with a higher tumor/lymph node staging .\n\nThe promoter methylation level of the THRB gene had a significant negative correlation with the expression level of the THRB gene .\n\nTreatment of FTC-236 cells with 5'-aza-CdR or zebularine induced reexpression of the THRB gene and inhibited cell proliferation and migration .\n\nFTC-236 cells stably expressing TR\u03b2 exhibited lower cell proliferation and migration through inhibition of \u03b2-catenin signaling pathways compared with FTC-236 without TR\u03b2. 5'-Aza-CdR also led to suppression of tumor growth in an in vivo xenograft model using FTC-236 cells consistent with the cell-based studies .\n\nThese finding indicate that TR\u03b2 is a tumor suppressor and could be tested as a potential therapeutic target .", "output": "Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "This study aimed to analyze the role of endothelial progenitor cell ( EPC)-derived angiogenic factors and chemokines in the multistep process driving angiogenesis with a focus on the recently discovered macrophage migration inhibitory factor ( MIF)/chemokine receptor axis .\n\nPrimary murine and murine embryonic EPCs ( eEPCs ) were analyzed for the expression of angiogenic/chemokines and components of the MIF/CXC chemokine receptor axis , focusing on the influence of hypoxic versus normoxic stimulation .\n\nHypoxia induced an upregulation of CXCR2 and CXCR4 but not CD74 on EPCs and triggered the secretion of CXCL12 , CXCL1 , MIF , and vascular endothelial growth factor ( VEGF ) .\n\nThese factors stimulated the transmigration activity and adhesive capacity of EPCs , with MIF and VEGF exhibiting the strongest effects under hypoxia .\n\nMIF- , VEGF- , CXCL12- , and CXCL1-stimulated EPCs enhanced tube formation , with MIF and VEGF exhibiting again the strongest effect following hypoxia .\n\nTube formation following in vivo implantation utilizing angiogenic factor-loaded Matrigel plugs was only promoted by VEGF .\n\nColoading of plugs with eEPCs led to enhanced tube formation only by CXCL12 , whereas MIF was the only factor which induced differentiation towards an endothelial and smooth muscle cell ( SMC ) phenotype , indicating an angiogenic and differentiation capacity in vivo .\n\nSurprisingly , CXCL12 , a chemoattractant for smooth muscle progenitor cells , inhibited SMC differentiation .\n\nWe have identified a role for EPC-derived proangiogenic MIF , VEGF and MIF receptors in EPC recruitment following hypoxia , EPC differentiation and subsequent tube and vessel formation , whereas CXCL12 , a mediator of early EPC recruitment , does not contribute to the remodeling process .\n\nBy discerning the contributions of key angiogenic chemokines and EPCs , these findings offer valuable mechanistic insight into mouse models of angiogenesis and help to define the intricate interplay between EPC-derived angiogenic cargo factors , EPCs , and the angiogenic target tissue .", "output": "Inducing angiogenesis" }, { "input": "There are contradictory observations about the different radiosensitivities of cancer stem cells and cancer non-stem cells .\n\nTo resolve these contradictory observations , we studied radiosensitivities by employing breast cancer stem cell ( CSC)-like MDA-MB231 and MDA-MB453 cells as well as their corresponding non-stem cells .\n\nCSC-like cells proliferate without differentiating and have characteristics of tumor-initiating cells [ 1 ] .\n\nThese cells were exposed to \\u03b3-rays ( 1.25-8.75 Gy ) and survival curves were determined by colony formation .\n\nA final slope , D(0) , of the survival curve for each cell line was determined to measure radiosensitivity .\n\nThe D(0) of CSC-like and non-stem MDA-MB-453 cells were 1.16 Gy and 1.55 Gy , respectively .\n\nSimilar results were observed in MDA-MB-231 cells ( 0.94 Gy vs. 1.56 Gy ) .\n\nAfter determination of radiosensitivity , we investigated intrinsic cellular determinants which influence radiosensitivity including cell cycle distribution , free-radical scavengers and DNA repair .\n\nWe observed that even though cell cycle status and antioxidant content may contribute to differential radiosensitivity , differential DNA repair capacity may be a greater determinant of radiosensitivity .\n\nUnlike non-stem cells , CSC-like cells have little/no sublethal damage repair , a low intracellular level of ataxia telangiectasia mutated ( ATM ) and delay of \\u03b3-H2AX foci removal ( DNA strand break repair ) .\n\nThese results suggest that low DNA repair capacity is responsible for the high radiosensitivity of these CSC-like cells .", "output": "Genomic instability and mutation" }, { "input": "We have already shown that IL-10 plays an important role in immunosuppression and metastatic dissemination in the rat B-cell lymphoma L-TACB model .\n\nIt was suggested that the up-regulation of IL-10 production and IL-10 receptor ( IL-10R ) expression would be part of the transition from primary tumor to metastatic phenotype and that IL-10 , besides its immunosuppressive activity , may act as a growth factor for metastatic L-TACB cells .\n\nThe treatment of L-TACB-bearing rats with a single low-dose cyclophosphamide decreased IL-10 production , reverted immunosuppression and induced the immunologic rejection of tumor metastasis without any effect on primary tumor growth .\n\nOur current aim was to investigate the effects of cyclophosphamide on the expression of IL-10 and IL-10R on primary and metastatic L-TACB cells .\n\nConsidering that cyclophosphamide is a prodrug , we used mafosfamide , a compound that yields in vitro the same active metabolites as cyclophosphamide does in vivo .\n\nMafosfamide induced down-regulation of IL-10 production and IL-10R expression on metastatic cells and , concomitantly , inhibited metastatic cell proliferation .\n\nWe suggest that mafosfamide would inhibit the regulatory loop mediated by the IL-10/IL-10R system and , as a consequence , metastatic cell proliferation .\n\nThese results may have a considerable impact on the design of new therapies for metastatic lymphomas .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "When the cell cycle is arrested , even though growth-promoting pathways such as mTOR are still active , then cells senesce .\n\nFor example , induction of either p21 or p16 arrests the cell cycle without inhibiting mTOR , which , in turn , converts p21/p16-induced arrest into senescence ( geroconversion ) .\n\nHere we show that geroconversion is accompanied by dramatic accumulation of cyclin D1 followed by cyclin E and replicative stress .\n\nWhen p21 was switched off , senescent cells ( despite their loss of proliferative potential ) progressed through S phase , and levels of cyclins D1 and E dropped .\n\nMost cells entered mitosis and then died , either during mitotic arrest or after mitotic slippage , or underwent endoreduplication .\n\nNext , we investigated whether inhibition of mTOR would prevent accumulation of cyclins and loss of mitotic competence in p21-arrested cells .\n\nBoth nutlin-3 , which inhibits mTOR in these cells , and rapamycin suppressed geroconversion during p21-induced arrest , decelerated accumulation of cyclins D1 and E and decreased replicative stress .\n\nWhen p21 was switched off , cells successfully progressed through both S phase and mitosis .\n\nAlso , senescent mouse embryonic fibroblasts ( MEFs ) overexpressed cyclin D1 .\n\nAfter release from cell cycle arrest , senescent MEFs entered S phase but could not undergo mitosis and did not proliferate .\n\nWe conclude that cellular senescence is characterized by futile hyper-mitogenic drive associated with mTOR-dependent mitotic incompetence .", "output": "Sustaining proliferative signaling, Enabling replicative immortality" }, { "input": "Approximately 50% of melanomas require oncogenic B-RAF(V600E) signaling for proliferation , survival , and metastasis , and the use of highly selective B-RAF inhibitors has yielded remarkable , although short-term , clinical responses .\n\nReactivation of signaling downstream of B-RAF is frequently associated with acquired resistance to B-RAF inhibitors , and the identification of B-RAF targets may therefore provide new strategies for managing melanoma .\n\nIn this report , we applied whole-genome expression analyses to reveal that oncogenic B-RAF(V600E) regulates genes associated with epithelial-mesenchymal transition in normal cutaneous human melanocytes .\n\nMost prominent was the B-RAF-mediated transcriptional repression of E-cadherin , a keratinocyte-melanoma adhesion molecule whose loss is intimately associated with melanoma invasion and metastasis .\n\nHere we identify a link between oncogenic B-RAF , the transcriptional repressor Tbx3 , and E-cadherin .\n\nWe show that B-RAF(V600E) induces the expression of Tbx3 , which potently represses E-cadherin expression in melanocytes and melanoma cells .\n\nTbx3 expression is normally restricted to developmental embryonic tissues and promoting cell motility , but it is also aberrantly increased in various cancers and has been linked to tumor cell invasion and metastasis .\n\nWe propose that this B-RAF/Tbx3/E-cadherin pathway has a critical role in promoting the metastasis of B-RAF-mutant melanomas .", "output": "Activating invasion and metastasis" }, { "input": "Gastrointestinal stromal tumor ( GIST ) is a prototype of mutant KIT oncogene-driven tumor .\n\nProlonged tyrosine kinase inhibitor ( TKI ) treatment may result in a resistant phenotype through acquired secondary KIT mutation .\n\nHeat shock protein 90 ( HSP90AA1 ) is a chaperone protein responsible for protein maturation and stability , and KIT is a known client protein of HSP90AA1 .\n\nInhibition of HSP90AA1 has been shown to destabilize KIT protein by enhancing its degradation via the proteasome-dependent pathway .\n\nIn this study , we demonstrated that NVP-AUY922 ( AUY922 ) , a new class of HSP90AA1 inhibitor , is effective in inhibiting the growth of GIST cells expressing mutant KIT protein , the imatinib-sensitive GIST882 and imatinib-resistant GIST48 cells .\n\nThe growth inhibition was accompanied with a sustained reduction of both total and phosphorylated KIT proteins and the induction of apoptosis in both cell lines .\n\nSurprisingly , AUY922-induced KIT reduction could be partially reversed by pharmacological inhibition of either autophagy or proteasome degradation pathway .\n\nThe blockade of autophagy alone led to the accumulation of the KIT protein , highlighting the role of autophagy in endogenous KIT turnover .\n\nThe involvement of autophagy in endogenous and AUY922-induced KIT protein turnover was further confirmed by the colocalization of KIT with MAP1LC3B- , acridine orange- or SQSTM1-labeled autophagosome , and by the accumulation of KIT in GIST cells by silencing either BECN1 or ATG5 to disrupt autophagosome activity .\n\nTherefore , the results not only highlight the potential application of AUY922 for the treatment of KIT-expressing GISTs , but also provide the first evidence for the involvement of autophagy in endogenous and HSP90AA1 inhibitor-induced KIT degradation .", "output": "Resisting cell death" }, { "input": "Prohibiting angiogenesis is an important therapeutic approach for fighting cancer and other angiogenic related diseases .\n\nResearch focused on proteins that regulate abnormal angiogenesis has attracted intense interest in both academia and industry .\n\nSuch proteins are able to target several angiogenic factors concurrently , thereby increasing the possibility of therapeutic success .\n\nAquaporin-1 ( AQP1 ) is a water channel membrane protein that promotes tumour angiogenesis by allowing faster endothelial cell migration .\n\nIn this study we test the hypothesis that AQP1 inhibition impairs tumour growth in a mouse model of melanoma .\n\nAfter validating the inhibitor efficacy of two different AQP1 specific siRNAs in cell cultures , RNA interference experiments were performed by intratumoural injections of AQP1 siRNAs in mice .\n\nAfter 6 days of treatment , AQP1 siRNA treated tumours showed a 75 % reduction in volume when compared to controls .\n\nAQP1 protein level , in AQP1 knockdown tumours , was around 75 % that of the controls and was associated with a significant 40 % reduced expression of the endothelial marker , Factor VIII .\n\nImmunofluorescence analysis of AQP1 siRNA treated tumours showed a significantly lower microvessel density .\n\nTime course experiments demonstrated that repeated injections of AQP1 siRNA over time are effective in sustaining the inhibition of tumour growth .\n\nFinally , we have confirmed the role of AQP1 in sustaining an active endothelium during angiogenesis and we have shown that AQP1 reduction causes an increase in VEGF levels .\n\nIn conclusion , this study validates AQP1 as a pro-angiogenic protein , relevant for the therapy of cancer and other angiogenic-related diseases such as psoriasis , endometriosis , arthritis and atherosclerosis .", "output": "Inducing angiogenesis" }, { "input": "Bisphenol A is a monomer used in the production of polycarbonate plastics , epoxy resins , and flame retardants .\n\nIt is an endocrine disruptor with a variety of other effects , including genotoxicity .\n\nOxidative metabolism of bisphenol A yields electophilic bisphenol A-3,4-quinone ( BPAQ ) , which may cause genotoxicity .\n\nTo determine the genotoxic potential of bisphenol A , the mechanism of the reaction between the BPAQ and deoxyadenosine ( dA ) was studied in detail .\n\nThe most probable reaction pathway was determined using quantum chemical methods .\n\nOur results demonstrate that the rate limiting step is Michael addition between BPAQ and dA , the main product being the unstable N7-modified adduct that rapidly undergoes depurination .\n\nIn addition , our calculations provide strong evidence for protonation of the adducts prior to depurination , which indicates pH dependence of the reaction .\n\nThe calculated activation barrier for Michael addition is 28.7 kcal/mol , indicating that the reaction with dA is very slow .\n\nComparison with the activation energy of 23.1 kcal/mol for the corresponding deoxyguanosine reaction indicates that most of the DNA damage by BPAQ will occur at the guanine site .\n\nThe detoxification reactions with glutathione compete with reactions between BPAQ and DNA .\n\nThe calculated free energy of activation for the reaction with glutathione is significantly lower than that for the corresponding reaction with dA .\n\nThis indicates that BPAQ will preferably react with glutathione and will only react with DNA when the level of glutathione in the cell is low .", "output": "Genomic instability and mutation" }, { "input": "BACKGROUND Cellular and clinical sensitivity to ionizing radiation ( IR ) is determined by DNA double-strand breaks ( DSB ) repair .\n\nHere , we investigate the molecular mechanism underlying the extreme response of a head and neck tumor case ( SKX ) to standard radiotherapy .\n\nMETHODS Immunofluorescence ( IF ) was used for the assessment of DSB repair , Western blot and real-time PCR for protein and mRNA expression , respectively .\n\nRESULTS SKX cells exhibited a pronounced radiosensitivity associated with numerous residual \\u03b3-H2AX foci after IR .\n\nThis was not associated with lacking canonical repair proteins .\n\nSKX cells did not express any ATM protein .\n\nAccordingly , immunoblotting revealed no ATM kinase activity toward substrates such as p-SMC1 , p-CHK2 and p-KAP1 .\n\nSequencing of all 66 exons of ATM showed no mutation .\n\nATM mRNA level was moderately reduced , which could be reverted by 5'-Aza-C treatment but without restoring protein levels .\n\nImportantly , we demonstrated a post-transcriptional regulation in SKX cells via 6-fold enhanced levels of miR-421 , which targets the 3'-UTR of ATM mRNA .\n\nTransfection of SKX cells with either anti-miR-421 inhibitor or a microRNA-insensitive ATM vector recovered ATM expression and abrogated the hyper-radiosensitivity .\n\nCONCLUSION This is the first report describing microRNA-mediated down-regulation of ATM leading to clinically manifest tumor radiosensitivity .", "output": "Genomic instability and mutation" }, { "input": "Autophagy , or autophagocytosis , is a selective intracellular degradative process involving the cell's own lysosomal apparatus .\n\nAn essential component in cell development , homeostasis , repair and resistance to stress , autophagy may result in either cell death or survival .\n\nThe targeted region of the cell is sequestered within a membrane structure , the autophagosome , for regulation of the catabolic process .\n\nA key factor in both autophagosome formation and autophagosome maturation is a protein encoded by the ultraviolet irradiation resistance-associated gene ( UVRAG ) .\n\nConversely , the serine/threonine-specific protein kinase B ( PKB , also known as Akt ) , which regulates survival in various cancers , inhibits autophagy through mTOR activation .\n\nWe found that Akt1 may also directly inhibit autophagy by down-regulating UVRAG both in a 293T transient transfection system and breast cancer cells stably expressing Akt1 .\n\nThe UVRAG with mutations at putative Akt1-phosphorylation sites were still inhibited by Akt1 , and dominant-negative Akt1 also inhibited UVRAG expression , suggesting that Akt1 down-regulates UVRAG by a kinase activity-independent mechanism .\n\nWe showed that Akt1 overexpression in MDA-MB-231 breast cancer cells down-regulated UVRAG transcription .\n\nCells over-expressing Akt1 were more resistant than control cells to ultraviolet light-induced autophagy and exhibited the associated reduction in cell viability .\n\nLevels of the autophagosome indicator protein LC3B-II and mRFP-GFP-LC3 were reduced in cells that over-expressing Akt1 .\n\nInhibiting Akt1 by siRNA or reintroducing UVRAG gene rescued the level of LC3B-II in UV-irradiation .\n\nAltogether , these data suggest that Akt1 may inhibit autophagy by decreasing UVRAG expression , which also sensitizes cancer cells to UV irradiation .", "output": "Resisting cell death" }, { "input": "Phyllanthus urinaria is widely used as anti-inflammatory , antiviral , antibacterial , and anti-hepatotoxic medicines in almost every tropical country .\n\nHowever , scientific evidence supporting its use in cancer metastasis is limited , particularly osteosarcoma .\n\nWe investigated the effect of P. urinaria extract ( PUE ) on cell viability , invasion , and migration in the human osteosarcoma Saos-2 cell line , and looked at the impact of PUE on several relevant proteases and signaling pathways .\n\nThis study demonstrates that PUE , at a range of concentrations ( from 0 to 100 \u03bcg/ml ) , concentration-dependently inhibited the migration/invasion capacities of Saos-2 without cytotoxic effects .\n\nZymographic and western blot analyses revealed that PUE inhibited the urokinase-type plasminogen activator ( u-PA ) and matrix metalloproteinase-2 ( MMP-2 ) enzyme activity , as well as protein expression .\n\nWestern blot analysis also showed that PUE inhibits phosphorylation of ERK1/2 and Akt .\n\nTesting of mRNA level , quantitative real-time PCR , and promoter assays evaluated the inhibitory effects of PUE on u-PA expression in Saos-2 cells .\n\nThe chromatin immunoprecipitation ( ChIP ) assay was reactive to the transcription protein SP-1 , which was inhibited by PUE .\n\nIn conclusion , PUE suppresses human osteosarcoma Saos-2 cell invasion and migration by transcriptionally inhibiting u-PA via ERK and Akt signaling pathways .\n\nTherefore , PUE produces anti-metastatic activity in Saos-2 cells .", "output": "Activating invasion and metastasis" }, { "input": "Many molecular mechanisms contribute to the development of doxorubicin resistance and different cancers can express wide and diverse arrays of drug-resistance genes .\n\nThe aim of this study was to identify the changes in gene expression associated with the development of doxorubicin resistance in MCF7 breast cancer cell line .\n\nThe doxorubicin resistant MCF7 cell line was developed by stepwise selection of MCF7 cells and was tested using the MTT assay .\n\nThe alterations in gene expression were examined using the real-time based PCR array .\n\nThe findings showed an up-regulation of many phase I/II metabolizing genes , specifically , the CYP1A1 and the CYP1A2 that were up-regulated by 206- and 96-fold respectively .\n\nDrug efflux pump genes were also up-regulated profoundly .\n\nTOP2A was strongly down-regulated by 202-fold .\n\nMany other changes were observed in genes crucial for cell cycle , apoptosis and DNA repair .\n\nThe findings of this project imply that the development of doxorubicin resistance is a multi-factorial process .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Resisting cell death" }, { "input": "Von Hippel-Lindau ( VHL ) is a tumor suppressor that negatively regulates the production of angiogenic factors .\n\nMutations in the VHL gene cause VHL syndrome , which is characterized by highly vascularized tumors .\n\nHere we report a c.464T>A mutation of the VHL gene in three patients with hemangioblastoma from a Chinese family .\n\nThis mutation was not reported previously and was absent in the unaffected family members .\n\nThe mutation is predicted to cause Val to Glu substitution at VHL protein residue 155 in a conserved region .\n\nPrevious biochemical studies demonstrated that residue Val-155 was critical for VHL protein binding to chaperonin TRiC/CCT , an essential step for proper VHL protein folding .\n\nOur finding of naturally occurring VHL V155E mutation in patients with VHL syndrome supports the functional importance of Val-155 residue in VHL protein and illustrates the diversity of VHL gene defects underlying VHL syndrome .", "output": "Genomic instability and mutation" }, { "input": "The \u03b22-adrenergic receptor ( \u03b22AR ) mediates the effects of chronic stress in several neoplasms , however , \u03b22AR signaling is impaired by hypoxia in various tissues .\n\nWhile hypoxia is a common feature significant in the progression of solid tumors , little is known about the effect of hypoxia on \u03b22AR signaling in the tumor microenvironment .\n\nPreviously , it has been reported that the systemic administration of mesenchymal stem cells ( MSCs ) increased the engraftment and metastatic colonization of rat osteosarcoma ( OS ) cells .\n\nIn the current study , the effect of MSCs on the hypoxia-induced desensitization of the \u03b22AR in OS cells was investigated .\n\nEpinephrine , norepinephrine and isoproterenol increased the cellular proliferation of the rat OS cell line COS1NR and rat MSCs in a dose-dependent and \u03b22AR antagonist-sensitive manner .\n\nWhile isoproterenol had significant proliferative effects on MSCs under normoxic and hypoxic conditions , COS1NR cells did not respond under hypoxic conditions .\n\nA sensitivity assay for the \u03b22AR revealed that hypoxia impaired the sensitivity of COS1NR cells , whereas hypoxia did not affect MSCs .\n\nAn immunoassay revealed no significant change in the expression of hypoxia-inducible factor-1\u03b1 ( HIF1\u03b1 ) in COS1NR cells , whilst an immunoassay demonstrated a 15% increase in MSCs following isoproterenol stimulation .\n\nIn COS1NR cells co-cultured with MSCs under hypoxic conditions , isoproterenol caused a significant increase in proliferation and this effect was inhibited by an anti-interleukin ( IL)-6 antibody .\n\nA tumor formation assay in syngeneic rats revealed that the systemic administration of MSCs enhances the growth of OS and the effect of MSCs was inhibited by IL-6 neutralization .\n\nIn conclusion , MSCs are resistant to the hypoxia-induced desensitization to \u03b22AR .\n\nHypoxia caused a siginificant desensitization of the \u03b22AR in COS1NR cells alone , whereas MSCs may support tumor progression through cellular interactions .", "output": "Sustaining proliferative signaling" }, { "input": "AIM To investigate the role of delta-like ligand 4 ( DLL4 ) in the angiogenesis of high-grade malignant glioma .\n\nMATERIALS AND METHODS DLL4 expression and microvessel density ( MVD ) were detected by immunohistochemistry in 51 human high-grade malignant glioma tissue samples .\n\nThe vessel maturation index ( VMI ) was calculated as the percentage of a-smooth muscle actin ( a-SMA)-positive vessels in relation to the amount of CD31-positive vessels .\n\nDouble fluorescent immunostaining for CD31 and EphrinB2 or EphB4 was performed to identify the arterial ( EphrinB2 ) or venous ( EphB4 ) origins of glioma microvessels .\n\nRESULTS Strong immunostaining of DLL4 and a positive correlation of DLL4 with the MVD were observed in high-grade malignant gliomas .\n\nThe VMI of the DLL4-positive group was significantly higher than that of the DLL4-negative group .\n\nHowever , no significant association was found between DLL4 and EphrinB2 or EphB4 in high-grade gliomas .\n\nCONCLUSION DLL4 may be an important regulator for vessel proliferation and maturation in human high-grade malignant gliomas .", "output": "Inducing angiogenesis" }, { "input": "Many viruses subvert the host cell's ability to mount and complete various DNA damage responses ( DDRs ) after infection .\n\nHCMV infection of permissive fibroblasts activates host DDRs at the time of viral deposition and during replication , but the DDRs remain uncompleted without arrest or apoptosis .\n\nWe believe this was in part due to partitioning of the damage response and double strand break repair components .\n\nAfter extraction of soluble proteins , the localization of these components fell into three groups : specifically associated with the viral replication centers ( RCs ) , diffused throughout the nucleoplasm and excluded from the RCs .\n\nOthers have shown that cells are incapable of processing exogenously introduced damage after infection .\n\nWe hypothesized that the inability of the cells to process damage might be due to the differential association of repair components within the RCs and , in turn , potentially preferential repair of the viral genome and compromised repair of the host genome .\n\nTo test this hypothesis we used multiple strategies to examine repair of UV-induced DNA damage in mock and virus-infected fibroblasts .\n\nComet assays indicated that repair was initiated , but was not completed in infected cells .\n\nQuantitative analysis of immunofluorescent localization of cyclobutane pyrimidine dimers ( CPDs ) revealed that after 24 h of repair , CPDs were significantly reduced in viral DNA , but not significantly changed in the infected host DNA .\n\nTo further quantitate CPD repair , we developed a novel dual-color Southern protocol allowing visualization of host and viral DNA simultaneously .\n\nCombining this Southern methodology with a CPD-specific T4 endonuclease V alkaline agarose assay to quantitate repair of adducts , we found efficient repair of CPDs from the viral DNA but not host cellular DNA .\n\nOur data confirm that NER functions in HCMV-infected cells and almost exclusively repairs the viral genome to the detriment of the host's genome .", "output": "Genomic instability and mutation" }, { "input": "Genomic instability drives tumorigenesis , but how it is initiated in sporadic neoplasias is unknown .\n\nIn early preneoplasias , alterations at chromosome fragile sites arise due to DNA replication stress .\n\nA frequent , perhaps earliest , genetic alteration in preneoplasias is deletion within the fragile FRA3B/FHIT locus , leading to loss of Fhit protein expression .\n\nBecause common chromosome fragile sites are exquisitely sensitive to replication stress , it has been proposed that their clonal alterations in cancer cells are due to stress sensitivity rather than to a selective advantage imparted by loss of expression of fragile gene products .\n\nHere , we show in normal , transformed , and cancer-derived cell lines that Fhit-depletion causes replication stress-induced DNA double-strand breaks .\n\nUsing DNA combing , we observed a defect in replication fork progression in Fhit-deficient cells that stemmed primarily from fork stalling and collapse .\n\nThe likely mechanism for the role of Fhit in replication fork progression is through regulation of Thymidine kinase 1 expression and thymidine triphosphate pool levels ; notably , restoration of nucleotide balance rescued DNA replication defects and suppressed DNA breakage in Fhit-deficient cells .\n\nDepletion of Fhit did not activate the DNA damage response nor cause cell cycle arrest , allowing continued cell proliferation and ongoing chromosomal instability .\n\nThis finding was in accord with in vivo studies , as Fhit knockout mouse tissue showed no evidence of cell cycle arrest or senescence yet exhibited numerous somatic DNA copy number aberrations at replication stress-sensitive loci .\n\nFurthermore , cells established from Fhit knockout tissue showed rapid immortalization and selection of DNA deletions and amplifications , including amplification of the Mdm2 gene , suggesting that Fhit loss-induced genome instability facilitates transformation .\n\nWe propose that loss of Fhit expression in precancerous lesions is the first step in the initiation of genomic instability , linking alterations at common fragile sites to the origin of genome instability .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Enabling replicative immortality" }, { "input": "K-Ras dependent non-small cell lung cancer ( NSCLC ) cells are ' addicted ' to basal autophagy that reprograms cellular metabolism in a lysosomal-sensitive manner .\n\nHere we demonstrate that the xenophagy-associated kinase TBK1 drives basal autophagy , consistent with its known requirement in K-Ras-dependent NSCLC proliferation .\n\nFurthermore , basal autophagy in this context is characterised by sequestration of the xenophagy cargo receptor Ndp52 and its paralogue Tax1bp1 , which we demonstrate here to be a bona fide cargo receptor .\n\nAutophagy of these cargo receptors promotes non-canonical NF-\u03baB signalling .\n\nWe propose that this TBK1-dependent mechanism for NF-\u03baB signalling contributes to autophagy addiction in K-Ras driven NSCLC .", "output": "Resisting cell death" }, { "input": "Excessive exposure to solar UVA and UVB radiation is widely considered to cause skin cancers such as squamous cell carcinoma and basalioma .\n\nDirect UVB damage to skin cell DNA as well as UV-induced chronic skin inflammation , accelerated keratinocyte proliferation , inhibited apoptosis , and immunosuppression seem to underlie the UV-induced carcinogenesis .\n\nAlso , UVB induces cytochrome P450 subfamilies ( CYP1A1 and CYP1B1 ) involved in metabolic activation of organic pro-carcinogens and their conversion to ultimate carcinogens .\n\nHere , the effects of several glycosylated and non-glycosylated plant polyphenols ( verbascoside , resveratrol , polydatin , rutin , and quercetin ) on the inflammatory , apoptotic , metabolic , and proliferative responses of cultured human epidermal keratinocytes ( HEK ) to non-cytotoxic doses of solar-simulated UVA+UVB and chemical mediators of UV signalling in HEK , 6-formylindolo[3,2-b]carbazole and squalene isolated from photo-oxidized skin surface lipids ( SSL ) , were evaluated .\n\nWe showed that the stilbenes and quercetin being exposed to UV were photo-destroyed within a short period of time , while verbascoside and rutin were photo-stable .\n\nWhen SSL were exposed to UV , the stilbenes and quercetin remarkably accelerated photo-oxidation of alpha-tocopherol , squalene , and cholesterol fractions , whilst verbascoside protected them .\n\nVerbascoside invariably inhibited molecular pathways in HEK leading to inflammatory cytokine expression ( NFkappaB and EGFR/ERK phosphorylation ) , and cell proliferation ( EGFR nuclear translocation ) , and displayed a stimulus-specific effect on the metabolic axis aryl hydrocarbon receptor ( AhR)-CYP1A1/CYP1B1 .\n\nBy contrast , the stilbenes inhibited UV-connected inflammatory cytokines excluding IL-8 , but they prevalently stimulated NFkappaB , EGFR nuclear translocation and the AhR-CYP pathway .\n\nWe conclude that , among the PPs investigated , verbascoside does interfere with multiple UV-sensitive signalling in HEK in a way that it could have a major impact on skin cancer chemoprevention .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Lesion-specific enzymes repair different forms of DNA damage , yet all lesions elicit the same checkpoint response .\n\nThe common intermediate required to mount a checkpoint response is thought to be single-stranded DNA ( ssDNA ) , coated by replication protein A ( RPA ) and containing a primer-template junction .\n\nTo identify factors important for initiating the checkpoint response , we screened for genes that , when overexpressed , could amplify a checkpoint signal to a weak allele of chk1 in fission yeast .\n\nWe identified Ast1 , a novel member of the XPG-related family of endo/exonucleases .\n\nAst1 promotes checkpoint activation caused by the absence of the other XPG-related nucleases , Exo1 and Rad2 , the homologue of Fen1 .\n\nEach nuclease is recruited to DSBs , and promotes the formation of ssDNA for checkpoint activation and recombinational repair .\n\nFor Rad2 and Exo1 , this is independent of their S-phase role in Okazaki fragment processing .\n\nThis XPG-related pathway is distinct from MRN-dependent responses , and each enzyme is critical for damage resistance in MRN mutants .\n\nThus , multiple nucleases collaborate to initiate DNA damage responses , highlighting the importance of these responses to cellular fitness .", "output": "Genomic instability and mutation" }, { "input": "MicroRNAs ( miRNAs ) are considered to be regulators of various biological processes in cancers , including the epithelial to mesenchymal transition ( EMT ) , which is a key factor in cancer metastasis .\n\nIn this study , we aimed to clarify the potential roles of miR-490-3p in hepatocellular carcinoma ( HCC ) cells .\n\nUsing real-time quantitative RT-PCR , we discovered that miR-490-3p was up-regulated in HCC tissues and cells compared with the adjacent non-tumor tissues and normal cells .\n\nWe also found that overexpression of miR-490-3p led to an increase in cell proliferation , migration , and invasion abilities and that it contributed to EMT .\n\nThe inhibition of miR-490-3p had the opposite effect on the cells .\n\nWe identified ERGIC3 ( endoplasmic reticulum-Golgi intermediate compartment protein 3 ) as a direct target gene for miR-490-3p .\n\nUnlike most miRNA-mRNA interactions , miR-490-3p increased ERGIC3 mRNA and protein levels as well as the intensity of expression of the EGFP reporter gene controlled by the 3'-UTR of ERGIC3 mRNA .\n\nThe up-regulation by miR-490-3p also required the participation of Ago2 .\n\nThe inhibition of miR-490-3p reduced the expression of ERGIC3 .\n\nOverexpression of ERGIC3 led to the same effect on HCC cells as miR-490-3p overexpression , including EMT .\n\nImportantly , silencing ERGIC3 reversed the cellular responses mediated by miR-490-3p overexpression .\n\nIn conclusion , our study indicated for the first time that miR-490-3p functioned like an oncogenic miRNA in HCC cells and that the inhibition of miR-490-3p might provide an potential treatment approach for HCC patients .", "output": "Activating invasion and metastasis" }, { "input": "Group VIB Phospholipase A(2) ( iPLA(2)\u03b3 ) is distributed in membranous organelles in which \u03b2-oxidation occurs , that is , mitochondria and peroxisomes , and is expressed by insulin-secreting pancreatic islet \u03b2-cells and INS-1 insulinoma cells , which can be injured by inflammatory cytokines , for example , IL-1\u03b2 and IFN-\u03b3 , and by oxidants , for example , streptozotocin ( STZ ) or t-butyl-hydroperoxide ( TBHP ) , via processes pertinent to mechanisms of \u03b2-cell loss in types 1 and 2 diabetes mellitus .\n\nWe find that incubating INS-1 cells with IL-1\u03b2 and IFN-\u03b3 , with STZ , or with TBHP causes increased expression of iPLA(2)\u03b3 mRNA and protein .\n\nWe prepared INS-1 knockdown ( KD ) cell lines with reduced iPLA(2)\u03b3 expression , and they proliferate more slowly than control INS-1 cells and undergo increased membrane peroxidation in response to cytokines or oxidants .\n\nAccumulation of oxidized phospholipid molecular species in STZ-treated INS-1 cells was demonstrated by LC/MS/MS scanning , and the levels in iPLA(2)\u03b3-KD cells exceeded those in control cells. iPLA(2)\u03b3-KD INS-1 cells also exhibited higher levels of apoptosis than control cells when incubated with STZ or with IL-1\u03b2 and IFN-\u03b3 .\n\nThese findings suggest that iPLA(2)\u03b3 promotes \u03b2-cell proliferation and that its expression is increased during inflammation or oxidative stress as a mechanism to mitigate membrane injury that may enhance \u03b2-cell survival .", "output": "Tumor promoting inflammation" }, { "input": "Reprogramming of cellular metabolism is a key eventduring tumorigenesis .\n\nDespite being known for decades ( Warburg effect ) , the molecular mechanisms regulating this switch remained unexplored .\n\nHere , we identify SIRT6 as a tumor suppressor thatregulates aerobic glycolysis in cancer cells .\n\nImportantly , loss of SIRT6 leads to tumor formation without activation of known oncogenes , whereas transformed SIRT6-deficient cells display increased glycolysis and tumor growth , suggesting that SIRT6 plays a role in both establishment and maintenance of cancer .\n\nBy using a conditional SIRT6 allele , we show that SIRT6 deletion invivo increases the number , size , and aggressiveness of tumors .\n\nSIRT6 also functions as a regulator of ribosome metabolismby corepressing MYC transcriptional activity .\n\nLastly,Sirt6 is selectively downregulated in several human cancers , and expression levels of SIRT6 predict prognosis and tumor-free survival rates , highlighting SIRT6 as a critical modulator of cancermetabolism .\n\nOur studies reveal SIRT6 to be a potent tumor suppressor acting to suppress cancer metabolism .", "output": "Genomic instability and mutation, Cellular energetics" }, { "input": "The mechanisms by which hematopoietic stem and progenitor cells ( HSC and HPC ) from myelodysplastic syndromes ( MDS ) undergo ineffective production of blood cells and disease transformation into acute myeloid leukemia remain to be investigated .\n\nIt has been confirmed that increased production of reactive oxygen species ( ROS ) under various pathological conditions impairs HSC self-renewal and causes HSC premature exhaustion and BM suppression primarily via induction of HSC senescence , and oncogene induces accumulation of ROS and DNA damage and subsequently cellular senescence , which functions as an important barrier to prevent the growth of transformed cells to form a neoplasia .\n\nHere we investigated whether MDS CD34(+) cells enriched with HSC and HPC undergo senescence through accumulation of ROS and DNA damage and their action mechanisms .\n\nIn this study , the percentages of SA-\u03b2-gal positive senescent CD34(+) cells increased in lower-risk MDS patients , but not in higher-risk MDS and AML patients , compared to that of healthy controls .\n\nThe increases were associated with an elevated expression of p21 but not the activation of p38 .\n\nFurther study found that there were increased ROS and DNA damage in CD34(+)CD38(-) cells enriched with HSC progression from lower-risk MDS , higher-risk MDS to AML .\n\nTherefore , these data suggest that CD34(+) cells from patients with lower-risk MDS present p21 dependent premature senescence , increased accumulation of ROS and DNA damage in CD34(+)CD38(-) cells could contribute to this process ; however , CD34(+) cells from patients with higher-risk MDS could develop some mechanisms to uncouple ROS and DNA damage induced senescence .", "output": "Genomic instability and mutation, Tumor promoting inflammation, Enabling replicative immortality, Avoiding immune destruction" }, { "input": "The functions of Rap-1A in oral carcinogenesis are largely unexplored .\n\nIn this study , we examined the expression of Rap-1A at different malignant stages of oral cavity squamous cell carcinoma ( OCSCC ) .\n\nSemiquantitative RT-PCR , quantitative RT-PCR , and Western blotting were used to evaluate Rap-1A mRNA and protein expressions , respectively , in paired OCSCC patient specimens .\n\nTo determine the possible correlation between Rap-1A expression and various clinical characteristics , 256 samples from patients with OCSCC were evaluated by immunohistochemical staining .\n\nStrong Rap-1A expression was a significant prognostic marker and predictor of aggressive OCSCC .\n\nThe overall and disease-specific 5-year survival rates were significantly correlated with strong expression of Rap-1A ( P < 0.001 ) .\n\nFunctionally , overexpressed Rap-1A could promote oral cancer cell migration and invasion by Transwell chambers and wound healing assay .\n\nConversely , the suppression of Rap-1A expression using Rap-1A-mediated siRNA was sufficient to decrease cell motility .\n\nFurthermore , our data also illustrated that Aurora-A could not only induce mRNA and protein expressions of Rap-1A for enhancing cancer cell motility but also co-localize and form a complex with Rap-1A in the oral cancer cell line .\n\nFinally , immunohistochemical staining , indirect immunofluorescence , and Western blotting analysis of human aggressive OCSCC specimens revealed a significantly positive correlation between Rap-1A and Aurora-A expression .\n\nTaken together , our results suggest that the Aurora-A/Rap-1A pathway is associated with survival , tumor progression , and metastasis of OCSCC patients .", "output": "Activating invasion and metastasis" }, { "input": "The flow of interstitial fluid and the associated interstitial fluid pressure ( IFP ) in solid tumors and surrounding host tissues have been identified as critical elements in cancer growth and vascularization .\n\nBoth experimental and theoretical studies have shown that tumors may present elevated IFP , which can be a formidable physical barrier for delivery of cell nutrients and small molecules into the tumor .\n\nElevated IFP may also exacerbate gradients of biochemical signals such as angiogenic factors released by tumors into the surrounding tissues .\n\nThese studies have helped to understand both biochemical signaling and treatment prognosis .\n\nBuilding upon previous work , here we develop a vascular tumor growth model by coupling a continuous growth model with a discrete angiogenesis model .\n\nWe include fluid/oxygen extravasation as well as a continuous lymphatic field , and study the micro-environmental fluid dynamics and their effect on tumor growth by accounting for blood flow , transcapillary fluid flux , interstitial fluid flow , and lymphatic drainage .\n\nWe thus elucidate further the non-trivial relationship between the key elements contributing to the effects of interstitial pressure in solid tumors .\n\nIn particular , we study the effect of IFP on oxygen extravasation and show that small blood/lymphatic vessel resistance and collapse may contribute to lower transcapillary fluid/oxygen flux , thus decreasing the rate of tumor growth .\n\nWe also investigate the effect of tumor vascular pathologies , including elevated vascular and interstitial hydraulic conductivities inside the tumor as well as diminished osmotic pressure differences , on the fluid flow across the tumor capillary bed , the lymphatic drainage , and the IFP .\n\nOur results reveal that elevated interstitial hydraulic conductivity together with poor lymphatic function is the root cause of the development of plateau profiles of the IFP in the tumor , which have been observed in experiments , and contributes to a more uniform distribution of oxygen , solid tumor pressure and a broad-based collapse of the tumor lymphatics .\n\nWe also find that the rate that IFF is fluxed into the lymphatics and host tissue is largely controlled by an elevated vascular hydraulic conductivity in the tumor .\n\nWe discuss the implications of these results on microenvironmental transport barriers , and the tumor invasive and metastatic potential .\n\nOur results suggest the possibility of developing strategies of targeting tumor cells based on the cues in the interstitial fluid .", "output": "Activating invasion and metastasis" }, { "input": "Methyleugenol ( MEG ) , a constituent of plants used in the human diet , is hepatocarcinogenic in rodents .\n\nIn an experiment to elucidate its mode of action in rat liver , male F344 rats were administered MEG intragastrically at 3 doses per week for up to 16 weeks in an initiation phase , after which half the rats were fed 500 ppm phenobarbital ( PB ) in the diet to promote liver neoplasia and the other half were maintained on control diet for 24 weeks .\n\nAt 8 and 16 week interim terminations , ( 32)P-nucleotide postlabeling assay revealed 3 adducts in livers of all MEG groups .\n\nThe hepatocellular replicating fractions , measured by proliferating cell nuclear antigen immunohistochemistry , were doubled or more in all MEG groups .\n\nHepatocellular altered foci , detected by glutathione S-transferase-placental type ( \\u03c0 ) immunohistochemistry , were present beginning with the high dose group at 8 weeks and extending to all MEG groups at 16 weeks .\n\nAt the end of maintenance/promotion phase , the incidences , multiplicity and size of foci was similar between control and low dose groups , while those of mid and high dose groups were increased .\n\nHepatocellular adenomas occurred in the mid and high dose groups , attaining higher multiplicity and size with PB .\n\nThus , MEG had rapid initiating activity , reflecting the formation of DNA adducts and possibly cell proliferation .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "The potential of cytologically reconstructed barley line D-2946 to cope with the major lesions that hamper genome integrity , namely DNA single- and double-strand breaks was investigated .\n\nStrand breaks induced by \\u03b3-rays and Li ions were assessed by neutral and alkaline comet assay .\n\nRepair capacity after bleomycin treatment was evaluated by agarose gel electrophoresis under neutral and alkaline conditions .\n\nFrequencies of radiation-induced chromosome aberrations were also determined .\n\nResults indicate that radiation-mediated constitutive rearrangement of the chromosome complement has led to a substantial modulation of the sensitivity of barley genome towards DNA strand breaks , produced by ionising radiation , Li ion implantation and bleomycin in an agent-specific manner , as well as of the clastogenic response to \\u03b3-rays .\n\nBased on these findings , reconstructed barley karyotype D-2946 can be considered a candidate radio-sensitive line with reduced ability to maintain genome integrity with respect to both DNA and chromosomal damage .", "output": "Genomic instability and mutation" }, { "input": "Tumor-infiltrating immune cells can promote chemoresistance and metastatic spread in aggressive tumors .\n\nConsequently , the type and quality of immune responses present in the neoplastic stroma are highly predictive of patient outcome in several cancer types .\n\nIn addition to host immune responses , intrinsic tumor cell activities that mimic stem cell properties have been linked to chemoresistance , metastatic dissemination and the induction of immune suppression .\n\nCancer stem cells are far from a static cell population ; rather , their presence appears to be controlled by highly dynamic processes that are dependent on cues from the tumor stroma .\n\nHowever , the impact immune responses have on tumor stem cell differentiation or expansion is not well understood .\n\nIn this study , we demonstrate that targeting tumor-infiltrating macrophages and inflammatory monocytes by inhibiting either the myeloid cell receptors CSF1R or CCR2 decreases the number of tumor-initiating cells in pancreatic tumors .\n\nTargeting CCR2 or CSF1R improves chemotherapeutic efficacy , inhibits metastasis and increases antitumor T-cell responses .\n\nTumor-educated macrophages also directly enhanced the tumor-initiating capacity of pancreatic tumor cells by activating the transcription factor STAT3 , thereby facilitating macrophage-mediated suppression of CD8+ T lymphocytes .\n\nTogether , our findings show how targeting tumor-infiltrating macrophages can effectively overcome therapeutic resistance mediated by tumor-initiating cells .", "output": "Tumor promoting inflammation, Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "The glutathione peroxidases , a family of selenocysteine-containing redox enzymes , play pivotal roles in balancing the signaling , immunomodulatory , and deleterious effects of reactive oxygen species ( ROS ) .\n\nThe glutathione peroxidase GPX3 is the only extracellular member of this family , suggesting it may defend cells against ROS in the extracellular environment .\n\nNotably , GPX3 hypermethylation and underexpression occur commonly in prostate , gastric , cervical , thyroid , and colon cancers .\n\nWe took a reverse genetics approach to investigate whether GPX3 would augment inflammatory colonic tumorigenesis , a process characterized by oxidative stress and inflammation , comparing Gpx3(-/-) mice in an established two-stage model of inflammatory colon carcinogenesis .\n\nGpx3-deficient mice exhibited an increased tumor number , though not size , along with a higher degree of dysplasia .\n\nIn addition , they exhibited increased inflammation with redistribution toward protumorigenic M2 macrophage subsets , increased proliferation , hyperactive WNT signaling , and increased DNA damage .\n\nTo determine the impact of acute gene loss in an established colon cancer line , we silenced GPX3 in human Caco2 cells , resulting in increased ROS production , DNA damage and apoptosis in response to oxidative stress , combined with decreased contact-independent growth .\n\nTaken together , our results suggested an immunomodulatory role for GPX3 that limits the development of colitis-associated carcinoma .", "output": "Tumor promoting inflammation, Genomic instability and mutation, Resisting cell death" }, { "input": "Collagen triple helix repeat containing-1 ( CTHRC1 ) is a secreted protein involved in vascular remodeling , bone formation and developmental morphogenesis .\n\nCTHRC1 has recently been shown to be expressed in human cancers such as breast cancer and melanoma .\n\nIn this study , we show that CTHRC1 is highly expressed in human pancreatic cancer tissues and plays a role in the progression and metastasis of the disease .\n\nCTHRC1 promoted primary tumor growth and metastatic spread of cancer cells to distant organs in orthotopic xenograft tumor mouse models .\n\nOverexpression of CTHRC1 in cancer cells resulted in increased motility and adhesiveness , whereas these cellular activities were diminished by down-regulation of the protein .\n\nCTHRC1 activated several key signaling molecules , including Src , focal adhesion kinase , paxillin , mitogen-activated protein kinase kinase ( MEK ) , extracellular signal-regulated kinase and Rac1 .\n\nTreatment with chemical inhibitors of Src , MEK or Rac1 and expression of dominant-negative Rac1 attenuated CTHRC1-induced cell migration and adhesion .\n\nCollectively , our results suggest that CTHRC1 has a role in pancreatic cancer progression and metastasis by regulating migration and adhesion activities of cancer cells .", "output": "Activating invasion and metastasis" }, { "input": "Low-dose cyclophosphamide ( CP ) therapy induces immunogenic tumor cell death and decreases regulatory T cell ( Treg ) numbers in mice with transplantable tumors .\n\nUsing the ret transgenic murine melanoma model that resembles human melanoma , we detected no beneficial antitumor effects with such treatment , despite a decrease in Tregs .\n\nOn the contrary , low-dose CP enhanced the production of chronic inflammatory mediators in melanoma lesions associated with increased accumulation of Gr1(+)CD11b(+) myeloid-derived suppressor cells ( MDSCs ) , which exhibit elevated suppressive activity and nitric oxide ( NO ) production as well as inhibition of T-cell proliferation .\n\nMoreover , the frequencies of CD8(+) T cells in the tumors and their ability to produce perforin were decreased .\n\nTo study whether the observed CP-induced MDSC expansion and activation also occurs under chronic inflammatory tumor-free conditions , mice exhibiting chronic inflammation were treated with CP .\n\nSimilar to tumor-bearing mice , CP-treated inflamed mice displayed elevated levels of MDSCs with enhanced production of NO , reactive oxygen species , and a suppressed in vivo natural killer ( NK ) cell cytotoxic activity indicating CP effects on the host immune system independent of the tumor .\n\nWe suggest that melanoma therapy with low-dose CP could be efficient only when combined with the neutralization of MDSC immunosuppressive function and chronic inflammatory microenvironment.Journal of Investigative Dermatology advance online publication , 6 December 2012 ; doi:10.1038/jid.2012.444 .", "output": "Tumor promoting inflammation" }, { "input": "BACKGROUND Young age is a favorable prognostic factor for patients with glioblastoma multiforme ( GBM ) .\n\nWe reviewed the outcomes and molecular tumor characteristics of adolescent and young adult patients with GBM treated in 2 Austrian centers .\n\nPATIENTS AND METHODS Data on patients with histologically proven primary GBM diagnosed from 18 through 40 years of age were retrospectively analyzed .\n\nAll patients were treated with standard first-line therapy .\n\nThe primary end points were overall survival ( OS ) and time to progression ( TTP ) .\n\nIDH1-R132H mutation status was analyzed using immunohistochemistry , and MGMT promoter methylation was assessed using methylation-specific polymerase chain reaction .\n\nRESULTS We included 70 patients ( 36 men and 34 women ) with a median age of 33 years .\n\nIDH1-R132H mutations were detected in 22 ( 39.3% ) of 56 cases and MGMT promoter methylation in 33 ( 61.1% ) of 54 cases with available tissue samples .\n\nIn patients with wild-type IDH , median TTP was 8.2 months and median OS was 24 months , compared with 18 months and 44 months , respectively , observed in patients with mutated IDH .\n\nNeither IDH1 nor MGMT status showed a statistically significant association with TTP or OS .\n\nOf note , the social and economical situation of the young patients with GBM was alarming , because only 17% succeeded in staying employed after receiving the diagnosis .\n\nCONCLUSIONS We found a high frequency of IDH1 mutations and MGMT promoter methylation among young adult patients with primary GBM that may contribute to the generally favorable outcome associated with young age .\n\nThe social and economic coverage of patients with glioma remains an unsolved socio-ethical problem .", "output": "Genomic instability and mutation" }, { "input": "Kinins and their receptors have been recently implicated in cancer .\n\nUsing functional and molecular approaches , we investigated the relevance of kinin B(1) and B(2) receptors in bladder cancer .\n\nFunctional studies were conducted using bladder cancer cell lines , and human biopsies were employed for molecular studies .\n\nBoth B(1) des-Arg(9)-BK and B(2) BK receptor agonists stimulated the proliferation of grade 3-derived T24 bladder cancer cells .\n\nFurthermore , treatment with B(1) and B(2) receptor antagonists ( SSR240612 and HOE140 ) markedly inhibited the proliferation of T24 cells .\n\nOnly higher concentrations of BK increased the proliferation of the grade 1 bladder cancer cell line RT4 , while des-Arg(9)-BK completely failed to induce its proliferation .\n\nReal-time PCR revealed that the mRNA expression of kinin receptors , particularly B(1) receptors , was increased in T24 cells relative to RT4 cells .\n\nData from bladder cancer human biopsies revealed that B(1) receptor expression was increased in all tumor samples and under conditions of chronic inflammation .\n\nWe also show novel evidence demonstrating that the pharmacological inhibition of PI3K\u03b3 ( phosphatidylinositol 3-kinase ) with AS252424 , concentration-dependently reduced T24 cell proliferation induced by BK or des-Arg(9)-BK .\n\nFinally , the incubation of T24 cells with kinin agonists led to a marked activation of the PI3K/AKT and ERK 1/2 signaling pathways , whereas p38 MAP kinase remained unaffected .\n\nKinin receptors , especially B(1) receptors , appear to be implicated in bladder cancer progression .\n\nIt is tempting to suggest that selective kinin antagonists might represent potential alternative therapies for bladder cancer .", "output": "Tumor promoting inflammation, Sustaining proliferative signaling" }, { "input": "Osteosarcoma is a high-grade malignant bone tumor that manifests ingravescent clinical behavior .\n\nThe intrinsic events that confer malignant properties on osteosarcoma cells have remained unclear , however .\n\nWe previously established two lines of mouse osteosarcoma cells : AX cells , which are able to form tumors in syngeneic mice , and AXT cells , which were derived from such tumors and acquired an increased tumorigenic capacity during tumor development .\n\nWe have now identified Igf2 mRNA-binding protein3 ( Imp3 ) as a key molecule responsible for this increased tumorigenicity of AXT cells in vivo .\n\nImp3 is consistently up-regulated in tumors formed by AX cells , and its expression in these cells was found to confer malignant properties such as anchorage-independent growth , loss of contact inhibition , and escape from anoikis in vitro .\n\nThe expression level of Imp3 also appeared directly related to tumorigenic ability in vivo which is the critical determination for tumor-initiating cells .\n\nThe effect of Imp3 on tumorigenicity of osteosarcoma cells did not appear to be mediated through Igf2-dependent mechanism .\n\nOur results implicate Imp3 as a key regulator of stem-like tumorigenic characteristics in osteosarcoma cells and as a potential therapeutic target for this malignancy .", "output": "Evading growth suppressors" }, { "input": "Oxaliplatin is included in a number of effective combination regimens used as first and subsequent lines of therapy for metastatic colorectal cancer .\n\nAccumulating evidence indicates that autophagy plays a significant role in response to cancer therapy .\n\nHowever , the role of autophagy in oxaliplatin-induced cell death remains to be clarified .\n\nIn this study , we showed that oxaliplatin induced cell death and autophagy in Caco-2 colorectal cancer cells .\n\nThe suppression of autophagy using either pharmacologic inhibitors ( 3-methyladenine , bafilomycin A1 ) or RNA interference in essential autophagy genes ( ATG5 or Beclin1 ) enhanced the cell death and reactive oxygen species ( ROS ) production induced by oxaliplatin in Caco-2 cells .\n\nBlocking oxaliplatin-induced ROS production by using ROS scavengers ( NAC or Tiron ) decreased autophagy .\n\nFurthermore , numerous dilated endoplasmic reticula ( ER ) were present in oxaliplatin-treated Caco-2 cells , and blocking ER stress by RNA interference against candidate of metastasis-1 ( P8 ) and C/EBP-homologous protein ( CHOP ) decreased autophagy and ROS production .\n\nTaken together , these data indicate that oxaliplatin activates autophagy as a cytoprotective response via ER stress and ROS in human colorectal cancer cells .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Lymphocytes are a key component of the immune system and their differentiation and function are directly influenced by cancer .\n\nWe examined peripheral blood lymphocyte ( PBL ) gene expression as a biomarker of illness and treatment effect using the Affymetrix Human Gene ST1 platform in patients with metastatic renal cell carcinoma ( mRCC ) who received combined treatment with IL-2 , interferon-?-2a and dendritic cell vaccine .\n\nWe examined gene expression , cytokine levels in patient serum and lymphocyte subsets as determined by flow cytometry ( FCM ) .\n\nPre-treatment PBLs from patients with mRCC exhibit a gene expression profile and serum cytokine profile consistent with inflammation and proliferation not found in healthy donors ( HD ) .\n\nPBL gene expression from patients with mRCC showed increased mRNA of genes involved with T-cell and T(REG)-cell activation pathways , which was also reflected in lymphocyte subset distribution .\n\nOverall , PBL gene expression post-treatment ( POST ) was not significantly different than pre-treatment ( PRE ) .\n\nNevertheless , treatment related changes in gene expression ( post-treatment minus pre-treatment ) revealed an increased expression of T-cell and B-cell receptor signaling pathways in responding ( R ) patients compared to non-responding ( NR ) patients .\n\nIn addition , we observed down-regulation of T(REG)-cell pathways post-treatment in R vs. NR patients .\n\nWhile exploratory in nature , this study supports the hypothesis that enhanced inflammatory cytotoxic pathways coupled with blunting of the regulatory pathways is necessary for effective anti-cancer activity associated with immune therapy .\n\nThis type of analysis can potentially identify additional immune therapeutic targets in patients with mRCC .", "output": "Tumor promoting inflammation" }, { "input": "Metastasis , commonly to the lung , is the major cause of mortality from testicular cancer .\n\nThe aim of the present study was to examine the effect of a novel nutrient mixture ( NM ) containing ascorbic acid , amino acids and green tea extract on the inhibition of melanoma growth and metastasis using a model of intratesticular inoculation of B16FO cells into nude mice .\n\nMale athymic mice ( n=12 ) , 10-12 weeks of age , were inoculated with 5\ufffd10(5) B16FO melanoma cells in 100 \u03bcl of PBS into the right testis , while the left testis was left untreated .\n\nFollowing inoculation , the mice were randomly divided into two groups .\n\nThe control group ( n=6 ) was fed a regular mouse chow diet and the NM 1% group ( n=6 ) the same diet , but supplemented with 1% NM .\n\nFour weeks later the mice were sacrificed and the abdominal cavity was opened .\n\nMice in the control group exhibited extensive metastasis in the peritoneal cavity and severely enlarged right testes and necrotic seminiferous tubules .\n\nBy contrast , in the NM 1% fed group there was no evidence of peritoneal metastasis in 50% of the animals and mild metastasis in the remaining 50% .\n\nThe right testes were enlarged and seminiferous tubules in the area of invasion showed evidence of degeneration .\n\nNo metastasis to the liver , kidney or spleen were evident in either group .\n\nHowever , severe lung metastasis was observed in 2 of 6 mice in the control group and mild metastasis in 2 of 6 mice in the NM 1% group .\n\nIn conclusion , these results confirm earlier studies and verify the anti-metastatic potential of NM .", "output": "Activating invasion and metastasis" }, { "input": "The prediction of tumor behavior for patients with oral carcinomas remains a challenge for clinicians .\n\nThe presence of lymph node metastasis is the most important prognostic factor but it is limited in predicting local relapse or survival .\n\nThis highlights the need for identifying biomarkers that may effectively contribute to prediction of recurrence and tumor spread .\n\nIn this study , we used one- and two-dimensional gel electrophoresis , mass spectrometry and immunodetection methods to analyze protein expression in oral squamous cell carcinomas .\n\nUsing a refinement for classifying oral carcinomas in regard to prognosis , we analyzed small but lymph node metastasis-positive versus large , lymph node metastasis-negative tumors in order to contribute to the molecular characterization of subgroups with risk of dissemination .\n\nSpecific protein patterns favoring metastasis were observed in the \" more-aggressive \" group defined by the present study .\n\nThis group displayed upregulation of proteins involved in migration , adhesion , angiogenesis , cell cycle regulation , anti-apoptosis and epithelial to mesenchymal transition , whereas the \" less-aggressive \" group was engaged in keratinocyte differentiation , epidermis development , inflammation and immune response .\n\nBesides the identification of several proteins not yet described as deregulated in oral carcinomas , the present study demonstrated for the first time the role of cofilin-1 in modulating cell invasion in oral carcinomas .", "output": "Activating invasion and metastasis, Sustaining proliferative signaling, Tumor promoting inflammation, Inducing angiogenesis, Resisting cell death" }, { "input": "The endothelial cell-specific microRNA ( miRNA ) , miR-126 , is considered a master regulator of physiological angiogenesis .\n\nTransplanted mesenchymal stem cells ( MSCs ) release soluble factors contributing to neoangiogenesis and cardiac repair .\n\nTherefore , we hypothesized that the over-expression of miR-126 may prolong MSC survival and enhance the cell secretome , thereby improving post-infarction angiogenesis and cardiac function .\n\nIn this study , MSCs harvested from male C57BL/6 mouse bone marrow were infected in vitro with miR-126 ( MSC(miR-126) ) by using recombinant lentiviral vectors ; the control cells were either non-transfected or transduced with mock vectors ( MSC(null) ) .\n\nThe results showed an increased secretion of angiogenic factors and a higher resistance against hypoxia in MSC(miR-126) compared with the control cells .\n\nThe expression of the Notch ligand Delta-like ( Dll)-4 in the MSC(miR-126) group was also increased .\n\nFor in vivo experiments , MSC(miR-126) cultures were intramyocardially injected into the infarct region of the hearts of female C57BL/6 mice ( an acute myocardial infarction model ) who had undergone ligation of the left anterior descending coronary artery .\n\nThe survival of MSC(miR-126) cultures , determined by Sry expression , was increased at 7 days after transplantation .\n\nMSC(miR-126)-treated animals showed significantly improved cardiac function as assessed by echocardiography 2 weeks later .\n\nThe expression levels of angiogenic factors and Dll-4 in the infarcted myocardium were further increased by MSC(miR-126) compared with MSCs or MSC(null) cultures .\n\nFurthermore , fluorescent microsphere and histological studies revealed that myocardial blood flow and microvessel density were significantly increased in the MSC(miR-126)-transplanted animals .\n\nIn addition , we found increased immature vessel proliferation following the transplantation of MSC(miR-126) cultures in which the expression of Dll-4 had been knocked down .\n\nHowever , blood vessels with lumen were barely detected , which indicated that Dll-4 plays a key role in tubulogenesis .\n\nWe conclude that the transplantation of MSCs overexpressing miR-126 can further enhance functional angiogenesis in the ischemic myocardium possibly by the secretion of angiogenic factors and the activation of Dll-4 , thus increasing MSC survival .\n\nTherefore , MSCs modified with miR-126 may represent a novel and efficient therapeutic approach for ischemic angiogenesis and the improvement of cardiac function .", "output": "Inducing angiogenesis" }, { "input": "Topoisomerase 1 ( Top1)-DNA cleavage complexes induced by camptothecin ( CPT ) cause DNA strand breaks during DNA replication or transcription .\n\nAlthough the cellular responses to replication-mediated DNA double-strand breaks have been well studied , the responses to transcription-mediated DNA strand breaks have not .\n\nHere , we show that poly ( ADP-ribose ) polymerase ( PARP ) and cockayne syndrome group B protein ( CSB ) modulate the CPT-induced formation of discrete p53-binding protein 1 ( 53BP1 ) nuclear foci at sites of transcription-mediated DNA strand breaks .\n\nInhibition of PARP activity enhanced the formation of these foci , while knockdown of essential components of the base excision repair ( BER ) pathway did not .\n\nThese findings suggest that PARP suppresses transcription-mediated 53BP1 foci formation , but that this does not occur through the BER pathway .\n\nIn addition , knockdown of CSB , one of the key factors of transcription-coupled repair , slowed the kinetics of 53BP1 foci formation .\n\nThese data suggest that PARP and CSB modulate the formation of 53BP1 foci during the processing of transcription-mediated DNA strand breaks .", "output": "Genomic instability and mutation" }, { "input": "Autophagy is an evolutionarily conserved , multi-step lysosomal degradation process in which a cell degrades its own long-lived proteins and damaged organelles .\n\nAtaxia telangiectasia mutated ( ATM ) has recently been shown to upregulate the process of autophagy .\n\nPrevious studies showed that certain microRNAs , including miR-18a , potentially regulate ATM in cancer cells .\n\nHowever , the mechanisms behind the modulation of ATM by miR-18a remain to be elucidated in colon cancer cells .\n\nIn the present study , we explored the impact of miR-18a on the autophagy process and ATM expression in HCT116 colon cancer cells .\n\nTo determine whether a preliminary link exists between autophagy and miR-18a , HCT116 cells were irradiated and quantitative ( q ) PCR was performed to measure miR-18a expression .\n\nHCT116 cells were transfected with an miR-18a mimic to study its impact on indicators of autophagy .\n\nWestern blotting and luciferase assays were implemented to explore the impact of miR-18a on ATM gene expression in HCT116 cells .\n\nThe results showed that miR-18a expression was strongly stimulated by radiation .\n\nEctopic overexpression of miR-18a in HCT116 cell lines potently enhanced autophagy and ionizing radiation-induced autophagy .\n\nMoreover , miR-18a overexpression led to the upregulation of ATM expression and suppression of mTORC1 activity .\n\nResults of the present study pertaining to the role of miR-18a in regulating autophagy and ATM gene expression in colon cancer cells revealed a novel function for miR-18a in a critical cellular event and on a crucial gene with significant impacts in cancer development , progression , treatment and in other diseases .", "output": "Resisting cell death" }, { "input": "SCOPE Glyceollins are a novel class of soybean phytoalexins with potential cancer-preventive and antiestrogenic effects .\n\nThe angiogenic cascade during tumor development consists of the release of angiogenic factors and binding of angiogenic factors to receptors on endothelial cells to activate downstream signaling pathways .\n\nHowever , the potential medicinal value of glyceollins , especially in antiangiogenesis , remains unexplored .\n\nMETHODS AND RESULTS Here , we investigated the antiangiogenic activity of glyceollins and their underlying mechanisms .\n\nGlyceollins inhibited vascular endothelial growth factor ( VEGF ) or basic fibroblast growth factor ( bFGF ) induced in vitro angiogenic activity .\n\nGlyceollins inhibited VEGF receptor-2 or FGF receptor-1 activity and their downstream signaling pathways such as extracellular regulated kinase 1/2 , c-Jun N-terminal kinase , as well as p38 mitogen-activated protein kinase and focal adhesion kinase induced by VEGF or bFGF .\n\nGlyceollins significantly suppressed VEGF receptor-2 kinase activity assayed by the ELISA .\n\nGlyceollins significantly attenuated in vivo and ex vivo microvessel development in a dose-dependent manner and tumor growth by suppressing microvessel density in Lewis lung carcinoma ( LLC ) mouse xenograft .\n\nCONCLUSION Thus , glyceollins , elicited ingredients of soy source , target the signaling pathways mediated by VEGF or bFGF , providing new perspectives into potential therapeutics for preventing and treating hypervascularized diseases including cancer .", "output": "Inducing angiogenesis" }, { "input": "The tumor suppressors Lats1 and Lats2 are mediators of the Hippo pathway that regulates tissue growth and proliferation .\n\nTheir N-terminal non-kinase regions are distinct except for Lats conserved domains 1 and 2 ( LCD1 and LCD2 ) , which may be important for Lats1/2-specific functions .\n\nLats1 knockout mice were generated by disrupting the N-terminal region containing LCD1 ( Lats1(\\u0394N/\\u0394N) ) .\n\nSome Lats1(\\u0394N/\\u0394N) mice were born safely and grew normally .\n\nHowever , mouse embryonic fibroblasts ( MEFs ) from Lats1(\\u0394N/\\u0394N) mice displayed mitotic defects , centrosomal overduplication , chromosomal misalignment , multipolar spindle formation , chromosomal bridging and cytokinesis failure .\n\nThey also showed anchorage-independent growth and continued cell cycles and cell growth , bypassing cell-cell contact inhibition similar to tumor cells .\n\nLats1(\\u0394N/\\u0394N) MEFs produced tumors in nude mice after subcutaneous injection , although the tumor growth rate was much slower than that of ordinary cancer cells .\n\nYap , a key transcriptional coactivator of the Hippo pathway , was overexpressed and stably retained in Lats1(\\u0394N/\\u0394N) MEFs in a cell density independent manner , and Lats2 mRNA expression was downregulated .\n\nIn conclusion , N-terminally truncated Lats1 induced Lats2 downregulation and Yap protein accumulation , leading to chromosomal instability and tumorigenesis .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Bigelovin is a sesquiterpene lactone isolated from the plant Inula helianthus-aquatica which was traditionally used in cancer treatment in Yunnan , China .\n\nThe potent apoptotic activities of bigelovin in human leukemia U937 cells were shown in our previous study .\n\nThe present study investigated the anti-angiogenic and immunomodulatory effects of bigelovin using transgenic zebrafish Tg(fli1a:EGFP)y1 with fluorescent blood vessels and human peripheral blood mononuclear cells ( PBMCs ) , respectively .\n\nFurthermore , the inhibitory activities of bigelovin on the human endothelial cell adhesion molecules ( CAMs ) were also examined .\n\nOur results showed that the growth of subintestinal vessels of the bigelovin-treated zebrafish embryos was significantly inhibited and the gene expressions in angiogenesis signaling pathways ( e.g .\n\nAng2 and Tie2 ) of the zebrafish were down-regulated after bigelovin treatment .\n\nBesides , the proliferation and Th1 cytokines productions ( e.g .\n\nIFN-\u03b3 , IL-2 and IL-12 ) were suppressed in bigelovin-treated PBMCs .\n\nOn the other hand , bigelovin was shown to significantly inhibit the human monocyte adhesion to human endothelial cells and the gene expressions of inflammation-related CAMs ( e.g .\n\nICAM-1 , VCAM-1 and E-selectin ) were significantly down-regulated in bigelovin-treated human endothelial cells .\n\nIn summary , our data provide the first evidence that bigelovin possesses anti-angiogenic and immunomodulatory activities , suggesting bigelovin may exert multi-target functions against cancer in animal models .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "Adipose tissue growth and development are thought to be associated with angiogenesis and extracellular matrix remodeling .\n\nBecause ginseng has been shown to inhibit angiogenesis and matrix metalloproteinase ( MMP ) activity , we hypothesized that adipose tissue growth and obesity can be regulated by Korean ginseng ( Panax ginseng C.A. Meyer ) .\n\nWild-type C57BL/6J mice were fed for 8 weeks with a low fat diet , a high fat diet ( HFD ) , or HFD supplemented with 0.5% or 5% Korean red ginseng extract .\n\nWe measured body weight , adipose tissue mass , food intake , MMP activity , and the expression of genes involved in angiogenesis and MMPs .\n\nAdministering ginseng to HFD-induced obese mice produced reductions in body weight and adipose tissue mass compared with untreated counterparts .\n\nGinseng treatment decreased blood vessel density and MMP activity in adipose tissues .\n\nGinseng also reduced mRNA levels of angiogenic factors ( e.g. , VEGF-A and FGF-2 ) and MMPs ( e.g. , MMP-2 and MMP-9 ) , whereas it increased mRNA levels of angiogenic inhibitors ( e.g. , TSP-1 , TIMP-1 , and TIMP-2 ) in adipose tissues .\n\nThese results demonstrate that ginseng effectively reduces adipose tissue mass and prevents obesity in diet-induced obese mice and that this process may be mediated in part through the anti-angiogenic actions of ginseng .", "output": "Inducing angiogenesis" }, { "input": "It is well known that ErbB2 , a receptor tyrosine kinase , localizes to the plasma membrane .\n\nHere we describe a novel observation that ErbB2 also localizes in mitochondria of cancer cells and patient samples .\n\nWe found that ErbB2 translocates into mitochondria through association with mtHSP70 .\n\nAdditionally , mitochondrial ErbB2 ( mtErbB2 ) negatively regulates mitochondrial respiratory functions .\n\nOxygen consumption and activities of complexes of the mitochondrial electron transport chain were decreased in mtErbB2-overexpressing cells .\n\nMitochondrial membrane potential and cellular ATP levels were also decreased .\n\nIn contrast , mtErbB2 enhanced cellular glycolysis .\n\nThe translocation of ErbB2 and its impact on mitochondrial function are kinase dependent .\n\nInterestingly , cancer cells with higher levels of mtErbB2 were more resistant to the ErbB2-targeting antibody trastuzumab .\n\nOur study provides a novel perspective on the metabolic regulatory function of ErbB2 and reveals that mtErbB2 has an important role in the regulation of cellular metabolism and cancer cell resistance to therapeutics .", "output": "Cellular energetics" }, { "input": "Vascular integrity is fundamental to the formation of mature blood vessels and depends on a functional , quiescent endothelial monolayer .\n\nHowever , how endothelial cells enter and maintain quiescence in the presence of angiogenic factors is still poorly understood .\n\nHere we identify the fibroblast growth factor ( FGF ) antagonist Sprouty2 ( Spry2 ) as a key player in mediating endothelial quiescence and barrier integrity in mouse aortic endothelial cells ( MAECs ) : Spry2 knockout MAECs show spindle-like shapes and are incapable of forming a functional , impermeable endothelial monolayer in the presence of FGF2 .\n\nWhereas dense wild type cells exhibit contact inhibition and stop to proliferate , Spry2 knockout MAECs remain responsive to FGF2 and continue to proliferate even at high cell densities .\n\nImportantly , the anti-proliferative effect of Spry2 is absent in sparsely plated cells .\n\nThis cell density-dependent Spry2 function correlates with highly increased Spry2 expression in confluent wild type MAECs .\n\nSpry2 protein expression is barely detectable in single cells but steadily increases in cells growing to high cell densities , with hypoxia being one contributing factor .\n\nAt confluence , Spry2 expression correlates with intact cell-cell contacts , whereas disruption of cell-cell contacts by EGTA , TNF\u03b1 and thrombin decreases Spry2 protein expression .\n\nIn confluent cells , high Spry2 levels correlate with decreased extracellular signal-regulated kinase 1/2 ( Erk1/2 ) phosphorylation .\n\nIn contrast , dense Spry2 knockout MAECs exhibit enhanced signaling by Erk1/2 .\n\nMoreover , inhibiting Erk1/2 activity in Spry2 knockout cells restores wild type cobblestone monolayer morphology .\n\nThis study thus reveals a novel Spry2 function , which mediates endothelial contact inhibition and barrier integrity .", "output": "Evading growth suppressors" }, { "input": "Pancreatic ductal adenocarcinoma ( PDAC ) is one of the most aggressive human neoplasms with extremely poor prognosis and a low survival rate .\n\nImmunosuppressive cell populations , e.g. regulatory T cells ( Treg ) , appear to be important in PDAC , contributing to patient's poor prognosis .\n\nTherefore , we investigated the PDAC microenvironment with a focus on conventional and regulatory T cells in view of their potential therapeutic importance .\n\nWe found that tumors from the murine Panc02 orthotopic model of PDAC were infiltrated with high numbers of Treg .\n\nRemarkably , these cells exhibited the effector/memory phenotype , suggesting their enhanced suppressive activity and higher proliferation capacity .\n\nAlthough we observed a steady increase in transforming growth factor-\u03b2 ( TGF-\u03b2 ) levels in the tumors , treatment with a specific inhibitor of TGF-\u03b2 receptor I kinase failed to abrogate Treg accumulation .\n\nA CCR4 antagonist did not affect Treg percentage in the tumor either .\n\nHowever , intense Treg cell division in the tumor microenvironment was demonstrated , suggesting local proliferation as a major mechanism of Treg accumulation in PDAC .\n\nNotably , this accumulation was reduced by low-dose gemcitabine administration , resulting in a modestly increased survival of PDAC mice .\n\nOur results provide an insight into mechanisms of immunosuppression in PDAC , suggesting an important role for proliferative expansion of effector/memory Treg .\n\nLow-dose gemcitabine therapy selectively depletes Treg , providing a basis for new modalities of PDAC therapy .", "output": "Avoiding immune destruction" }, { "input": "Liver resection is now widely accepted as a potentially curative treatment for colorectal liver metastasis .\n\nHowever , the efficacy of surgical resection for gastric cancer liver metastasis(GLM)remains unclear .\n\nBased on our 18-year experience with 64 patients who underwent curative hepatectomy for GLM , we discuss the indication and efficacy of surgical resection for GLM .\n\nFrom January 1993 to January 2011 , 73 patients underwent hepatectomy for GLM in the Department of Gastroenterological Surgery , Cancer Institute Ariake Hospital(Japanese Foundation for Cancer Research ) , Japan .\n\nThe actuarial1 - , 3- , and 5-year overall survival rates and 1- , 3- , and 5-year recurrence-free survival rates of those 64 patients who achieved curative resections were 84 , 50 , and 37% , and 42 , 27 , and 27% , respectively .\n\nBy multivariate analysis , serosal invasion of the primary gastric cancer and larger hepatic tumor(>5 cm in diameter)were found to be independent indicators of poor prognosis .\n\nBased on the multivariate analysis results , all patients were divided into three groups no poor prognostic factor(n=38) , one poor prognostic factor(n=24) , and two poor prognostic factors(n=2) .\n\nThe actuarial overall survival rates of each group were 63 , 36 , and 0% at 3 years , and 53 , 15 , and 0% at 5 years .\n\nGLM patients having hepatic tumors with the maximum diameter of <5 cm , and without serosalinvasion of the primary gastric cancer , are the best candidates for hepatectomy .", "output": "Activating invasion and metastasis" }, { "input": "BACKGROUND Treatment of head and neck cancer with radiation often results in damage to surrounding normal tissues such as salivary glands .\n\nPermanent loss of function in the salivary glands often leads patients to discontinue treatment due to incapacitating side effects .\n\nIt has previously been shown that IGF-1 suppresses radiation-induced apoptosis and enhances G2/M arrest leading to preservation of salivary gland function .\n\nIn an effort to recapitulate the effects of IGF-1 , as well as increase the likelihood of translating these findings to the clinic , the small molecule therapeutic Roscovitine , is being tested .\n\nRoscovitine is a cyclin-dependent kinase inhibitor that acts to transiently inhibit cell cycle progression and allow for DNA repair in damaged tissues .\n\nMETHODOLOGY/PRINCIPAL FINDINGS Treatment with Roscovitine prior to irradiation induced a significant increase in the percentage of cells in the G(2)/M phase , as demonstrated by flow cytometry .\n\nIn contrast , mice treated with radiation exhibit no differences in the percentage of cells in G(2)/M when compared to unirradiated controls .\n\nSimilar to previous studies utilizing IGF-1 , pretreatment with Roscovitine leads to a significant up-regulation of p21 expression and a significant decrease in the number of PCNA positive cells .\n\nRadiation treatment leads to a significant increase in activated caspase-3 positive salivary acinar cells , which is suppressed by pretreatment with Roscovitine .\n\nAdministration of Roscovitine prior to targeted head and neck irradiation preserves normal tissue function in mouse parotid salivary glands , both acutely and chronically , as measured by salivary output .\n\nCONCLUSIONS/SIGNIFICANCE These studies suggest that induction of transient G(2)/M cell cycle arrest by Roscovitine allows for suppression of apoptosis , thus preserving normal salivary function following targeted head and neck irradiation .\n\nThis could have an important clinical impact by preventing the negative side effects of radiation therapy in surrounding normal tissues .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND : The p38\u03b1 MAP kinase pathway is involved in inflammation , cell differentiation , growth , apoptosis and production of pro-inflammatory cytokines TNF-\u03b1 and IL-1\u03b2 .\n\nThe overproduction of these cytokines plays an important role in cancer .\n\nThe aim of this work was to design a peptide inhibitor on the basis of structural information of the active site of p38\u03b1 .\n\nMETHODS : A tetrapeptide , VWCS as p38\u03b1 inhibitor was designed on the basis of structural information of the ATP binding site by molecular modeling .\n\nThe inhibition study of peptide with p38\u03b1 was performed by ELISA , binding study by Surface Plasmon Resonance and anti-proliferative assays by MTT and flow cytometry .\n\nRESULTS : The percentage inhibition of designed VWCS against pure p38\u03b1 protein and serum of HNSCC patients was 70.30 and 71.5% , respectively .\n\nThe biochemical assay demonstrated the K(D) and IC(50) of the selective peptide as 7.22\ufffd10(-9)M and 20.08nM , respectively .\n\nThe VWCS as inhibitor significantly reduced viability of oral cancer KB cell line with an IC(50) value of 10\u03bcM and induced apoptosis by activating Caspase 3 and 7 .\n\nCONCLUSIONS : VWCS efficiently interacted at the ATP binding pocket of p38\u03b1 with high potency and can be used as a potent inhibitor in case of HNSCC .\n\nGENERAL SIGNIFICANCE : VWCS can act as an anticancer agent as it potentially inhibits the cell growth and induces apoptosis in oral cancer cell-line in a dose as well as time dependent manner .\n\nHence , p38\u03b1 MAP kinase inhibitor can be a potential therapeutic agent for human oral cancer .", "output": "Resisting cell death" }, { "input": "TP53 mutations compromising p53 transcriptional function occur in more than 50% of human cancers , including pancreatic adenocarcinoma , and render cancer cells more resistant to conventional therapy .\n\nIn the last few years , many efforts have been addressed to identify p53-reactivating molecules able to restore the wild-type transcriptionally competent conformation of the mutated proteins .\n\nHere , we show that two of these compounds , CP-31398 and RITA , induce cell growth inhibition , apoptosis , and autophagy by activating p53/DNA binding and p53 phosphorylation ( Ser15 ) , without affecting the total p53 amount .\n\nThese effects occur in both wild-type and mutant p53 pancreatic adenocarcinoma cell lines , whereas they are much less pronounced in normal human primary fibroblasts .\n\nFurthermore , CP-31398 and RITA regulate the axis SESN1-2/AMPK/mTOR by inducing AMPK phosphorylation on Thr172 , which has a crucial role in the autophagic response .\n\nThe protective role of autophagy in cell growth inhibition by CP-31398 and RITA is supported by the finding that the AMPK inhibitor compound C or the autophagy inhibitors chloroquine or 3-methyladenine sensitize both pancreatic adenocarcinoma cell lines to the apoptotic response induced by p53-reactivating molecules .\n\nOur results demonstrate for the first time a survival role for autophagy induced by p53-reactivating molecules , supporting the development of an anti-cancer therapy based on autophagy inhibition associated to p53 activation .", "output": "Resisting cell death" }, { "input": "Heparanase is a mammalian endoglycosidase that degrades heparan sulfate at the cell surface and in the extracellular matrix .\n\nThe expression of heparanase was detected in a wide variety of human malignant tumors and closely associated with tumor invasion , metastasis , and angiogenesis .\n\nHowever , the specific roles of heparanase and its mechanisms of regulating the malignant potential of non-small cell lung cancer ( NSCLC ) cells still remain unclear .\n\nIn the present study , the expression of heparanase was down-regulated in NSCLC cell line by antisense oligodeoxynucleotide .\n\nResults showed that down-regulation of heparanase led to significant inhibition of invasive and proliferative potentials of A549 cells in vitro and in vivo .\n\nFurther research demonstrated that down-regulation of heparanase significantly inhibited the angiogenic potential of A549 cells , which might be the mechanism responsible for the inhibition of A549 cell proliferation in BALB/c nude mice in vivo .\n\nThese findings demonstrate that heparanase plays essential roles in regulating the invasion , proliferation , and angiogenesis of A549 cells .", "output": "Inducing angiogenesis, Sustaining proliferative signaling, Activating invasion and metastasis" }, { "input": "The role of regulatory T cells ( T(regs) ) in human colon cancer ( CC ) remains controversial : high densities of tumor-infiltrating T(regs) can correlate with better or worse clinical outcomes depending on the study .\n\nIn mouse models of cancer , T(regs) have been reported to suppress inflammation and protect the host , suppress T cells and protect the tumor , or even have direct cancer-promoting attributes .\n\nThese different effects may result from the presence of different T(reg) subsets .\n\nWe report the preferential expansion of a T(reg) subset in human CC with potent T cell-suppressive , but compromised anti-inflammatory , properties ; these cells are distinguished from T(regs) present in healthy donors by their coexpression of Foxp3 and ROR\u03b3t .\n\nT(regs) with similar attributes were found to be expanded in mouse models of hereditary polyposis .\n\nIndeed , ablation of the ROR\u03b3t gene in Foxp3(+) cells in polyp-prone mice stabilized T(reg) anti-inflammatory functions , suppressed inflammation , improved polyp-specific immune surveillance , and severely attenuated polyposis .\n\nAblation of interleukin-6 ( IL-6 ) , IL-23 , IL-17 , or tumor necrosis factor-\u03b1 in polyp-prone mice reduced polyp number but not to the same extent as loss of ROR\u03b3t .\n\nSurprisingly , loss of IL-17A had a dual effect : IL-17A-deficient mice had fewer polyps but continued to have ROR\u03b3t(+) T(regs) and developed invasive cancer .\n\nThus , we conclude that ROR\u03b3t has a central role in determining the balance between protective and pathogenic T(regs) in CC and that T(reg) subtype regulates inflammation , potency of immune surveillance , and severity of disease outcome .", "output": "Tumor promoting inflammation" }, { "input": "\u039duclear factor-\u03baB ( NF-\u03baB ) and activator protein-1 ( AP-1 ) are major transcription factors that have been associated with breast cancer metastasis by inducing matrix metalloproteinase-9 ( MMP-9 ) expression .\n\nIn this study , we investigated the inhibitory effects of guggulsterone isomers ( cis or trans ) on 12-O-tetradecanoylpho-bol-13-acetate ( TPA)-induced MMP-9 expression .\n\nCis-guggulsterone inhibited TPA-induced MMP expression by blocking I\u03baB kinase ( IKK)/NF-\u03baB signaling , whereas trans-guggulsterone blocked mitogen-activated protein kinase ( MAPK)/AP-1 signaling .\n\nCis-guggulsterone was more potent than trans-guggulsterone in the inhibition of TPA-induced MMP-9 expression and invasion of MCF-7 cells .\n\nFurthermore , we found that the combination of these isomers exerted an additive effect on the inhibition of MCF-7 cell invasion .\n\nThese results suggest that guggulsterone isomers downregulate MMP-9 expression and tumor cell invasion through the isomer-specific suppression of IKK/NF-\u03baB and MAPK/AP-1 activation .\n\nIn addition , the suppression of MMP-9 expression correlated well with the inhibition of cell invasion .", "output": "Activating invasion and metastasis" }, { "input": "Calcium ( Ca(2+) ) signals are involved in important checkpoints in cell death pathways and promote both apoptosis and autophagy .\n\nHowever , the relationship between autophagy and apoptosis in response to Ca(2+) level elevation is poorly understood .\n\nHere , we provided evidence that the influx of extracellular Ca(2+) triggered by Trichokonin VI ( TK VI ) , an antimicrobial peptide , induced calpain-dependent apoptosis and autophagy in hepatocellular carcinoma ( HCC ) cells .\n\nRemarkably , TK VI preferentially induced apoptosis that was associated with calpain-mediated Bax and Atg5 cleavage , which resulted in the collapse of the mitochondrial membrane potential and cytochrome c release .\n\nInterestingly , truncated , but not full-length Atg5 , associated with Bcl-xL and promoted the intrinsic pathway .\n\nMoreover , TK VI treatment induced reactive oxygen species ( ROS ) accumulation , an effect in which Bak might play a major role .\n\nThis accumulation of ROS resulted in the subsequent disposal of damaged mitochondria within autophagosomes via Atg5-mediated and mitochondria-selective autophagy .\n\nBoth the inhibition of calpain activity and Bax deficiency activated a switch that promoted an enhancement of autophagy .\n\nThe inhibition of both apoptosis and autophagy significantly attenuated the TK VI cytotoxicity , indicating that the two processes had stimulatory effects during TK VI-meditated cell death .\n\nThese results suggested that calpain , Bak and Atg5 were molecular links between autophagy and apoptosis and revealed novel aspects of the crosstalk between these two processes .\n\nThe potential of TK VI is proposed as a promising anticancer agent for its well-characterized activity of Ca(2+) agonist and as a possible novel therapeutic strategy that acts on cancer cell mitochondria .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "MYC deregulation is common in human cancer .\n\nIG-MYC translocations that are modeled in E\u03bc-Myc mice occur in almost all cases of Burkitt lymphoma as well as in other B-cell lymphoproliferative disorders .\n\nDeregulated expression of MYC results in increased mTOR complex 1 ( mTORC1 ) signaling .\n\nAs tumors with mTORC1 activation are sensitive to mTORC1 inhibition , we used everolimus , a potent and specific mTORC1 inhibitor , to test the requirement for mTORC1 in the initiation and maintenance of E\u03bc-Myc lymphoma .\n\nEverolimus selectively cleared premalignant B cells from the bone marrow and spleen , restored a normal pattern of B-cell differentiation , and strongly protected against lymphoma development .\n\nEstablished E\u03bc-Myc lymphoma also regressed after everolimus therapy .\n\nTherapeutic response correlated with a cellular senescence phenotype and induction of p53 activity .\n\nTherefore , mTORC1-dependent evasion of senescence is critical for cellular transformation and tumor maintenance by MYC in B lymphocytes .", "output": "Enabling replicative immortality" }, { "input": "Integrin \\u03b1v\\u03b23 plays a role in insulin-like growth factor 1 ( IGF1 ) signaling ( integrin-IGF1 receptor ( IGF1R ) cross-talk ) in non-transformed cells in anchorage-dependent conditions .\n\nWe reported previously that IGF1 directly binds to \\u03b1v\\u03b23 and induces \\u03b1v\\u03b23-IGF1-IGF1R ternary complex formation in these conditions .\n\nThe integrin-binding defective IGF1 mutant ( R36E/R37E ) is defective in inducing ternary complex formation and IGF signaling , whereas it still binds to IGF1R .\n\nWe studied if IGF1 can induce signaling in anchorage-independent conditions in transformed Chinese hamster ovary cells that express \\u03b1v\\u03b23 ( \\u03b23-CHO ) cells .\n\nHere we describe that IGF1 signals were more clearly detectable in anchorage-independent conditions ( polyHEMA-coated plates ) than in anchorage-dependent conditions .\n\nThis suggests that IGF signaling is masked by signals from cell-matrix interaction in anchorage-dependent conditions .\n\nIGF signaling required \\u03b1v\\u03b23 expression , and R36E/R37E was defective in inducing signals in polyHEMA-coated plates .\n\nThese results suggest that \\u03b1v\\u03b23-IGF1 interaction , not \\u03b1v\\u03b23-extracellular matrix interaction , is essential for IGF signaling .\n\nInhibitors of IGF1R , Src , AKT , and ERK1/2 did not suppress \\u03b1v\\u03b23-IGF-IGF1R ternary complex formation , suggesting that activation of these kinases are not required for ternary complex formation .\n\nAlso , mutations of the \\u03b23 cytoplasmic tail ( Y747F and Y759F ) that block \\u03b23 tyrosine phosphorylation did not affect IGF1R phosphorylation or AKT activation .\n\nWe propose a model in which IGF1 binding to IGF1R induces recruitment of integrin \\u03b1v\\u03b23 to the IGF-IGF1R complex and then \\u03b23 and IGF1R are phosphorylated .\n\nIt is likely that \\u03b1v\\u03b23 should be together with the IGF1-IGF1R complex for triggering IGF signaling .", "output": "Genomic instability and mutation, Sustaining proliferative signaling" }, { "input": "The ARF locus is frequently inactivated in human cancer .\n\nThe oncosuppressor ARF has indeed been described as a general sensor for different situation of cellular stress .\n\nWe have previously demonstrated that ARF deficiency severely impairs inflammatory responses in vitro and in vivo , establishing a role for ARF in the regulation of innate immunity .\n\nThe aim of the present work was to get further insights into the immune functions of ARF and to evaluate its possible contribution to the polarization of macrophages toward the M1 or M2 phenotype .\n\nOur results demonstrate that resting Arf(-/-) macrophages express high levels of Ym1 and Fizz-1 , two typical markers of alternatively-activated macrophages ( M2 ) .\n\nAdditionally , Arf(-/-) peritoneal macrophages showed an impaired response to lipopolysaccharide ( a classical inducer of M1 polaryzation ) and a reduced production of pro-inflammatory cytokines/chemokines .\n\nMoreover , upon stimulation with interleukin-4 ( IL-4 ) , an inducer of the M2 phenotype , well established M2 markers such as Fizz-1 , Ym1 and arginase-1 were upregulated in Arf(-/-) as compared with wild type macrophages .\n\nAccordingly , the cytokine and chemokine profile associated with the M2 phenotype was significantly overexpressed in Arf(-/-) macrophages responding to IL-4 .\n\nIn addition , multiple pro-angiogenic factors such as VEGF and MMP-9 were also increased .\n\nIn summary , these results indicate that ARF contributes to the polarization and functional plasticity of macrophages .", "output": "Inducing angiogenesis, Tumor promoting inflammation" }, { "input": "ABSTRACT : PurposeAutophagy has attracted attentions as a novel mechanism for tumor development .\n\nIn this study Human ovarian carcinoma cell line SKOV3 and multidrug-resistant phenotype SKVCR cells were used and the roles of autophagy in radiation-induced cell death were analyzed.Methods and materialsCell viability was examined by colony formation and cell counting kit-8 ( CCK-8 ) assay , 3MA and ZVAD were used to block autophagy and apoptosis , respectively .\n\nQuantitative real-time PCR was used to detect mRNA level and Western blot was used to detect protein expression , monodansylcadaverine ( MDC ) staining and flow cytometery were used for autophagy , apoptosis and cell cycle dynamics , respectively .\n\nRESULTS : ( 1 ) The radiosensitivity exhibited differently in SKOV3 and SKVCR cells ( SKOV3 : D0=3.37 , SKVCR : D0= 4.18 ) ; compared with SKOV3 the constitutive expression of MAPLC3 in SKVCR was higher , but no change of Caspase-3 and cleaved Caspase-3 .\n\n( 2 ) The ionizing radiation ( IR)- induced apoptosis and autophagy were significant in both cells ( P<0.05 ) ; inhibition of apoptosis with ZVAD showed no impact on survival of SKOV3 and SKVCR cells after radiation , while inhibition of autophagy significantly decreased viability in SKVCR cells , for SKVO3 cells only low level of radiation ( 2 Gy and 4 Gy ) could decrease the viability(P<0.05) .\n\n( 3 ) ZVAD inhibited apoptosis and autophagy in both cells , 3MA inhibit apoptosis in SKOV3 , and promote apoptosis in SKVCR , together with inhibition of autophagy .\n\n( 4 ) G2/M arrest was induced by radiation in both cells ; the accumulation of G2/M was more significant in SKOV3 , 3MA attenuated the radiation-induced S phase delay in SKVCR .\n\nCONCLUSION : IR-induced autophagy provides a self-protective mechanism against radiotherapy in SKVCR cells , the use of autophagy inhibitor , 3MA , increases the killing effects of radiation by inhibiting autophagy and radiation- induced S phase delay , also by the increase of apoptosis , which suggests a better therapeutic strategy in drug- resistant SKVCR ovarian cancer cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Tumor cells exhibit enhanced glucose consumption and lactate production even when supplied with adequate oxygen ( a phenomenon known as the Warburg effect , or aerobic glycolysis ) .\n\nPharmacological inhibition of aerobic glycolysis represents a potential tumor-selective approach that targets the metabolic differences between normal and malignant tissues .\n\nHuman breast tumor MDA-MB-231 cells were used to develop an assay system to discover natural product-based glycolysis inhibitors .\n\nThe assay employed was based on hypersensitivity to glycolytic inhibition in tumor cells treated with the mitochondrial electron transport inhibitor rotenone .\n\nUnder such conditions , ATP supply , and hence cell viability , depends exclusively on glycolysis .\n\nThis assay system was used to evaluate 10648 plant and marine organism extracts from the U.S. National Cancer Institute's Open Repository .\n\nBioassay-guided isolation of an active Moronobea coccinea extract yielded the new bis-geranylacylphloroglucinol derivative moronone ( 1 ) .\n\nCompound 1 exhibited enhanced antiproliferative/cytotoxic activity in the presence of rotenone-imposed metabolic stress on tumor cells .\n\nSurprisingly , mechanistic studies revealed that 1 did not inhibit glycolysis , but rather functions as a protonophore that dissipates the mitochondrial proton gradient .\n\nIn the presence of rotenone , tumor cells may be hypersensitive to protonophores due to increased ATP utilization by the ATP synthase .", "output": "Cellular energetics" }, { "input": "In normal human cells , oncogene-induced senescence ( OIS ) depends on induction of DNA damage response .\n\nOxidative stress and hyperreplication of genomic DNA have been proposed as major causes of DNA damage in OIS cells .\n\nHere , we report that down-regulation of deoxyribonucleoside pools is another endogenous source of DNA damage in normal human fibroblasts ( NHFs ) undergoing HRAS(G12V)-induced senescence .\n\nNHF-HRAS(G12V) cells underexpressed thymidylate synthase ( TS ) and ribonucleotide reductase ( RR ) , two enzymes required for the entire de novo deoxyribonucleotide biosynthesis , and possessed low dNTP levels .\n\nChromatin at the promoters of the genes encoding TS and RR was enriched with retinoblastoma tumor suppressor protein and histone H3 tri-methylated at lysine 9 .\n\nImportantly , ectopic coexpression of TS and RR or addition of deoxyribonucleosides substantially suppressed DNA damage , senescence-associated phenotypes , and proliferation arrest in two types of NHF-expressing HRAS(G12V) .\n\nReciprocally , short hairpin RNA-mediated suppression of TS and RR caused DNA damage and senescence in NHFs , although less efficiently than HRAS(G12V) .\n\nHowever , overexpression of TS and RR in quiescent NHFs did not overcome proliferation arrest , suggesting that unlike quiescence , OIS requires depletion of dNTP pools and activated DNA replication .\n\nOur data identify a previously unknown role of deoxyribonucleotides in regulation of OIS .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "DNA double-strand breaks ( DSBs ) representa serious threat for hematopoietic stem cells ( HSCs ) .\n\nHow cytokines and environmental signals integrate the DNA damage response and contribute to HSC-intrinsic DNA repair processes remains unknown .\n\nThrombopoietin ( TPO ) and its receptor , Mpl , are critical factors supporting HSC self-renewal and expansion .\n\nHere , we uncover an unknown function for TPO-Mpl in the regulation of DNA damage response .\n\nWe show that DNA repair following \\u03b3-irradiation ( \\u03b3-IR ) or the action of topoisomerase-II inhibitors is defective in Mpl(-/-) and in wild-type mouse or human hematopoietic stem and progenitor cells treated in the absence of TPO .\n\nTPO stimulates DNA repair invitro and invivo by increasing DNA-PK-dependent nonhomologous end-joining efficiency .\n\nThis ensures HSC chromosomal integrity and limits their long-term injury in response to IR.This shows that niche factors can modulate theHSC DSB repair machinery and opens new avenues for administration of TPO agonists for minimizing radiotherapy-induced HSC injury and mutagenesis .", "output": "Genomic instability and mutation" }, { "input": "In this study , laboratory experiments were carried out in order to come to a better understanding of the fate of polycyclic aromatic hydrocarbons ( PAHs ) in the marine environment and especially on their bioaccumulation , biotransformation and genotoxic effects in fish .\n\nJuveniles of turbot ( Scophthalmus maximus ) were exposed to PAHs through different routes via ( 1 ) a mixture of dissolved PAHs , ( 2 ) a PAH-polluted sediment and ( 3 ) an oil fuel elutriate .\n\nFish were exposed 4 days followed by a 6-day depuration period .\n\nIn each experiment , PAH concentrations in the seawater of the tanks were analysed regularly by gas chromatography coupled with mass spectrometry .\n\nMuscle and liver samples were also analysed for parent PAH levels and PAH bioconcentration factors were calculated .\n\nBiotransformation was evaluated by measuring the levels of PAH metabolites in fish bile .\n\nGenotoxicity was assessed by the alkaline comet assay .\n\nRegardless of exposure route , the parent PAH concentrations in the liver and muscle showed a peak level 1 day after the beginning of the exposure , followed by a decrease up to the background level towards the end of the experiment , except for the exposure to dissolved PAHs for which levels were relatively low throughout the study .\n\nAs a consequence , no bioaccumulation was observed in fish tissues at the end of the experiment .\n\nIn contrast , regardless of exposure routes , a rapid production of biliary metabolites was observed throughout the whole exposure experiment .\n\nThis was especially true for 1-hydroxypyrene , the major metabolite of pyrene .\n\nAfter 6 days of recovery in clean water , a significant decrease in the total metabolite concentrations occurred in bile .\n\nFish exposed through either route displayed a significant increase in DNA strand breaks after 4 days of exposure , and significant correlations were observed between the level of biliary PAH metabolites and the level of DNA lesions in fish erythrocytes .\n\nOverall results indicate that exposure to either a mixture of dissolved PAHs , a PAH-contaminated sediment or a dispersed oil fuel elutriate leads to biotransformation and increase in DNA damage in fish .\n\nThe quantification of PAH metabolites in bile and DNA damage in erythrocytes appear to be suitable for environmental monitoring of marine pollution either in the case of accidental oil spills or sediment contamination .", "output": "Genomic instability and mutation" }, { "input": "Cancer vaccines based on human tumor-associated antigens ( TAA ) have been tested in patients with advanced or recurrent cancer , in combination with or following standard therapy .\n\nTheir immunogenicity and therapeutic efficacy has been difficult to properly evaluate in that setting characterized by multiple highly suppressive effects of the tumor and the standard therapy on the patient's immune system .\n\nIn animal models of human cancer , vaccines administered in the prophylactic setting are most immunogenic and effectively prevent cancer development and progression .\n\nWe report results of a clinical study that show that in patients without cancer but with a history of premalignant lesions ( advanced colonic adenomas , precursors to colon cancer ) , a vaccine based on the TAA MUC1 was highly immunogenic in 17 of 39 ( 43.6% ) of vaccinated individuals , eliciting high levels of anti-MUC1 immunoglobulin G ( IgG ) and long-lasting immune memory .\n\nLack of response in 22 of 39 individuals was correlated with high levels of circulating myeloid-derived suppressor cells ( MDSC ) prevaccination .\n\nVaccine-elicited MUC1-specific immune response and immune memory were not associated with significant toxicity .\n\nOur study shows that vaccines based on human TAAs are immunogenic and safe and capable of eliciting long-term memory that is important for cancer prevention .\n\nWe also show that in the premalignant setting , immunosuppressive environment ( e.g. , high levels of MDSC ) might already exist in some individuals , suggesting an even earlier premalignant stage or preselection of nonimmunosuppressed patients for prophylactic vaccination .\n\nCancer Prev Res ; 6(1) ; 18-26. \ufffd2012 AACR .", "output": "Avoiding immune destruction" }, { "input": "Evaluation of immune dysfunction during the tumor-bearing state is a critical issue in combating cancer .\n\nIn this study , we initially found that IL-6 , one of the cachectic factors , suppressed CD4(+) T cell-mediated immunity through downregulation of MHC class II by enhanced arginase activity of dendritic cells ( DC ) in tumor-bearing mice .\n\nWe demonstrated that administration of Ab against IL-6R ( anti-IL-6R mAb ) greatly enhanced T cell responses and inhibited the growth of tumor in vivo .\n\nWe also found that IL-6 upregulated the expression of arginase-1 and arginase activity of DC in vitro .\n\nTumor-infiltrating CD11c(+) DC exhibited upregulated mRNA expression of arginase-1 but reduced expression of MHC class II in parallel with the increase in serum IL-6 levels at the late stage in tumor-bearing hosts .\n\nHowever , the administration of anti-IL-6R mAb into tumor-bearing mice inhibited both the downmodulation of MHC class II and the upregulation of arginase-1 mRNA levels in DC .\n\nFurthermore , we noted that N(\u03c9)-hydroxy-L-arginine or L-arginine , an arginase-1 inhibitor , blocked the reduction in MHC class II levels on CD11c(+) DC during the tumor-bearing state .\n\nFinally , we demonstrated that the administration of N(\u03c9)-hydroxy-L-arginine at the peritumor site significantly enhanced CD4(+) T cell responses and inhibited tumor growth .\n\nThus , IL-6-mediated arginase activation and the subsequent reduction in MHC class II expression on DC appeared to be critical mechanisms for inducing dysfunction of the immune system in the tumor-bearing state .\n\nBlockade of the IL-6-arginase cascade is a promising tool to overcome the dysfunction of antitumor immunity in tumor-bearing hosts .", "output": "Avoiding immune destruction" }, { "input": "The ASPP2 ( also known as 53BP2L ) tumor suppressor is a proapoptotic member of a family of p53 binding proteins that functions in part by enhancing p53-dependent apoptosis via its C-terminal p53-binding domain .\n\nMounting evidence also suggests that ASPP2 harbors important nonapoptotic p53-independent functions .\n\nStructural studies identify a small G protein Ras-association domain in the ASPP2 N terminus .\n\nBecause Ras-induced senescence is a barrier to tumor formation in normal cells , we investigated whether ASPP2 could bind Ras and stimulate the protein kinase Raf/MEK/ERK signaling cascade .\n\nWe now show that ASPP2 binds to Ras-GTP at the plasma membrane and stimulates Ras-induced signaling and pERK1/2 levels via promoting Ras-GTP loading , B-Raf/C-Raf dimerization , and C-Raf phosphorylation .\n\nThese functions require the ASPP2 N terminus because BBP ( also known as 53BP2S ) , an alternatively spliced ASPP2 isoform lacking the N terminus , was defective in binding Ras-GTP and stimulating Raf/MEK/ERK signaling .\n\nDecreased ASPP2 levels attenuated H-RasV12-induced senescence in normal human fibroblasts and neonatal human epidermal keratinocytes .\n\nTogether , our results reveal a mechanism for ASPP2 tumor suppressor function via direct interaction with Ras-GTP to stimulate Ras-induced senescence in nontransformed human cells .", "output": "Enabling replicative immortality" }, { "input": "ABSTRACT : BACKGROUND : Chemokine receptor CXCR4 , together with its ligand CXCL12 , plays critical roles in cancer progression , including growth , metastasis and angiogenesis .\n\nEwing sarcoma is a sarcoma with poor prognosis despite current therapies , particularly for patients with advanced-stage disease .\n\nLungs and bone ( marrow ) , organs of predilection for ( primary/metastatic ) Ewing sarcoma , represent predominant CXCL12 sources .\n\nMETHODS : To gain insight into the role of the CXCR4-CXCL12 axis in Ewing sarcoma , CXCR4 , CXCL12 and hypoxia-inducible factor-1alpha protein expression was studied in therapy-naive and metastatic tumors by immunohistochemistry .\n\nCXCR4 function was assessed in vitro , by flow cytometry and proliferation/ cell viability assays , in the presence of recombinant CXCL12 and/or CXCR4-antagonist AMD3100 or under hypoxic conditions .\n\nRESULTS : Whereas CXCR4 was predominantly expressed by tumor cells , CXCL12 was observed in both tumor and stromal areas .\n\nSurvival analysis revealed an ( expression level-dependent ) negative impact of CXCR4 expression ( p < 0.04 ) .\n\nA role for the CXCR4-CXCL12 axis in Ewing sarcoma growth was suggested by our observations that i ) CXCR4 expression correlated positively with tumor volume at diagnosis ( p = 0.013 ) , ii ) CXCL12 was present within the microenvironment of virtually all cases , iii ) CXCL12 induced proliferation of CXCR4-positive Ewing sarcoma cell lines , which could be abrogated by AMD3100 .\n\nCXCR4 expression was not correlated with occurrence of metastatic disease .\n\nAlso , therapy-naive tumors demonstrated higher CXCR4 expression as compared to metastases ( p = 0.027 ) .\n\nEvaluation of in vivo hypoxia-inducible factor-1alpha expression and culture of cells under hypoxic conditions revealed no role for hypoxia in CXCR4 expression .\n\nCONCLUSIONS : Together , our results imply a crucial role for the CXCR4-CXCL12 axis in auto- and/or paracrine growth stimulation .\n\nIntegration of CXCR4-targeting strategies into first- and/or second-line treatment regimens may represent a promising treatment option for Ewing sarcoma .", "output": "Activating invasion and metastasis" }, { "input": "The down-regulation of dominant oncogenes , including C-MYC , in tumor cells often leads to the induction of senescence via mechanisms that are not completely identified .\n\nIn the current study , we demonstrate that MYC-depleted melanoma cells undergo extensive DNA damage that is caused by the underexpression of thymidylate synthase ( TS ) and ribonucleotide reductase ( RR ) and subsequent depletion of deoxyribonucleoside triphosphate pools .\n\nSimultaneous genetic inhibition of TS and RR in melanoma cells induced DNA damage and senescence phenotypes very similar to the ones caused by MYC-depletion .\n\nReciprocally , overexpression of TS and RR in melanoma cells or addition of deoxyribo-nucleosides to culture media substantially inhibited DNA damage and senescence-associated phenotypes caused by C-MYC depletion .\n\nOur data demonstrate the essential role of TS and RR in C-MYC-dependent suppression of senescence in melanoma cells .", "output": "Genomic instability and mutation, Enabling replicative immortality" }, { "input": "The activity of DNA methyltransferase 1 ( DNMT1 ) is associated with diverse biological activities , including cell proliferation , senescence and cancer development .\n\nHere , we demonstrated that the HMG box-containing protein 1 ( HBP1 ) transcription factor is a new repressor of DNMT1 in a complex mechanism during senescence .\n\nThe DNMT1 gene contains an HBP1-binding site at position -115 to -134bp from the transcriptional start site .\n\nHBP1 repressed the endogenous DNMT1 gene through sequence-specific binding , resulting in both gene-specific ( e.g. p16(INK4) ) and global DNA hypomethylation changes .\n\nThe HBP1-mediated repression by DNMT1 contributed to replicative and premature senescence , the latter of which could be induced by Ras and HBP1 itself .\n\nA detailed investigation unexpectedly revealed that HBP1 has dual and complex transcriptional functions-both of which contribute to premature senescence .\n\nHBP1 both repressed the DNMT1 gene and activated the p16 gene in premature senescence .\n\nThe opposite transcriptional functions proceeded through different DNA sequences and differential protein acetylation .\n\nWhile intricate , the reciprocal partnership between HBP1 and DNMT1 has exceptional importance , since its abrogation compromises senescence and promotes tumorigenesis .\n\nTogether , our results suggest that the HBP1 transcription factor orchestrates a complex regulation of key genes during cellular senescence with an impact on overall DNA methylation state .", "output": "Enabling replicative immortality" }, { "input": "Androgens regulate both the physiological development of the prostate and the pathology of prostatic diseases .\n\nHowever , the mechanisms by which androgens exert their regulatory activities on these processes are poorly understood .\n\nIn this study , we have determined that androgens regulate overall cell metabolism and cell growth , in part , by increasing autophagy in prostate cancer cells .\n\nImportantly , inhibition of autophagy using either pharmacological or molecular inhibitors significantly abrogated androgen-induced prostate cancer cell growth .\n\nMechanistically , androgen-mediated autophagy appears to promote cell growth by augmenting intracellular lipid accumulation , an effect previously demonstrated to be necessary for prostate cancer cell growth .\n\nFurther , autophagy and subsequent cell growth is potentiated , in part , by androgen-mediated increases in reactive oxygen species .\n\nThese findings demonstrate a role for increased fat metabolism and autophagy in prostatic neoplasias and highlight the potential of targeting underexplored metabolic pathways for the development of novel therapeutics .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "The aim of the present work was to study the expression of the proinflammatory cytokine , interleukin-6 ( IL-6 ) , mediated by bFGF signaling and its possible crosstalk with prostate-specific membrane antigen ( PSMA ) in LNCaP and PC3-PSMA prostate cancer cell lines .\n\nPC3 cells stably transfected with PSMA gene were used for restoring PSMA expression .\n\nLNCaP and PC3-PSMA cells were exposed to 10ng/mL of basic fibroblast growth factor ( bFGF ) .\n\nIL-6 production was measured by ELISA assay , and levels of PSMA expression were assessed by flow cytometry .\n\nAKT , ERK1/2 , and p38 phosphorylation were detected by Western blot. bFGF enhances IL-6 production in LNCaP and PC3-PSMA prostate cancer cells .\n\nThe effect of bFGF on stimulating IL-6 secretion was greater in LNCaP than in PC3-PSMA cells .\n\nIn the presence of bFGF , PSMA expression was activated after 4days of treatment in LNCaP and PC3-PSMA cells .\n\nThis activation was not maintained after long term of treatment in both metastatic cell lines .\n\nSolely MAPKs pathways ( ERK1/2 and p38 ) were activated after bFGF stimulation in both metastatic cell lines , whereas AKT did not show any activation .\n\nThe interference of the proinflammatory cytokine , IL-6 , with bFGF signaling and PSMA , should be of high clinical relevance in the treatment of metastatic prostate cancer .\n\nIn developing novel therapeutic modalities targeting IL-6 , significant attention should be given to PSMA and its inactivation to fight against prostate cancer .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "MicroRNAs ( miRNAs or miR ) have been integrated into tumorigenic programs as either oncogenes or tumor suppressor genes .\n\nThe miR-124 was reported to be attenuated in several tumors , such as glioma , medulloblastoma and hepatocellular carcinoma .\n\nHowever , its role in cancer remains greatly elusive .\n\nIn this study , we show that the miR-124 expression is significantly suppressed in human breast cancer specimens , which is reversely correlated to histological grade of the cancer .\n\nMore intriguingly , ectopic expression of miR-124 in aggressive breast cancer cell lines MDA-MB-231 and BT-549 strongly inhibits cell motility and invasive capacity , as well as the epithelial-mesenchymal transition process .\n\nAlso , lentivirus-delivered miR-124 endows MDA-MB-231 cells with the ability to suppress cell colony formation in vitro and pulmonary metastasis in vivo .\n\nFurther studies have identified the E-cadherin transcription repressor Slug as a direct target gene of miR-124 ; its downregulation by miR-124 increases the expression of E-cadherin , a hallmark of epithelial cells and a repressor of cell invasion and metastasis .\n\nMoreover , knockdown of Slug notably impairs the motility of MDA-MB-231 cells , whereas re-expression of Slug abrogates the reduction of motility and invasion ability induced by miR-124 in MDA-MB-231 cells .\n\nThese findings highlight an important role for miR-124 in the regulation of invasive and metastatic potential of breast cancer and suggest a potential application of miR-124 in cancer treatment .", "output": "Activating invasion and metastasis" }, { "input": "The aim of this study was to evaluate the effects and molecular mechanisms of everolimus on Panc-1 human pancreatic cancer cells .\n\nPanc-1 human pancreatic cancer cells were treated with everolimus ( 10 \u03bcg/ml ) at selected time points ( 6 , 12 and 24 h ) .\n\nCell proliferation and apoptosis were evaluated by MTT and flow cytometric analyses .\n\nThe glycolytic activity was determined by measuring the activity of the key enzyme lactate dehydrogenase ( LDH ) and lactate production .\n\nThe activity of mammalian target of rapamycin ( mTOR ) signaling was measured by western blotting .\n\nThe expression of genes , including hexokinase 2 ( HK2 ) and microRNA-143 ( miR-143 ) , was evaluated by real-time polymerase chain reaction ( PCR ) .\n\nThe administration of everolimus time-dependently inhibited proliferation and glycolysis and induced apoptosis in the Panc-1 human pancreatic cancer cells .\n\nAs the time of treatment with everolimus increased , the mTOR signaling activity decreased , indicated by lower phosphorylation levels of S6 kinase ; however , the phosphorylation levels of mTOR barely changed .\n\nMoreover , our data showed an everolimus-induced increase in miR-143 and decrease in HK2 in Panc-1 cells in a time-dependent manner .\n\nIn conclusion , the current study indicates a novel role of everolimus in its antitumor effect as an inhibitor of glycolysis in Panc-1 human pancreatic cancer cells .\n\nFurthermore , our data highlights the significance of exploring the mechanisms of everolimus and miR-143 in malignant tumors .", "output": "Cellular energetics, Resisting cell death" }, { "input": "Testicular Germ Cell Tumors ( TGCT ) and patient-derived cell lines are extremely sensitive to cisplatin and other interstrand cross-link ( ICL ) inducing agents .\n\nNevertheless , a subset of TGCTs are either innately resistant or acquire resistance to cisplatin during treatment .\n\nUnderstanding the mechanisms underlying TGCT sensitivity/resistance to cisplatin as well as the identification of novel strategies to target cisplatin-resistant TGCTs have major clinical implications .\n\nHerein , we have examined the proficiency of five embryonal carcinoma ( EC ) cell lines to repair cisplatin-induced ICLs .\n\nUsing \\u03b3H2AX staining as a marker of double strand break formation , we found that EC cell lines were either incapable of or had a reduced ability to repair ICL-induced damage .\n\nThe defect correlated with reduced Homologous Recombination ( HR ) repair , as demonstrated by the reduction of RAD51 foci formation and by direct evaluation of HR efficiency using a GFP-reporter substrate .\n\nHR-defective tumors cells are known to be sensitive to the treatment with poly(ADP-ribose) polymerase ( PARP ) inhibitor .\n\nIn line with this observation , we found that EC cell lines were also sensitive to PARP inhibitor monotherapy .\n\nThe magnitude of sensitivity correlated with HR-repair reduced proficiency and with the expression levels and activity of PARP1 protein .\n\nIn addition , we found that PARP inhibition strongly enhanced the response of the most resistant EC cells to cisplatin , by reducing their ability to overcome the damage .\n\nThese results point to a reduced proficiency of HR repair as a source of sensitivity of ECs to ICL-inducing agents and PARP inhibitor monotherapy , and suggest that pharmacological inhibition of PARP can be exploited to target the stem cell component of the TGCTs ( namely ECs ) and to enhance the sensitivity of cisplatin-resistant TGCTs to standard treatments .", "output": "Genomic instability and mutation" }, { "input": "Multiple myeloma ( MM ) is a clonal disease of plasma cells that remains incurable despite the advent of several novel therapeutics .\n\nIn this study , we aimed to delineate the impact of snake venom extracted from Walterinnesia aegyptia ( WEV ) alone or in combination with silica nanoparticles ( WEV+NP ) on primary MM cells isolated from patients diagnosed with MM as well as on two MM cell lines , U266 and RPMI 8226 .\n\nThe IC(50) values of WEV and WEV+NP that significantly decreased MM cell viability without affecting the viability of normal peripheral mononuclear cells ( PBMCs ) were determined to be 25 ng/ml and 10 ng/ml , respectively .\n\nAlthough both WEV ( 25 ng/ml ) and WEV+NP ( 10 ng/ml ) decreased the CD54 surface expression without affecting the expression of CXCR4 ( CXCL12 receptor ) on MM cells , they significantly reduced the ability of CXC chemokine ligand 12 ( CXCL12 ) to induce actin cytoskeleton rearrangement and the subsequent reduction in chemotaxis .\n\nIt has been established that the binding of CXCL12 to its receptor CXCR4 activates multiple intracellular signal transduction pathways that regulate MM cell chemotaxis , adhesion , and proliferation .\n\nWe found that WEV and WEV+NP clearly decreased the CXCL12/CXCR4-mediated activation of AKT , ERK , NF\u03baB and Rho-A using western blot analysis ; abrogated the CXCL12-mediated proliferation of MM cells using the CFSE assay ; and induced apoptosis in MM cell as determined by PI/annexin V double staining followed by flow cytometry analysis .\n\nMonitoring the expression of B-cell CCL/Lymphoma 2 ( Bcl-2 ) family members and their role in apoptosis induction after treatment with WEV or WEV+NP revealed that the combination of WEV with NP robustly decreased the expression of the anti-apoptotic effectors Bcl-2 , Bcl(XL) and Mcl-1 ; conversely increased the expression of the pro-apoptotic effectors Bak , Bax and Bim ; and altered the mitochondrial membrane potential in MM cells .\n\nTaken together , our data reveal the biological effects of WEV and WEV+NP and the underlying mechanisms against myeloma cancer cells .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Heparin-binding epidermal growth factor-like growth factor ( HB-EGF ) is a member of the epidermal growth factor family and has a variety of physiological and pathological functions .\n\nModulation of HB-EGF activity might have a therapeutic potential in the oncology area .\n\nWe explored the therapeutic possibilities by characterizing the in vitro biological activity of anti-HB-EGF monoclonal antibody Y-142 .\n\nEGF receptor ( EGFR ) ligand and species specificities of Y-142 were tested .\n\nNeutralizing activities of Y-142 against HB-EGF were evaluated in EGFR and ERBB4 signaling .\n\nBiological activities of Y-142 were assessed in cancer cell proliferation and angiogenesis assays and compared with the anti-EGFR antibody cetuximab , the HB-EGF inhibitor CRM197 , and the anti-vascular endothelial growth factor ( VEGF ) antibody bevacizumab .\n\nThe binding epitope was determined with alanine scanning .\n\nY-142 recognized HB-EGF as well as the EGFR ligand amphiregulin , and bound specifically to human HB-EGF , but not to rodent HB-EGF .\n\nIn addition , Y-142 neutralized HB-EGF-induced phosphorylation of EGFR and ERBB4 , and blocked their downstream ERK1/2 and AKT signaling .\n\nWe also found that Y-142 inhibited HB-EGF-induced cancer cell proliferation , endothelial cell proliferation , tube formation , and VEGF production more effectively than cetuximab and CRM197 and that Y-142 was superior to bevacizumab in the inhibition of HB-EGF-induced tube formation .\n\nSix amino acids in the EGF-like domain were identified as the Y-142 binding epitope .\n\nAmong the six amino acids , the combination of F115 and Y123 determined the amphiregulin cross-reactivity and that F115 accounted for the species selectivity .\n\nFurthermore , it was suggested that the potent neutralizing activity of Y-142 was derived from its recognition of R142 and Y123 and its high affinity to HB-EGF .\n\nY-142 has a potent HB-EGF neutralizing activity that modulates multiple biological activities of HB-EGF including cancer cell proliferation and angiogenic activities .\n\nY-142 may have a potential to be developed into a therapeutic agent for the treatment of HB-EGF-dependent cancers .", "output": "Inducing angiogenesis, Sustaining proliferative signaling" }, { "input": "Breast cancer incidence is increased in women receiving menopausal hormone therapy with estrogen plus progestin but not with estrogen alone .\n\nThe use of a tissue-selective estrogen complex ( TSEC ) has been proposed as a novel menopausal hormone therapy strategy to eliminate the requirement for a progestogen .\n\nCombination of bazedoxifene ( BZA ) with conjugated estrogens ( CEs ) , the first TSEC , has shown beneficial effects .\n\nWhether it would exert antiestrogenic effects on breast cancer is not clear .\n\nTo address this issue , we compared estradiol ( E(2) ) and CE alone on proliferation and apoptosis in MCF-7 breast cancer cells .\n\nCE stimulated growth of MCF-7 cells at a peak concentration 10-fold higher than required for E(2) .\n\nBoth CE and E(2) alone increased DNA synthesis and reduced apoptosis with activation of MAPK , Akt , and p70S6K and up-regulation of antiapoptotic factors survivin , Bcl-2 , and X-linked inhibitor of apoptosis protein , These effects could be completely blocked by BZA .\n\nGene expression studies demonstrated that CE and E(2) were equally potent on expression of cMyc , pS2 , and WNT1 inducible signaling pathway protein 2 , whereas the stimulatory effects of CE on progesterone receptor and amphiregulin expression were weaker than E(2) .\n\nBZA effectively blocked each of these effects and showed no estrogen agonistic effects when used alone .\n\nOur results indicate that the stimulatory effects of E(2) or CE on breast cancer cells could be completely abrogated by BZA .\n\nThese studies imply that the CE/BZA , TSEC , exerts antiestrogenic effects on breast cancer cells and might block the growth of occult breast neoplasms in postmenopausal women , resulting in an overall reduction in tumor incidence .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Gliomas display anoikis resistance , enhanced invasion in to the adjacent brain parenchyma and eventually recur despite using the standard therapies .\n\nOur studies on increased anoikis sensitization in matrix metalloproteinase-2 ( MMP-2)-knockdown 4910 and 5310 human glioma xenograft cells were interestingly correlated with p21-activated kinase 4 ( PAK4 ) inhibition , prompting us to further investigate the role of PAK4 in glioma .\n\nHere , we report the PAK4 upregulation in positive correlation with increasing glioma pathological grades .\n\nThe siRNA-mediated PAK4 knockdown elevated anoikis , and inhibited invasion and migration by downregulating MMP-2 , \u03b1v\u03b23-integrin and phospho-epidermal growth factor receptor ( phospho-EGFR ) .\n\nThe cDNA-PCR arrays revealed a transcriptional suppression of essential proteins involved in cell proliferation and adhesion in PAK4-knockdown cells .\n\nMost importantly , glutathione S-transferase pull-down assays demonstrated the MMP-2 as a new PAK4-interacting protein which binds to PAK4 kinase domain .\n\nIndividual EGFR/ErbB2 inhibitor and \u03b1v\u03b23 antibody treatments in PAK4si-treated cells indicated the regulation of \u03b1v\u03b23/EGFR survival signaling by PAK4 .\n\nOverexpression of PAK4 significantly reversed the MMP2si-induced cell death in both cell lines .\n\nCodepletion of PAK4 and MMP-2 resulted in robust anoikis-mediated cell death , and severely inhibited invasive and migratory properties in these cells .\n\nPAK4si inhibited in vivo tumor growth in nude mice by inhibiting MMP-2 , \u03b23-integrin and phospho-EGFR levels in tumors .\n\nOur findings indicate a physical association between PAK4 and MMP-2 , and suggest the future therapeutic potential of PAK4/MMP-2 dual targeting in glioma treatment .", "output": "Sustaining proliferative signaling, Resisting cell death, Activating invasion and metastasis" }, { "input": "Lowering the threshold of cellular senescence , the process employed by cells to thwart abnormal cell proliferation , though inhibition of CDK2 or Skp2 ( regulator of CDK inhibitors ) has been recently suggested as a potential avenue for cancer treatment .\n\nIn this study , we employ a published mathematical model of G1/S transition involving the DNA-damage signal transduction pathway to conduct carefully constructed computational experiments to highlight the effectiveness of manipulating cellular senescence in inhibiting damaged cell proliferation .\n\nWe first demonstrate the suitability of the mathematical model to explore senescence by highlighting the overlap between senescence pathways and those involved in G1/S transition and DNA damage signal transduction .\n\nWe then investigate the effect of CDK2 deficiency on senescence in healthy cells , followed by effectiveness of CDK2 deficiency in triggering senescence in DNA damaged cells .\n\nFor this , we focus on the behaviour of CycE , whose peak response indicates G1/S transition , for several reduced CDK2 levels in healthy as well as two DNA-damage conditions to calculate the probability ( \\u03b2 ) or the percentage of CDK2 deficient cells passing G1/S checkpoint ( (1-\\u03b2) indicates level of senescence ) .\n\nResults show that 50% CDK2 deficiency can cause senescence in all healthy cells in a fairly uniform cell population ; whereas , most healthy cells ( \\u224867% ) in a heterogeneous population escape senescence .\n\nThis finding is novel to our study .\n\nUnder both low- and high-DNA damaged conditions , 50% CDK deficiency can cause 65% increase in senescence in a heterogeneous cell population .\n\nFurthermore , the model analyses the relationship between CDK2 and its CKIs ( p21 , p27 ) to help search for other effective ways to bring forward cellular senescence .\n\nResults show that the degradation rate of p21 and initial concentration of p27 are effective in lowering CDK2 levels to lower the senescence threshold .\n\nSpecifically , CDK2 and p27 are the most effective in triggering senescence while p21 having a smaller influence .\n\nWhile receiving experimental support , these findings specify in detail the inhibitory effects of CKIs .\n\nHowever , simultaneous variation of CDK2 and CKIs produces a dramatic reduction of damage cells passing the G1/S with CDK2&p27 combination causing senescence in almost all damaged cells .\n\nThis combined effect of CDK2&CKIs on senescence is a novel contribution in this study .\n\nA review of the crucial protein complexes revealed that the concentration of active CycE/CDK2-p that controls cell cycle arrest provides support for the above findings with CycE/CDK2-p undergoing the largest reduction ( over 100% ) under the combined CDK2&CKI conditions leading to the arrest of most of the damaged cells .\n\nOur study thus provides quantitative assessments for the previously published qualitative findings on senescence and highlights new avenues for bringing forward senescence bar .", "output": "Genomic instability and mutation, Enabling replicative immortality, Evading growth suppressors" }, { "input": "Aurora A kinase has drawn considerable attention as a therapeutic target for cancer therapy .\n\nHowever , the underlying molecular and cellular mechanisms of the anticancer effects of Aurora A kinase inhibition are still not fully understood .\n\nHerein , we show that depletion of Aurora A kinase by RNA interference ( RNAi ) in hepatocellular carcinoma ( HCC ) cells upregulated FoxO1 in a p53-dependent manner , which induces cell cycle arrest .\n\nIntroduction of an RNAi-resistant Aurora A kinase into Aurora A-knockdown cells resulted in downregulation of FoxO1 expression and rescued proliferation .\n\nIn addition , silencing of FoxO1 in Aurora A-knockdown cells allowed the cells to exit cytostatic arrest , which , in turn , led to massive cell death .\n\nOur results suggest that FoxO1 is responsible for growth arrest at the G 2/M phase that is induced by Aurora A kinase inhibition .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "It is now largely accepted that ribosomal proteins may be implicated in a variety of biological functions besides that of components of the translation machinery .\n\nMany evidences show that a subset of ribosomal proteins are involved in the regulation of the cell cycle and apoptosis through modulation of p53 activity .\n\nIn addition , p53-independent mechanisms of cell cycle arrest in response to alterations of ribosomal proteins availability have been described .\n\nHere , we identify human rpL3 as a new regulator of cell cycle and apoptosis through positive regulation of p21 expression in a p53-independent system .\n\nWe demonstrate that the rpL3-mediated p21 upregulation requires the specific interaction between rpL3 and Sp1 .\n\nFurthermore , in our experimental system , p21 overexpression leads to a dual outcome , activating the G\\u2081/S arrest of the cell cycle or the apoptotic pathway through mitochondria , depending on its intracellular levels .\n\nIt is noteworthy that depletion of p21 abrogates both effects .\n\nTaken together , our findings unravel a novel extraribosomal function of rpL3 and reinforce the proapoptotic role of p21 in addition to its widely reported ability as an inhibitor of cell proliferation .", "output": "Evading growth suppressors, Resisting cell death" }, { "input": "BACKGROUND: : Radiotherapy is an efficient remedy in the treatment of hepatocellular carcinoma ( Hca ) ; however , some cancer cells can still survive from the radiation and the therapeutic effect is to be improved .\n\nRegulatory T cell ( Tregs)-induced tumor tolerance and Akt expression play important roles in the tumor survival .\n\nThis study aims to elucidate the role of radiation induces Akt expression in Tregs .\n\nMETHODS: : A rat Hca model was developed .\n\nHca tissue was collected from the rats with or without radiotherapy .\n\nThe frequency of Treg and apoptotic Treg in Hca tissue was assessed by flow cytometry .\n\nA cell culture model was used to investigate the mechanism by which the tumor-infiltrating Tregs survive from irradiation .\n\nRESULTS: : After radiotherapy , the frequency of Treg was increased in Hca , the frequency of apoptotic Tregs was decreased and the expression of Akt was increased in the remaining Tregs .\n\nThe results were reproduced in vitro with a cell culture model .\n\nThe addition of Akt inhibitor blocked the irradiation-induced Treg survival .\n\nTregs with high levels of Akt still preserve the immune suppressor function .\n\nCONCLUSIONS: : Akt plays an important role in the radiation-induced tumor-infiltrating Treg survival in Hca .", "output": "Avoiding immune destruction, Resisting cell death" }, { "input": "Environmental or occupational exposure to low doses of arsenic induces a series of health problems including cancer .\n\nThe molecular events in arsenic-induced carcinogenicity remain to be defined .\n\nIn the NuLi-1 immortalized human lung epithelial cell line with p53 and pRb deficiency , exposure to low doses of arsenic trioxide for 72\u2009h promoted cell proliferation and upregulated the gene transcription levels of FOXM1 , CDC6 , CDC25A , and cyclin D1 , which are both critical cell cycle regulatory genes and proto-oncogenes .\n\nContinuous in vitro exposure to 1\u2009\ufffdM arsenic trioxide for 34 wks induced malignant cell transformation , as evidenced by enhanced anchorage-independent cell growth .\n\nThe expression of FOXM1 , CDC6 , CDC25A , and Cyclin D1 was dynamically elevated at the gene transcription and protein levels in the process of cell transformation .\n\nThe carcinogenic ability of transformed cell colonies coincides with the expression levels of FOXM1 in in vitro anchorage-independent growth assays and in vivo tumor xenograft formation assays .\n\nIn reverse , the knockdown of FOXM1 in lung adenocarcinoma A549 cells or arsenic-transformed NuLi-1 cells significantly decreased anchorage-independent cell growth and tumor xenograft formation .\n\nThe transformed NuLi-1 cells showed genomic instability in the form of copy number variation ( CNV ) at chromosome 1 , 5 , 6 , 18 , and 20 , but not loss of heterozygosity ( LOH ) .\n\nThese results showed for the first time that chronic exposure to low doses of arsenic trioxide promoted lung carcinogenicity , in part by aberrantly upregulating FOXM1 and its associated oncogenes , when the tumor suppressor genes p53 and pRb were inactivated. \ufffd 2012 Wiley Periodicals , Inc .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "cAMP-signaling plays an essential role in modulating the proliferation of different cell types , including cancer cells .\n\nUntil now , the regulation of this pathway was restricted to the transmembrane class of adenylyl cyclases .\n\nIn the present study , a significant overexpression of soluble adenylyl cyclase ( sAC ) , an alternative source of cAMP , was found in human prostate carcinoma and , therefore , the contribution of this cyclase was investigated in the prostate carcinoma cell lines LNCaP and PC3 .\n\nSuppression of sAC activity by treatment with the sAC-specific inhibitor KH7 or by sAC-specific knockdown mediated by siRNA or shRNA transfection prevented the proliferation of prostate carcinoma cells , led to lactate dehydrogenase release , and induced apoptosis .\n\nCell cycle analysis revealed a significant rise in the G2-phase population 12 hours after sAC inhibition , which was accompanied by the down-regulation of cyclin B1 and CDK1. sAC-dependent regulation of proliferation involves the EPAC/Rap-1/B-Raf signaling pathway .\n\nIn contrast , protein kinase A does not play a role .\n\nIn conclusion , the present study suggests a novel sAC-dependent signaling pathway that controls the proliferation of prostate carcinoma cells .", "output": "Resisting cell death" }, { "input": "Capsaicin , one of the major pungent ingredients found in red peppers , has been recently demonstrated to induce apoptosis in various malignant cell lines through an unclear mechanism .\n\nIn this study , the effect of capsaicin on proliferation and apoptosis in the human pancreatic cancer cell line PANC-1 and its possible mechanism(s) of action were investigated .\n\nThe results of a Cell Counting Kit-8 ( CCK-8 ) assay revealed that capsaicin significantly decreased the viability of PANC-1 cells in a dose-dependent manner .\n\nCapsaicin induced G0/G1 phase cell cycle arrest and apoptosis in PANC-1 cells as demonstrated by a flow cytometric assessment .\n\nCaspase-3 expression at both the protein and mRNA level was promoted following capsaicin treatment .\n\nFurthermore , we revealed that phospho-PI3 Kinase p85 ( Tyr458 ) and phospho-Akt ( Ser473 ) in PANC-1 cells were downregulated in response to capsaicin .\n\nMoreover , capsaicin gavage significantly inhibited the growth of pancreatic cancer PANC-1 cell xenografts in athymic nude mice .\n\nAn increased number of TUNEL-positive cells and cleaved caspase-3 were observed in capsaicin-treated mice .\n\nIn vivo , capsaicin downregulated the expression of phospho-PI3 Kinase p85 ( Tyr458 ) and phospho-Akt ( Ser473 ) .\n\nIn conclusion , we have demonstrated that capsaicin is an inhibitor of growth of PANC-1 cells , and downregulation of the phosphoinositide 3-kinase/Akt pathway may be involved in capsaicin-induced apoptosis in vitro and in vivo .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Sorafenib is a multi-targeted agent and has been reported to have potent antitumor effects against various types of tumors , including human non-small cell lung cancer ( NSCLC ) .\n\nIn this study , we explored in vitro the antitumor effects of sorafenib alone and in combination with gemcitabine in epidermal growth factor receptor-tyrosine kinase inhibitor ( EGFR-TKI)-resistant human lung cancer cell lines and the related molecular mechanisms .\n\nThe NSCLC cell lines A549 ( mutant KRAS ) , H1666 ( mutant BRAF ) and H1975 ( mutant EGFR-T790M ) were treated with sorafenib and gemcitabine alone and in combination .\n\nThe cytotoxicity was assessed by MTT assay , cell cycle distribution was analyzed by flow cytometry , and alterations in signaling pathways were analyzed by western blotting .\n\nWe found that sorafenib exhibited dose-dependent growth inhibition in all three EGFR-TKI-resistant NSCLC cell lines .\n\nWhen sorafenib was combined with gemcitabine , synergistic activity was observed in the A549 and H1666 cells and antagonistic activity was observed in the H1975 cells .\n\nSorafenib arrested the cell cycle at the G1 phase , whereas gemcitabine arrested the cell cycle at the S phase .\n\nSorafenib inhibited C-RAF and p-ERK in the A549 cells and B-RAF and p-ERK in the H1666 and H1975 cells .\n\nThe molecular mechanism of this synergism is that RAF/MEK/ERK which are activated by gemcitabine are efficiently suppressed by simultaneously administered sorafenib .\n\nBy contrast , the mechanism of antagonism may be due to mutual interference with the cell cycle in the H1975 cells .\n\nIn conclusion , we found that sorafenib exhibits antiproliferative effects in EGFR-TKI-resistant NSCLC cell lines and when combined with gemcitabine demonstrates synergistic activity in A549 and H1666 cells but antagonistic activity in H1975 cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Cancer immunotherapies are designed to elicit T-cell responses that inhibit tumor growth .\n\nPrevious studies have demonstrated that interleukin 21 ( IL-21 ) is a promising cytokine for cancer immunotherapy due to its ability to induce the immunity of T cells and natural killer cells , whereas blockade of the interaction of programmed death receptor-1 ( PD-1 ) with its ligand ( PD-L1 ) reduces peripheral tolerance .\n\nIn the current study , we investigated IL-21 alone and in combination with soluble PD-1 ( sPD-1 ) for the treatment of experimental H22 murine hepatocarcinoma .\n\nThe naked plasmids pmIL-21 and/or psPD-1 were used for local gene transfer by injection .\n\nIn these assays , sPD-1 combined with IL-21 was found to significantly inhibit the growth of the tumors in mice .\n\nCombined treatment with IL-21 and sPD-1 enhanced the antitumor immune response compared with that induced by IL-21 alone .\n\nCombined treatment was found to increase CTL cytotoxicity , increase the number of CTLs and NK cells in splenocytes , upregulate the cytokines IFN-\u03b3 and IL-2 and downregulate IL-10 .\n\nThus , immunotherapy with IL-21 in combination with sPD-1 was found to induce a more efficacious antitumor immune response , which may have potential clinical implications .", "output": "Avoiding immune destruction" }, { "input": "Cancer therapy with rapamycin has been successfully implemented for kidney cancer , glioblastoma and prostate cancer .\n\nHowever , there are few studies concerning the effects of rapamycin on the treatment of human melanoma .\n\nIn this study , we investigated whether rapamycin may be a promising strategy for the effective treatment of melanoma and explored the possible mechanism for this by culturing M14 cells in vitro and treating with rapamycin at three concentrations ( 10 , 50 or 100 nmol/l ) .\n\nMDC and LC3B staining , western blot analysis , flow cytometry and transmission electron microscopy were employed .\n\nWe revealed that rapamycin induced autophagy and inhibited the proliferation of M14 cells in a concentration-dependent manner , Furthermore , western blot analysis revealed an upregulated expression of Bcl-2 and downregulated expression of Bax in M14 cells .\n\nIn conclusion , rapamycin induced autophagy and inhibited the growth of M14 cells .\n\nThe mechanism may involve regulation of the expression of Bcl-2 family proteins .\n\nRapamycin appears to be a promising strategy for the effective treatment of melanoma .", "output": "Resisting cell death" }, { "input": "Radix of Asiasarum heterotropoides var. mandshuricum F. Maekawa ( A. radix ) has been prescribed for treating pain , allergies and inflammatory disorders in traditional oriental medicine .\n\nHowever , only limited information on the anticancer effects of A. radix is currently available .\n\nThe aim of this study was to determine the anticancer effect of the ethanol extract of A. radix ( EEAR ) on HCT-116 human colon cancer cells and to investigate its underlying mechanisms of action .\n\nEEAR significantly induced G2/M cell cycle arrest and apoptosis in HCT-116 cells .\n\nEEAR-induced apoptosis was observed in parallel with activation of caspases and an increased ratio of Bax ( pro-apoptotic)/Bcl2 ( anti-apoptotic ) .\n\nWestern blot analyses revealed that EEAR elevated the expression of p53 and p21(Waf/Cip1) and decreased the expression of the regulator proteins of G2/M phase progression , such as cdc2 and cyclin B. The upregulation of p53 by EEAR was due to the increased levels of p53 mRNA without a similar increase in proteasome-mediated p53 degradation .\n\nEEAR-induced apoptosis in HCT-116 cells was dependent on p53 expression , as determined by siRNA-mediated p53 knockdown .\n\nTaken together , these results suggest that EEAR inhibits the growth of the HCT-116 cells through induction of G2/M cell cycle arrest and apoptosis , which are mediated by p53 expression .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "The prediction of response or severe toxicity and therapy individualisation are extremely important in cancer chemotherapy .\n\nThere are few tools to predict chemoresponse or toxicity in cancer patients .\n\nWe investigated the correlation between the induction and repair of DNA double-strand breaks ( DSBs ) using constant-field gel electrophoresis ( CFGE ) and evaluating cell cycle progression and the sensitivity of four cancer cell lines to 5-fluorouracil ( 5FU ) .\n\nUsing a sulphorhodamine-B assay , colon carcinoma cells ( HCT116 ) were found to be the most sensitive to 5FU , followed by liver carcinoma cells ( HepG2 ) and breast carcinoma cells ( MCF-7 ) .\n\nCervical carcinoma cells ( HeLa ) were the most resistant .\n\nAs measured by CFGE , DSB induction , but not residual DSBs , exhibited a significant correlation with the sensitivity of the cell lines to 5FU .\n\nFlow cytometric cell cycle analysis revealed that 14% of HCT116 or HepG2 cells and 2% of MCF-7 cells shifted to sub-G1 phase after a 96-h incubation with 5FU .\n\nAnother 5FU-induced cell cycle change in HCT116 , HepG2 and MCF-7 cells was the mild arrest of cells in G1 and/or G2/M phases of the cell cycle .\n\nIn addition , 5FU treatment resulted in the accumulation of HeLa cells in the S and G2/M phases .\n\nDetermination of Fas ligand ( Fas-L ) and caspase 9 as representative markers for the extrinsic and intrinsic pathways of apoptosis , respectively , revealed that 5FU-induced apoptosis in HCT116 and HepG2 results from the expression of Fas-L ( extrinsic pathway ) .\n\nTherefore , the induction of DNA DSBs by 5FU , detected using CFGE , and the induction of apoptosis are candidate predictive markers that may distinguish cancer cells which are likely to benefit from 5FU treatment and the measurement of DSBs using CFGE may aid the prediction of clinical outcome .", "output": "Genomic instability and mutation, Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Piceatannol has potent anti-inflammatory , immunomodulatory , anticancer and antiproliferative effects .\n\nHowever , little is known about the mechanism by which piceatannol inhibits invasion and metastasis .\n\nThe aim of the current study was to investigate the effects of piceatannol on the expression of matrix metalloproteinase-9 ( MMP-9 ) in DU145 human prostate cancer cells .\n\nThe results revealed that MMP-9 activity was significantly increased in response to tumor necrosis factor-\\u03b1 ( TNF-\\u03b1 ) .\n\nHowever , treatment with piceatannol reversed TNF-\\u03b1- and MMP-9-induced gelatin zymography and its gene expression .\n\nIn addition , a Matrigel invasion assay determined that piceatannol reduces the TNF-\\u03b1-induced invasion of DU145 cells .\n\nNuclear factor-\\u03ba B ( NF-\\u03baB ) is a significant transcription factor that regulates numerous genes involved in tumor cell invasion and metastasis .\n\nTherefore , whether piceatannol acts on NF-\\u03baB to regulate MMP-9 gene expression was analyzed .\n\nThe results revealed that piceatannol attenuates MMP-9 gene expression via the suppression of NF-\\u03baB activity .\n\nUsing a specific NF-\\u03baB inhibitor , pyrrolidine dithiocarbamate , it was confirmed that TNF-\\u03b1-induced MMP-9 gene expression is primarily regulated by NF-\\u03baB activation .\n\nPiceatannol inhibited NF-\\u03baB activity by suppressing nuclear translocation of the NF-\\u03baB p65 and p50 subunits .\n\nFurthermore , TNF-\\u03b1-induced Akt phosphorylation was significantly downregulated in the presence of piceatannol .\n\nThe Akt inhibitor LY294002 caused a significant decrease in TNF-\\u03b1-induced NF-\\u03baB activity and MMP-9 gene expression .\n\nOverall , these data suggest that piceatannol inhibits TNF-\\u03b1-induced invasion by suppression of MMP-9 activation via the Akt-mediated NF-\\u03baB pathway in DU145 prostate cancer cells .", "output": "Activating invasion and metastasis" }, { "input": "Gefitinib , the specific inhibitor of the epidermal growth factor receptor ( EGFR ) , may cause growth delay in cancer cell lines .\n\nThorough understanding of the downstream cellular signaling of gefitinib will facilitate the discovery of biomarkers for predicting outcomes and monitoring anti-EGFR therapies , and provide information for key targets for therapeutic intervention .\n\nIn this study , we investigated the role of transducer of erbB2.1 ( TOB1 ) in gefitinib therapy .\n\nUsing the lung carcinoma cell lines A549 and NCI-H1975 , the results suggested that gefitinib might mediate cell cycle arrest in lung cancer cells at least by targeting TOB1 expression .\n\nGefitinib treatment caused cell cycle arrest predominantly at the G1 phase , which is associated with TOB1 nuclear translocation and its interaction with cyclin D1 .\n\nWe also showed that knockdown of TOB1 expression by RNAi rescued lung cancer cells from gefitinib-induced cell-proliferative arrest .\n\nThese results suggest that TOB1 interaction with cyclin D1 and nuclear translocation is directly involved in the gefitinib-induced anti-proliferative cell cycle arrest .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Acute leukemia is a disorder of the hematopoietic system characterized by the expansion of a clonal population of cells blocked from differentiating into mature cells .\n\nRecent studies have shown that chalcones and their derivatives induce apoptosis in different cell lines .\n\nSince new compounds with biological activity are needed , the aim of this study was to evaluate the cytotoxic effect of three synthetic chalcones , derived from 1-naphthaldehyde and 2-naphthaldehyde , on human acute myeloid leukemia K562 cells and on human acute lymphoblastic leukemia Jurkat cells .\n\nBased on the results , the most cytotoxic compound ( A1 ) was chosen for further analysis in six human acute leukemia cells and in a human colon adenocarcinoma cell line ( HT-29 ) .\n\nChalcone A1 significantly reduced the cell viability of K562 , Jurkat , Kasumi , U937 , CEM and NB4 cells in a concentration and time-dependent manner when compared with the control group ( IC(50) values between \u223c1.5\u03bcM and 40\u03bcM ) .\n\nIt was also cytotoxic to HL-29 cells .\n\nTo further examine its effect on normal cells , peripheral blood lymphocytes collected from healthy volunteers were incubated with the compound .\n\nIt has also been incubated with human fibroblasts cultured from bone marrow ( JMA ) .\n\nChalcone A1 is non-cytotoxic to PBL cells and to JMA cells .\n\nA1 caused significant cell cycle arrest in all phases according to the cell line , and increased the proportion of cells in the sub G0/G1 phase .\n\nTo evaluate whether this chalcone induced cell death via an apoptotic or necrotic pathway , cell morphology was examined using fluorescence microscopy .\n\nCells treated with A1 at IC(50) demonstrated the morphological characteristic of apoptosis , such as chromatin condensation and formation of apoptotic bodies .\n\nApoptosis was confirmed by externalization of phosphatidylserine , which was detected by the Annexin V-FITC method , and by DNA fragmentation .\n\nThe results suggest that chalcone A1 has potential as a new lead compound for cancer therapy .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Metformin is a well-established diabetes drug that prevents the onset of most types of human cancers in diabetic patients , especially by targeting cancer stem cells .\n\nMetformin exerts its protective effects by functioning as a weak \" mitochondrial poison, \" as it acts as a complex I inhibitor and prevents oxidative mitochondrial metabolism ( OXPHOS ) .\n\nThus , mitochondrial metabolism must play an essential role in promoting tumor growth .\n\nTo determine the functional role of \" mitochondrial health \" in breast cancer pathogenesis , here we used mitochondrial uncoupling proteins ( UCPs ) to genetically induce mitochondrial dysfunction in either human breast cancer cells ( MDA-MB-231 ) or cancer-associated fibroblasts ( hTERT-BJ1 cells ) .\n\nOur results directly show that all three UCP family members ( UCP-1/2/3 ) induce autophagy and mitochondrial dysfunction in human breast cancer cells , which results in significant reductions in tumor growth .\n\nConversely , induction of mitochondrial dysfunction in cancer-associated fibroblasts has just the opposite effect .\n\nMore specifically , overexpression of UCP-1 in stromal fibroblasts increases \u03b2-oxidation , ketone body production and the release of ATP-rich vesicles , which \" fuels \" tumor growth by providing high-energy nutrients in a paracrine fashion to epithelial cancer cells .\n\nHence , the effects of mitochondrial dysfunction are truly compartment-specific .\n\nThus , we conclude that the beneficial anticancer effects of mitochondrial inhibitors ( such as metformin ) may be attributed to the induction of mitochondrial dysfunction in the epithelial cancer cell compartment .\n\nOur studies identify cancer cell mitochondria as a clear target for drug discovery and for novel therapeutic interventions .", "output": "Resisting cell death" }, { "input": "BACKGROUND Sorafenib has recently been shown to reduce tumour growth in hepatoblastoma ( HB ) xenografts .\n\nThe effect of a combined administration with cytostatic agents was now investigated .\n\nMETHODS Cell viability after treatment with sorafenib and different cytostatic agents was evaluated in two HB cell lines ( HUH6 and HepT1 ) using MTT assay .\n\nERK signalling was investigated by western blot , NOXA expression by rt-PCR , and formation of DNA adducts using immunocytology .\n\nNMRI mice bearing subcutaneous HUH6-derived tumours were treated with sorafenib alone or in combination with cisplatin .\n\nTumour progression , viability , apoptosis , and vascularisation were monitored by tumour volume , AFP levels , TUNEL assay , and CD31 immunostaining , respectively .\n\nRESULTS The combination of sorafenib and cisplatin led to a remarkable decrease in cell viability .\n\nThe cisplatin-induced enhanced ERK1/2 activation , but not NOXA expression and the formation of DNA adducts was partly abrogated by sorafenib .\n\nIn HB xenografts , both , sorafenib and alternated application of sorafenib and cisplatin significantly reduced tumour growth ( P<0.05 ) .\n\nLevels of AFP were lower in both treated groups ( P=0.08 ) .\n\nRelative apoptotic areas were increased ( P=0.003 ) .\n\nMean vascular density was the lowest in the sorafenib/CDDP group ( P=0.02 ) .\n\nCONCLUSION The combination of sorafenib with cisplatin might be a promising treatment option for high risk or recurrent HB .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "Background:Sorafenib is the only drug approved for the treatment of hepatocellular carcinoma ( HCC ) .\n\nThe bioenergetic propensity of cancer cells has been correlated to anticancer drug resistance , but such correlation is unclear in sorafenib resistance of HCC.Methods:Six sorafenib-naive HCC cell lines and one sorafenib-resistant HCC cell line ( Huh-7R ; derived from sorafenib-sensitive Huh-7 ) were used .\n\nThe bioenergetic propensity was calculated by measurement of lactate in the presence or absence of oligomycin .\n\nDichloroacetate ( DCA ) , a pyruvate dehydrogenase kinase ( PDK ) inhibitor , and siRNA of hexokinase 2 ( HK2 ) were used to target relevant pathways of cancer metabolism .\n\nCell viability , mitochondrial membrane potential , and sub-G1 fraction were measured for in vitro efficacy .\n\nReactive oxygen species ( ROS ) , adenosine triphosphate ( ATP ) and glucose uptake were also measured .\n\nA subcutaneous xenograft mouse model was used for in vivo efficacy.Results:The bioenergetic propensity for using glycolysis correlated with decreased sorafenib sensitivity ( R(2)=0.9067 , among sorafenib-naive cell lines ; P=0.003 , compared between Huh-7 and Huh-7\u2009R ) .\n\nDCA reduced lactate production and increased ROS and ATP , indicating activation of oxidative phosphorylation ( OXPHOS ) .\n\nDCA markedly sensitised sorafenib-resistant HCC cells to sorafenib-induced apoptosis ( sub-G1 ( combination vs sorafenib ) : Hep3B , 65.4\u00b18.4% vs 13\u00b12.9% ; Huh-7\u2009R , 25.3\u00b1 5.7% vs 4.3\u00b11.5% ; each P<0.0001 ) , whereas siRNA of HK2 did not .\n\nSorafenib ( 10\u2009mg\u2009kg(-1) per day ) plus DCA ( 100\u2009mg\u2009kg(-1) per day ) also resulted in superior tumour regression than sorafenib alone in mice ( tumour size : -87% vs -36% , P<0.001).Conclusion:The bioenergetic propensity is a potentially useful predictive biomarker of sorafenib sensitivity , and activation of OXPHOS by PDK inhibitors may overcome sorafenib resistance of HCC.British Journal of Cancer advance online publication , 20 December 2012 ; doi:10.1038/bjc.2012.559 www.bjcancer.com .", "output": "Tumor promoting inflammation, Cellular energetics, Resisting cell death" }, { "input": "Trinucleotide repeat ( TNR ) expansions and deletions are associated with human neurodegenerative diseases and prostate cancer .\n\nRecent studies have pointed to a linkage between oxidative DNA damage , base excision repair ( BER ) and TNR expansion , which is demonstrated by the observation that DNA polymerase \\u03b2 ( pol \\u03b2 ) gap-filling synthesis acts in concert with alternate flap cleavage by flap endonuclease 1 ( FEN1 ) to mediate CAG repeat expansions .\n\nIn this study , we provide the first evidence that the repair of a DNA base lesion can also contribute to CAG repeat deletions that were initiated by the formation of hairpins on both the template and the damaged strand of a continuous run of ( CAG)(20 ) or ( CAG)(25 ) repeats .\n\nMost important , we found that pol \\u03b2 not only bypassed one part of the large template hairpin but also managed to pass through almost the entire length of small hairpin .\n\nThe unique hairpin bypass of pol \\u03b2 resulted in large and small deletions in coordination with FEN1 alternate flap cleavage .\n\nOur results provide new insight into the role of BER in modulating genome stability that is associated with human diseases .", "output": "Genomic instability and mutation" }, { "input": "PURPOSE : Prostate cancer ( PCa ) development is often associated with deletion or silencing of tumor suppressor PTEN , a negative regulator of the PI3K-Akt pathway , leading to resistance to various therapies .\n\nTherefore , the PI3K/Akt pathway plays a central role in various cellular processes leading to malignant phenotype , and , consequently is an attractive target .\n\nHowever , the efficacy of AKT inhibitors is limited by resistance mechanisms that result in minimal tumor cell death .\n\nMethods:Effects of Akt inhibitor AZD5363 were investigated on cell proliferation , apoptosis , Akt pathway and survival mechanisms .\n\nWe then examined the impacts of inhibition of autophagy combined with AZD5363 on cell proliferation and apoptosis .\n\nFurthermore , the anti-cancer activity of combination treatment of the lysosomotropic inhibitor of autophagy ( chloroquine ) with Akt inhibitor AZD5363 was evaluated in PC-3 prostate cancer xenografts .\n\nRESULTS : we show that AZD5363 affected Akt downstream pathway by reducing p-mTOR , p-P70S6K and p-S6K .\n\nWhile AZD5363 monotherapy induced G2 growth arrest and autophagy , it failed to induce significant apoptosis in PC-3 and DU145 PCa cells .\n\nBlocking autophagy using pharmacological inhibitors or genetic inhibitors enhanced cell death induced by AZD5363 in these PCa cells .\n\nImportantly , the combination of AZD5363 with chloroquine significantly reduced tumor volume by 84.9% compared with the control group and by 77.5% compared with either drug alone in PC3 xenografts .\n\nCONCLUSIONS : These data demonstrate that Akt inhibitor AZD5363 , synergizes with the lysosomotropic inhibitor of autophagy , chloroquine , to induce apoptosis and delay tumor progression in PCa models that are resistant to monotherapy AZD5363 , providing a new therapeutic approach potentially translatable to patients .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "BACKGROUND : Although quiescence ( reversible cell cycle arrest ) is a key part in the life history and fate of many mammalian cell types , the mechanisms of gene regulation in quiescent cells are poorly understood .\n\nWe sought to clarify the role of microRNAs as regulators of the cellular functions of quiescent human fibroblasts .\n\nRESULTS : Using microarrays , we discovered that the expression of the majority of profiled microRNAs differed between proliferating and quiescent fibroblasts .\n\nFibroblasts induced into quiescence by contact inhibition or serum starvation had similar microRNA profiles , indicating common changes induced by distinct quiescence signals .\n\nBy analyzing the gene expression patterns of microRNA target genes with quiescence , we discovered a strong regulatory function for miR-29 , which is downregulated with quiescence .\n\nUsing microarrays and immunoblotting , we confirmed that miR-29 targets genes encoding collagen and other extracellular matrix proteins and that those target genes are induced in quiescence .\n\nIn addition , overexpression of miR-29 resulted in more rapid cell cycle re-entry from quiescence .\n\nWe also found that let-7 and miR-125 were upregulated in quiescent cells .\n\nOverexpression of either one alone resulted in slower cell cycle re-entry from quiescence , while the combination of both together slowed cell cycle re-entry even further .\n\nCONCLUSIONS : microRNAs regulate key aspects of fibroblast quiescence including the proliferative state of the cells as well as their gene expression profiles , in particular , the induction of extracellular matrix proteins in quiescent fibroblasts .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "Our study was to elucidate the mechanisms whereby BMS-345541 ( BMS , a specific I\\u03baB kinase \\u03b2 inhibitor ) inhibits the repair of DNA double-strand breaks ( DSBs ) and evaluate whether BMS can sensitize MCF-7 breast cancer cells ( MCF-7 cells ) to ionizing radiation ( IR ) in an apoptosis-independent manner .\n\nIn this study , MCF-7 cells were exposed to IR in vitro and in vivo with or without pretreatment of BMS .\n\nThe effects of BMS on the repair of IR-induced DSBs by homologous recombination ( HR ) and non-homologous end-joining ( NHEJ ) were analyzed by the DR-GFP and EJ5-GFP reporter assays and IR-induced \\u03b3-H2AX , 53BP1 , Brca1 and Rad51 foci assays .\n\nThe mechanisms by which BMS inhibits HR were examined by microarray analysis and quantitative reverse transcription PCR .\n\nThe effects of BMS on the sensitivity of MCF-7 cells to IR were determined by MTT and clonogenic assays in vitro and tumor growth inhibition in vivo in a xenograft mouse model .\n\nThe results showed that BMS selectively inhibited HR repair of DSBs in MCF-7 cells , most likely by down-regulation of several genes that participate in HR .\n\nThis resulted in a significant increase in the DNA damage response that sensitizes MCF-7 cells to IR-induced cell death in an apoptosis-independent manner .\n\nFurthermore , BMS treatment sensitized MCF-7 xenograft tumors to radiation therapy in vivo in an association with a significant delay in the repair of IR-induced DSBs .\n\nThese data suggest that BMS is a novel HR inhibitor that has the potential to be used as a radiosensitizer to increase the responsiveness of cancer to radiotherapy .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "High-grade prostate cancers express high levels of matrix metalloproteinases ( MMPs ) , major enzymes involved in tumor invasion and metastasis .\n\nHowever , the tumor cell lines commonly employed for prostate cancer research express only small amounts of MMPs when cultivated as monolayer cultures , in common culture media .\n\nThe present study was conducted to ascertain whether culture conditions that include fibronectin can alter MMP2 and MMP9 expression by the human prostatic epithelial cell lines RWPE-1 , LNCaP and PC-3 .\n\nThese cells were individually seeded at 2\u00d710(4) cells/cm(2) , cultivated until they reached 80% confluence , and then exposed for 4h to fibronectin , after which the conditioned medium was analyzed by gelatin zymography .\n\nUntreated cells were given common medium .\n\nOnly RWPE-1 cells express detectable amounts of MMP9 when cultivated in common medium , whereas the addition of fibronectin induced high expression levels of pro and active forms of MMP2 in all tested cell lines .\n\nOur findings demonstrate that normal and tumor prostate cell lines express MMP2 activity when in contact with extracellular matrix components or blood plasma proteins such as fibronectin .\n\nFuture studies of transcriptomes and proteomes in prostate cancer research using these cell lines should not neglect these important conclusions .", "output": "Activating invasion and metastasis" }, { "input": "p53 plays an important role in regulating a wide variety of cellular processes , such as cell cycle arrest and/or apoptosis .\n\nDysfunction of p53 is frequently associated with several pathologies , such as cancer and neurodegenerative diseases .\n\nIn recent years substantial progress has been made in developing novel p53-activating molecules .\n\nImportantly , modulation of p53 interaction with its main inhibitor , Mdm2 , has been highlighted as a promising therapy target .\n\nIn this regard , bimolecular fluorescence complementation ( BiFC ) analysis , by providing direct visualization of protein interactions in living cells , offers a straightforward method to identify potential modulators of protein interactions .\n\nIn this study , we developed a simple and robust Venus-based BiFC system to screen for modulators of p53-p53 and p53-Mdm2 interactions in live mammalian cells .\n\nWe used nutlin-3 , a well-known disruptor of p53-Mdm2 interaction , to validate the specificity of the assay .\n\nThe reduction of BiFC signal mediated by nutlin-3 was correlated with an increase in Puma transactivation , PARP cleavage , and cell death .\n\nFinally , this novel BiFC approach was exploited to identify potential modulators of p53-Mdm2 complex formation among a commercially available chemical library of 33 protein phosphatase inhibitors .\n\nOur results constitute \" proof-of-concept \" that this model has strong potential as an alternative to traditional target-based drug discovery strategies .\n\nIdentification of new modulators of p53-p53 and p53-Mdm2 interactions will be useful to achieve synergistic drug efficacy with currently used anti-tumor therapies .", "output": "Resisting cell death" }, { "input": "Breast cancer constitutes a major health problem for women worldwide .\n\nHowever , its incidence varies between populations and geographical locations .\n\nThese variations could be diet-related , since there are several carcinogenic compounds in the modern diet , while natural products contain various anti-cancer elements .\n\nSeveral lines of evidence indicate that , in addition to their clear preventive effect , these compounds could also be used as therapeutic agents .\n\nIn the present report we have shown that oleuropein , a pharmacologically safe natural product of olive leaf , has potent anti-breast cancer properties .\n\nIndeed , oleuropein exhibits specific cytotoxicity against breast cancer cells , with higher effect on the basal-like MDA-MB-231 cells than on the luminal MCF-7 cells .\n\nThis effect is mediated through the induction of apoptosis via the mitochondrial pathway .\n\nMoreover , oleuropein inhibits cell proliferation by delaying the cell cycle at S phase and up-regulated the cyclin-dependent inhibitor p21 .\n\nFurthermore , oleuropein inhibited the anti-apoptosis and pro-proliferation protein NF-\u03baB and its main oncogenic target cyclin D1 .\n\nThis inhibition could explain the great effect of oleuropein on cell proliferation and cell death of breast cancer cells .\n\nTherefore , oleuropein warrants further investigations to prove its utility in preventing/treating breast cancer , especially the less-responsive basal-like type .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "PURPOSE : Acid ceramidase ( AC ) occupies an important place in the control of cancer cell proliferation .\n\nWe tested the influence of AC inhibition on the effects of PSC 833 , a P-glycoprotein antagonist with potent ceramide-generating capacity , to determine whether AC could be a therapeutic target in pancreatic cancer .\n\nMETHODS : Ceramide metabolism was followed using ( 3)H-palmitate , and molecular species were determined by mass spectroscopy .\n\nApoptosis was measured by DNA fragmentation , autophagy by acridine orange staining , and cell cycle was assessed by flow cytometry and RB phosphorylation .\n\nAC was measured in intact cells using fluorescent substrate .\n\nRESULTS : Exposure of human PANC-1 or MIA-PaCa-2 cells to PSC 833 promoted increases in de novo ( dihydro)ceramides , ( dihydro)glucosylceramides , and ( dihydro)sphingomyelins , demarking ceramide generation and robust metabolism .\n\nDespite the multifold increases in ( dihydro)ceramide levels , cells were refractory to PSC 833 .\n\nHowever , PSC 833 produced a dose-dependent decrease in DNA synthesis and dose- and time-dependent decreases in RB phosphorylation , consistent with cell cycle arrest as demonstrated at G1 .\n\nCytostatic effects of PSC 833 were converted to cytotoxic end-point by acid ceramidase inhibition .\n\nCytotoxicity was accompanied by formation of acridine orange-stained acidic vesicles and an increase in LC3 expression , indicative of autophagic response .\n\nCell death was not reversed by preexposure to myriocin , which blocks PSC 833-induced ceramide generation .\n\nCONCLUSION : Although the role of ceramide in end-point cytotoxicity is unclear , our results suggest that acid ceramidase is a viable target in pancreatic cancer .\n\nWe propose that AC inhibition will be effective in concert with other anticancer therapies .", "output": "Sustaining proliferative signaling, Resisting cell death" }, { "input": "Epirubicin was developed as a semi-synthetic anthracycline derivative to circumvent the cardiotoxic limitations associated with the use of doxorubicin in the clinic .\n\nAnthracycline compounds have been demonstrated to form covalent drug-DNA adducts utilising endogenous and exogenous sources of formaldehyde ; however , previous investigations of the formation of epirubicin-DNA adducts provide conflicting evidence for adduct formation .\n\nThis work provides evidence that epirubicin acts to form drug-DNA adducts at physiologically relevant concentrations and demonstrates that the rate of formation of epirubicin-DNA adducts is slower than that observed for other anthracycline compounds , explaining why they are only detectable under defined experimental conditions .\n\nFormation of covalent epirubicin-DNA adducts improves the apoptotic profile of epirubicin and provides opportunities to overcome drug resistance and cardiotoxic limitations .", "output": "Genomic instability and mutation, Resisting cell death" }, { "input": "The mechanisms by which tumour cells metastasize and the role that cell polarity proteins play in this process are not well understood .\n\nWe report that partitioning defective protein 3 ( Par3 ) is dysregulated in metastasis in human breast cancer , and is associated with a higher tumour grade and ErbB2-positive status .\n\nDownregulation of Par3 cooperated with ErbB2 to induce cell invasion and metastasis in vivo .\n\nInterestingly , the metastatic behaviour was not associated with an overt mesenchymal phenotype .\n\nHowever , loss of Par3 inhibited E-cadherin junction stability , disrupted membrane and actin dynamics at cell-cell junctions and decreased cell-cell cohesion in a manner dependent on the Tiam1/Rac-GTP pathway .\n\nInhibition of this pathway restored E-cadherin junction stability and blocked invasive behaviour of cells lacking Par3 , suggesting that loss of Par3 promotes metastatic behaviour of ErbB2-induced tumour epithelial cells by decreasing cell-cell cohesion .", "output": "Activating invasion and metastasis" }, { "input": "BACKGROUND : CD81 is a transmembrane protein that serves as a putative receptor for hepatitis C virus .\n\nIn addition , CD81 has been suggested to be involved in a broad range of other cellular functions .\n\nIts putative implication in tumorigenesis has so far , however , remained largely unexplored .\n\nTo assess the candidacy of CD81 as a tumor suppressor in gastric cancer development , we investigated its expression and function in a series of primary gastric tumors and gastric tumor-derived cell lines .\n\nMETHODS : The expression and concomitant methylation status of the CD81 gene and its effect on tumor development and cellular signaling were evaluated .\n\nRESULTS : CD81 mRNA levels were found to be low in 16 of 40 ( 40% ) primary tumors and 9 of 14 ( 64.2% ) cell lines , and these low expression levels were found to correlate with the stage and grade of the tumors .\n\nGenomic alterations of CD81 were not encountered , whereas its expression could be re-activated in low expressing cells upon 5-aza-dC treatment .\n\nBisulfite DNA sequencing analysis of 10 CpG sites within the 5 ' proximal region of the CD81 gene promoter revealed that the observed transcriptional silencing was tightly associated with aberrant hypermethylation .\n\nSubsequent restoration of CD81 expression induced a G(1) cell cycle arrest and apoptosis , whereas siRNA-mediated CD81 down-regulation promoted cell proliferation and attenuated cellular responses to various apoptotic stress stimuli .\n\nAlso the colony-forming ability of the tumor cells could be inhibited and enhanced through CD81 up- and down-regulation , respectively .\n\nCD81 was found to inhibit p38 ( but not ERK , JNK and AKT ) phosphorylation and its growth suppressive effect could be abolished through p38 up- and down-regulation .\n\nCONCLUSION : From our data we conclude that epigenetic inactivation of CD81 is a common feature of gastric tumors and that this inactivation may render growth and survival advantages to the tumor cells , at least partially through p38 signaling .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Pterostilbene , a polyphenolic compound present in grapes and other fruits , has been demonstrated to inhibit growth and induce apoptosis and autophagy in some cancer cell types .\n\nWe found that pterostilbene at the IC90 concentration of 44 \ufffdM inhibited proliferation and induced apoptosis in MOLT4 human leukemia cells .\n\nTreatment with pterostilbene resulted in a transient accumulation of cells in the G0/G1-cell cycle phase followed by the S-phase arrest .\n\nPterostilbene-induced apoptotic death of MOLT4 cells was mediated by caspase-3 activation and was accompanied by the disruption of mitochondrial membrane potential , phosphatidylserine externalization and internucleosomal DNA fragmentation .\n\nOur results suggest that pterostilbene could serve as a potential additional chemotherapeutic agent for the treatment of leukemia .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Context:The ubiquitin-proteasome system and macroautophagy are two major pathways for intracellular protein degradation .\n\nEmerging lines of evidence have shown that blockade of ubiquitin-proteasome system by proteasome inhibitors activates macroautophagy.Objective:The purpose of this study was to determine the involvement of autophagy essential gene Beclin 1 in cytotoxicity of thyroid cancer cells mediated by proteasome inhibitors.Design:Autophagy was measured by acidic-trophic dye staining and EGF-LC3 distribution using fluorescence microscopy , as well as LC3-II transition using Western blot .\n\nTo ascertain the effect of Beclin 1 , cells were transfected with Beclin 1 plasmid or shRNA against Beclin 1 .\n\nCell viability and apoptotic cells were measured using MTT assay and flow cytometry , respectively.Results:Proteasome inhibitors decreased Beclin 1 expression .\n\nIn addition , treatment with PI3K inhibitors 3-MA or wortmannin , as well as knockdown of Beclin 1 expression , was unable to affect autophagic responses mediated by proteasome inhibitors .\n\nOverexpression of Beclin 1 enhanced proteasome inhibitor-mediated cytotoxicity of thyroid cancer cells via suppression of survivin.Conclusions:Proteasome inhibitors cause Beclin 1-independent macroautophagic responses of thyroid cancer cells in a Beclin 1-independent manner .\n\nBeclin 1 possesses autophagy-independent antitumoral effects upon exposure of thyroid cancer cells to proteasome inhibitors .", "output": "Resisting cell death" }, { "input": "The Wnt/\u03b2-catenin pathway is constitutively activated in over 90% of human colorectal cancer .\n\nActivated \u03b2-catenin stimulates cell proliferation and survival , however , its anti-apoptotic mechanisms are not fully understood .\n\nWe show here that activated \u03b2-catenin is required to suppress caspase-8 activation but only in colon cancer cells that are resistant to tumor necrosis factor-a ( TNF ) induced apoptosis .\n\nWe found that lysosomal delivery of internalized TNF occurred at a faster pace in apoptosis-resistant than in apoptosis-sensitive colon cancer cells .\n\nRetardation of endosomal trafficking through V-ATPase inhibition enhanced caspase-8 activation in the apoptosis-resistant but not sensitive cells .\n\nInterestingly , knockdown of \u03b2-catenin also prolonged TNF association with the early endosome and enhanced caspase-8 activation in the apoptosis-resistant but not sensitive colon cancer cells .\n\nIn a mouse model of inflammation-associated colon tumors , we found nuclear expression of \u03b2-catenin , resistance to TNF-induced apoptosis , and reactivation of apoptosis in vivo by cotreatment of TNF with a V-ATPase inhibitor .\n\nTogether , these results suggest that activated \u03b2-catenin can facilitate endosomal trafficking of internalized TNF to suppress caspase-8 activation in colon cancer cells .", "output": "Tumor promoting inflammation, Resisting cell death" }, { "input": "Kaposi sarcoma is the most common neoplasm caused by Kaposi sarcoma-associated herpesvirus ( KSHV ) .\n\nIt is prevalent among the elderly in the Mediterranean , inhabitants of sub-Saharan Africa , and immunocompromised individuals such as organ transplant recipients and AIDS patients .\n\nCurrent treatments for Kaposi sarcoma can inhibit tumor growth but are not able to eliminate KSHV from the host .\n\nWhen the host's immune system weakens , KSHV begins to replicate again , and active tumor growth ensues .\n\nNew therapeutic approaches are needed .\n\nCannabidiol ( CBD ) , a plant-derived cannabinoid , exhibits promising antitumor effects without inducing psychoactive side effects .\n\nCBD is emerging as a novel therapeutic for various disorders , including cancer .\n\nIn this study , we investigated the effects of CBD both on the infection of endothelial cells ( ECs ) by KSHV and on the growth and apoptosis of KSHV-infected ECs , an in vitro model for the transformation of normal endothelium to Kaposi sarcoma .\n\nWhile CBD did not affect the efficiency with which KSHV infected ECs , it reduced proliferation and induced apoptosis in those infected by the virus .\n\nCBD inhibited the expression of KSHV viral G protein-coupled receptor ( vGPCR ) , its agonist , the chemokine growth-regulated protein \u03b1 ( GRO-\u03b1 ) , vascular endothelial growth factor receptor 3 ( VEGFR-3 ) , and the VEGFR-3 ligand , vascular endothelial growth factor C ( VEGF-C ) .\n\nThis suggests a potential mechanism by which CBD exerts its effects on KSHV-infected endothelium and supports the further examination of CBD as a novel targeted agent for the treatment of Kaposi sarcoma .", "output": "Inducing angiogenesis, Resisting cell death" }, { "input": "In spite of the fact that they occur at high rates , the clinical responses of BRAF(V600) mutant metastatic melanoma to BRAF inhibitors are usually short-lasting , with most cases progressing within less than 8 mo .\n\nImmunomodulatory strategies initiated after progression have recently been reported to be poorly efficient .\n\nBy characterizing the immunological interactions between T cells and cancer cells in clinical material as well as the influence of the FDA-approved BRAF inhibitor vemurafenib on the immune system , we aimed at unraveling new strategies to expand the efficacy of adoptive T-cell transfer , which represents one of the most promising approaches currently in clinical development for the treatment of metastatic melanoma .\n\nHere we show that blocking the BRAF-MAPK pathway in BRAF signaling-addicted melanoma cells significantly increases the ability of T cells contained in clinical grade tumor-infiltrating lymphocytes to recognize autologous BRAF(V600) mutant melanoma cell lines in vitro .\n\nAntitumor reactivity was improved regardless of the class of antigen recognized by tumor-specific CD8(+) T cells .\n\nMicroarray data suggests that improved tumor recognition is associated with modified expression of MHC Class I-associated proteins as well as of heat-shock proteins .\n\nIn conclusion , our preclinical data suggest that an appropriately timed sequential treatment of BRAF(V600) mutant melanoma with vemurafenib and adoptive T-cell transfer might result in synergistic antineoplastic effects owing to an increased immunogenicity of cancer cells .", "output": "Avoiding immune destruction" }, { "input": "Inflammation is a major risk factor for carcinogenesis in patients affected by chronic colitis , yet the molecular mechanisms underlying the progression from chronic inflammation to cancer are not completely understood .\n\nActivation of the Toll-like receptor 4 ( TLR4)-NF\u03baB signaling axis is associated with inflammation .\n\nThus , we hypothesized that inhibition of TLR4-NF\u03baB signaling might help in limiting inflammatory responses and inflammation-induced oncogenesis .\n\nIn this work , we studied the effects of a TLR4-interacting surfactant protein A-derived ( SPA4 ) peptide on lipopolysaccharide ( LPS)-induced TLR4-NF\u03baB signaling and cancer progression .\n\nWe first characterized this peptide for its ability to bind the TLR4 ligand-LPS and for physico-chemical characteristics .\n\nInflammation was induced by challenging the colon cancer SW480 cells with Escherichia coli LPS .\n\nCells were then treated with varying amounts of the SPA4 peptide .\n\nChanges in the expression of TLR4 , interleukin ( IL)-1\u03b2 and IL-6 , in intracellular NF\u03baB-related signal transducers ( IKB\u03b1 , p65 , phosphorylated IKB\u03b1 , phosphorylated p65 , RelB , COX-2 ) as well as in the transcriptional activity of NF\u03baB were studied by immunocytochemistry , immunoblotting and NF\u03baB reporter assay , respectively .\n\nSimultaneously , the effects on LPS-induced cell migration and invasion were determined .\n\nWe found that the SPA4 peptide does not bind to LPS .\n\nRather , its binding to TLR4 inhibits the LPS-induced phosphorylation of p65 , production of IL-1\u03b2 and IL-6 , activity of NF\u03baB , migration and invasion of SW480 cells .\n\nIn conclusion , our results suggest that the inhibition of TLR4-NF\u03baB signaling by a TLR4-binding peptide may help for the treatment of chronic inflammation and prevention of inflammation-induced cancer in patients with colitis .", "output": "Tumor promoting inflammation, Activating invasion and metastasis" }, { "input": "Members of the Orai family are highly selective calcium ion channels that play an important role in store-operated calcium entry .\n\nAmong the three known Orai isoforms , Orai3 has gained increased attention , notably for its emerging role in cancer .\n\nWe recently demonstrated that Orai3 channels are over-expressed in breast cancer ( BC ) biopsies , and involved specifically in proliferation , cell cycle progression and survival of MCF-7 BC cells .\n\nHere , we investigate the downstream signaling mechanisms affected by Orai3 silencing , leading to the subsequent functional impact specifically seen in MCF-7 cancer cells .\n\nWe report a correlation between Orai3 and c-myc expression in tumor tissues and in the MCF-7 cancer cell line by demonstrating that Orai3 down-regulation reduces both expression and activity of the proto-oncogene c-myc .\n\nThis is likely mediated through the MAP Kinase pathway , as we observed decreased pERK1/2 levels and cell-cycle arrest in G1 phase after Orai3 silencing .\n\nOur results provide strong evidence that the c-myc proto-oncogene is influenced by the store-operated calcium entry channel Orai3 through the MAP kinase pathway .\n\nThis connection provides new clues in the downstream mechanism linking Orai3 channels and proliferation , cell cycle progression and survival of MCF-7 BC cells .", "output": "Sustaining proliferative signaling, Evading growth suppressors" }, { "input": "A predictive mathematical model of the transition from the G2 phase in the cell cycle to mitosis ( M ) was constructed from the known interactions of the proteins that are thought to play significant roles in the G2 to M transition as well as the DNA damage- induced G2 checkpoint .\n\nThe model simulates the accumulation of active cyclin B1/Cdk1 ( MPF ) complexes in the nucleus to activate mitosis , the inhibition of this process by DNA damage , and transport of component proteins between cytoplasm and nucleus .\n\nInteractions in the model are based on activities of individual phospho-epitopes and binding sites of proteins involved in G2/M .\n\nBecause tracking phosphoforms leads to combinatorial explosion , we employ a rule-based approach using the BioNetGen software .\n\nThe model was used to determine the effects of depletion or over-expression of selected proteins involved in the regulation of the G2 to M transition in the presence and absence of DNA damage .\n\nDepletion of Plk1 delayed mitotic entry and recovery from the DNA damage-induced G2 arrest and over-expression of MPF attenuated the DNA damage-induced G2 delay .\n\nThe model recapitulates the G2 delay observed in the biological response to varying levels of a DNA damage signal .\n\nThe model produced the novel prediction that depletion of pkMyt1 results in an abnormal biological state in which G2 cells with DNA damage accumulate inactive nuclear MPF .\n\nSuch a detailed model may prove useful for predicting DNA damage G2 checkpoint function in cancer and , therefore , sensitivity to cancer therapy .", "output": "Genomic instability and mutation, Evading growth suppressors" }, { "input": "\u03b3-Tocotrienol and sesamin are phytochemicals that display potent anticancer activity .\n\nSince sesamin inhibits the metabolic degradation of tocotrienols , studies were conducted to determine if combined treatment with sesamin potentiates the antiproliferative effects of \u03b3-tocotrienol on neoplastic mouse ( +SA ) and human ( MCF-7 and MDA-MB-231 ) mammary cancer cells .\n\nResults showed that treatment with \u03b3-tocotrienol or sesamin alone induced a significant dose-responsive growth inhibition , whereas combination treatment with these agents synergistically inhibited the growth of +SA , MCF-7 and MDA-MB-231 mammary cancer cells , while similar treatment doses were found to have little or no effect on normal ( mouse CL-S1 and human MCF-10A ) mammary epithelial cell growth or viability .\n\nHowever , sesamin synergistic enhancement of \u03b3-tocotrienol-induced anticancer effects was not found to be mediated from a reduction in \u03b3-tocotrienol metabolism .\n\nRather , combined treatment with subeffective doses of \u03b3-tocotrienol and sesamin was found to induce G1 cell cycle arrest , and a corresponding decrease in cyclin D1 , CDK2 , CDK4 , CDK6 , phospho-Rb , and E2F1 levels , and increase in p27 and p16 levels .\n\nAdditional studies showed that the antiproliferative effect of combination treatment did not initiate apoptosis or result in a decrease in mammary cancer cell viability .\n\nTaken together , these findings indicate that the synergistic antiproliferative action of combined \u03b3-tocotrienol and sesamin treatment in mouse and human mammary cancer cells is cytostatic , not cytotoxic , and results from G1 cell cycle arrest .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Bone marrow transplantation ( BMT ) , used to treat severe hematological malignancies , often leads to potentially fatal acute graft-versus-host disease ( GVHD ) , despite attempts for better matching and/or use of immune suppressive agents .\n\nWe reported that embryo-derived PreImplantation-Factor plays a determining role in developing maternal/host tolerance towards the semi/allogenic embryo and regulating systemic immune response .\n\nSynthetic PIF ( PIF ) treatment is effective preventing immune attack in non-pregnant models of autoimmunity .\n\nHerein , we test PIF ability to prevent acute GVHD development in semi or totally allogenic murine models .\n\nWe examine PIF's regulatory effect in vivo and in vitro , to control deleterious GVHD while maintaining its ability to preserve beneficial graft vs. leukemia ( GVL effect ) .\n\nBone marrow and spleen cells from C57BL/6 donors were transplanted to semi-allogenic ( C57BL/6\ufffdBALB/c ) F1 or allogenic ( BALB/c ) recipients , and then treated with PIF1mg/kg/day/2weeks .\n\nShort-term PIF administration reduced acute GVHD in both models and increased survival for up to four months after semi-(or totally ) allogenic BMT .\n\nThe obtained effect was coupled with lessened skin ( semi-allogenic ) and decreased liver inflammation ( both models ) as well as reduced colon ulceration ( allogenic ) .\n\nBoth GVHD associated cytokines and chemokines gene expression were decreased in the liver .\n\nPIF further lowered circulating interleukin-17 , but not interferon-\u03b3 levels .\n\nPIF treatment was demonstrated in vivo and in vitro to lead to decreased iNOS expression and in LPS-activated macrophages to lower nitric oxide secretion .\n\nSignificantly , PIF did not impair the beneficial GVL effect in the B-cell leukemia model .\n\nPIF primarily acts by inducing regulatory phenotype on monocytes/APC , which controls T-cell proliferation .\n\nOverall our data demonstrates that PIF protects against semi/allogenic GVHD long-term by reducing both target organ and systemic inflammation and by lowering oxidative stress , all while preserving the beneficial GVL effect .", "output": "Tumor promoting inflammation" }, { "input": "There are many growth factors influencing the expansion of multiple myeloma ( MM ) .\n\nAngiogenesis is a process that may enhance MM growth , in various manners .\n\nAmong them , insulin-like growth factor-1 ( IGF-1 ) is a major factor , acting in many levels .\n\nThe aim of the study was to measure serum levels of IGF-1 in newly diagnosed MM patients and to correlate them with clinical stage of the disease and with markers of angiogenesis , such as vascular endothelial growth factor ( VEGF ) , hepatocyte growth factor ( HGF ) , and interleukin-6 and 15 ( IL-6 and IL-15 ) .\n\nSerum levels of the above factors were measured , by ELISA , in 57 newly diagnosed MM patients and in 20 healthy controls .\n\nThere was no difference in serum levels of IGF-1 in MM patients and in controls , contrary to angiogenic factors , which were higher in MM patients ( p < 0.001 ) .\n\nSimilarly , IGF-1 did not correlate with clinical stage of the disease nor the other angiogenic factors , which also correlated with each other ( p < 0.001 ) .\n\nSerum IGF-1 concentrations are not influenced in MM patients .\n\nTherefore , although it is a proliferation cytokine , it cannot be used as marker of disease activity .", "output": "Inducing angiogenesis" }, { "input": "DNA damage signaling and repair take place in a chromatin context .\n\nConsequently , chromatin-modifying enzymes , including adenosine triphosphate-dependent chromatin remodeling enzymes , play an important role in the management of DNA double-strand breaks ( DSBs ) .\n\nHere , we show that the p400 ATPase is required for DNA repair by homologous recombination ( HR ) .\n\nIndeed , although p400 is not required for DNA damage signaling , DNA DSB repair is defective in the absence of p400 .\n\nWe demonstrate that p400 is important for HR-dependent processes , such as recruitment of Rad51 to DSB ( a key component of HR ) , homology-directed repair , and survival after DNA damage .\n\nStrikingly , p400 and Rad51 are present in the same complex and both favor chromatin remodeling around DSBs .\n\nAltogether , our data provide a direct molecular link between Rad51 and a chromatin remodeling enzyme involved in chromatin decompaction around DNA DSBs .", "output": "Genomic instability and mutation" }, { "input": "ABSTRACT : BACKGROUND : Survivin , a member of the family of inhibitor of apoptosis proteins , functions as a key regulator of mitosis and programmed cell death .\n\nYM155 , a novel molecular targeted agent , suppresses survivin , which is overexpressed in many tumor types .\n\nThe aim of this study was to determine the antitumor activity of YM155 in SK-NEP-1 cells .\n\nMETHODS : SK-NEP-1 cell growth in vitro and in vivo was assessed by MTT and nude mice experiments .\n\nAnnexin V/propidium iodide staining followed by flow cytometric analysis was used to detect apoptosis in cell culture .\n\nThen gene expression profile of tumor cells treated with YM155 was analyzed with real-time PCR arrays .\n\nWe then analyzed the expression data with MEV ( Multi Experiment View ) cluster software .\n\nDatasets representing genes with altered expression profile derived from cluster analyses were imported into the Ingenuity Pathway Analysis tool .\n\nRESULTS : YM155 treatment resulted in inhibition of cell proliferation of SK-NEP-1cells in a dose-dependent manner .\n\nAnnexin V assay , cell cycle , and activation of caspase-3 demonstrates that YM155 induced apoptosis in SK-NEP-1 cells .\n\nYM155 significantly inhibited growth of SK-NEP-1 xenografts ( YM155 5 mg/kg : 1.45 +/- 0.77 cm3 ; YM155 10 mg/kg : 0.95 +/- 0.55 cm3 ) compared to DMSO group ( DMSO : 3.70 +/- 2.4 cm3 ) or PBS group cells ( PBS : 3.78 +/- 2.20 cm3 , ANOVA P < 0.01 ) .\n\nYM155 treatment decreased weight of tumors ( YM155 5 mg/kg : 1.05 +/- 0.24 g ; YM155 10 mg/kg : 0.72 +/- 0.17 g ) compared to DMSO group ( DMSO : 2.06 +/- 0.38 g ) or PBS group cells ( PBS : 2.36 +/- 0.43 g , ANOVA P < 0.01 ) .\n\nReal-time PCR array analysis showed between Test group and control group there are 32 genes significantly up-regulated and 54 genes were significantly down-regulated after YM155 treatment .\n\nIngenuity pathway analysis ( IPA ) showed cell death was the highest rated network with 65 focus molecules and the significance score of 44 .\n\nThe IPA analysis also groups the differentially expressed genes into biological mechanisms that are related to cell death , cellular function maintenance , cell morphology , carbohydrate metabolism and cellular growth and proliferation .\n\nDeath receptor signaling ( 3.87E-19 ) , TNFR1 signaling , induction of apoptosis by HIV1 , apoptosis signaling and molecular mechanisms of cancer came out to be the top four most significant pathways .\n\nIPA analysis also showed top molecules up-regulated were BBC3 , BIRC3 , BIRC8 , BNIP1 , CASP7 , CASP9 , CD5 , CDKN1A , CEBPG and COL4A3 , top molecules down-regulated were ZNF443 , UTP11L , TP73 , TNFSF10 , TNFRSF1B , TNFRSF25 , TIAF1 , STK17A , SST and SPP1 , upstream regulator were NR3C1 , TP53 , dexamethasone , TNF and Akt .\n\nCONCLUSIONS : The present study demonstrates that YM155 treatment resulted in apoptosis and inhibition of cell proliferation of SK-NEP-1cells .\n\nYM155 had significant role and little side effect in the treatment of SK-NEP-1 xenograft tumors .\n\nReal-time PCR array analysis firstly showed expression profile of genes dyes-regulated after YM155 treatment .\n\nIPA analysis also represents new molecule mechanism of YM155 treatment , such as NR3C1 and dexamethasone may be new target of YM155 .\n\nAnd our results may provide new clues of molecular mechanism of apoptosis induced by YM155 .", "output": "Resisting cell death" }, { "input": "We evaluated the clinical significance of indoleamine 2 , 3-dioxygenase ( IDO ) expression in breast cancer patients with bone metastasis .\n\nIDO activity can be measured by the tryptophan(Trp)/kynurenine(Kyn) ratio .\n\nTrp and Kyn levels were measured by high-performance liquid chromatography ( HPLC ) .\n\nThe serum IDO levels of postoperative breast cancer patients with a high number of bone metastases were lower than those of patients with a single metastasis lesion .\n\nIn addition , IDO activity increased in the cases in which the number of metastatic lesions to the bone increased .\n\nThese results suggest that the expression of IDO in breast cancer patients with bone metastasis may play a critical role in immunosuppression in these patients .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "We report a case of recurrent gastric cancer successfully treated by a combination of CPT-11 and CDDP as the third- line chemotherapy .\n\nA 78-year-old man with advanced gastric cancer underwent a curative distal gastrectomy .\n\nThree years later , the tumor marker level began to rise and computed tomography ( CT ) revealed lymph node metastasis invading the pancreas resulting in pancreatic duct dilatation .\n\nS-1 treatment was initiated but was discontinued because of a systemic exanthem .\n\nPaclitaxel was administrated as secondary chemotherapy .\n\nBut , after 2 courses , a further increase in the tumor marker level and portal vein invasion were observed .\n\nCombination therapy of CPT-11 and CDDP was administered as the third-line chemotherapy .\n\nAfter 3 courses , the tumor marker level normalized , tumor size decreased , and the invasion was eliminated .\n\nThird-line chemotherapy against recurrent gastric cancer should be considered if patient performance status ( PS ) is maintained .", "output": "Activating invasion and metastasis" }, { "input": "Vaccination is , in theory , a safe and effective approach for controlling disseminated or metastatic cancer due to the specificity of the mammalian immune system , yet its success in the clinic has been hampered thus far by the problem of immune tolerance to tumor self-antigen .\n\nHere we describe a DNA vaccination strategy that is able to control cancer by overcoming immune tolerance to tumor self-antigen .\n\nWe engineered a DNA construct encoding a dimeric form of a secreted single chain trimer of major histocompatibility complex class I heavy chain , \u03b22-microglobulin , and peptide antigen linked to immunoglobulin G ( SCT-Ag/IgG ) .\n\nThe chimeric protein was able to bind to antigen-specific CD8+ T cells with nearly 100% efficiency and strongly induce their activation and proliferation .\n\nIn addition , the chimeric protein was able to coat professional antigen-presenting cells through the Fc receptor to activate antigen-specific CD8+ T cells .\n\nFurthermore , intradermal vaccination with DNA encoding SCT-Ag/IgG could generate significant numbers of cytotoxic effector T cells against tumor self-antigen and leads to successful therapeutic outcomes in a preclinical model of metastatic melanoma .\n\nOur data suggest that the DNA vaccine strategy described in the current study is able to break immune tolerance against endogenous antigen and result in potent therapeutic antitumor effects .\n\nSuch strategy may be used in other antigenic systems for the control of infections and/or cancers .", "output": "Avoiding immune destruction, Activating invasion and metastasis" }, { "input": "Abstract Different cyclooxygenase ( COX)-2 inhibitors were known to cause different cell cycle changes .\n\nWe investigated whether this different effect on cell cycle change was due to concentration-dependent effect .\n\nWe investigated the effects of celecoxib , a COX-2 selective inhibitor , on cell cycle regulation in irradiated cancer cells that express high or low levels of COX-2 .\n\nFour stably COX-2 knocked-down or overexpressed cell lines were treated with various concentrations of celecoxib with or without radiation .\n\nCelecoxib differentially modulated the cell cycle according to the concentrations applied .\n\nG(1) arrest was induced at lower concentrations , whereas G(2)/M arrest was induced at higher concentrations in each cell line tested .\n\nRadiation-induced G(2)/M arrest was enhanced at lower concentrations but reduced at higher concentrations .\n\nThe cutoff values to divide lower and higher concentrations were cell-type specific .\n\nCelecoxib treatment activated Cdc25C and inhibited p21 expression in both unirradiated and irradiated cells , regardless of COX-2 expression .\n\nApoptosis was induced in irradiated cells 48 hours after treatment with celecoxib dependent of COX-2 .\n\nThese results imply that celecoxib deactivates the G(2) checkpoint via both Cdc25C- and p21-dependent pathways in irradiated cells , which subsequently die by secondary apoptosis .\n\nCell cycle modulating effects in irradiated cells resulting from treatment with celecoxib may have clinical importance with regard to the potential application of celecoxib in cancer patients undergoing radiotherapy .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "Abstract Purpose : Cell death is one of the most important endpoints of radiosensitivity .\n\nThe tumor suppressor p53 participates not only in regulation of apoptosis , but also in autophagy mechanism .\n\nIn this study , H1299-P53 ( with wild-type p53 ) and H1299-175H ( with mutant 175H ) were used , and the effects of p53 on radiosensitivity were analyzed .\n\nMethods : Cell models with different p53 status were established by gene engineering , and cell viability was examined by colony formation assay , and cell counting kit-8 ( CCK-8 ) , 3-Methyladenine , and Z-VAD were used to block autophagy and apoptosis , respectively .\n\nWestern blot was used to detect protein expression ; monodansylcadaverine ( MDC ) staining was used to analyze autophagy rate ; DAPI/Propidium Iodide ( PI ) staining and flow cytometry were used to assess apoptosis and necrosis .\n\nResults : In parental H1299 , H1299-P53 , and H1299-175H cells , radiosensitivity exhibited different by colony formation and CCK-8 assay ( D0 : 1.764 Gy , 1.407 Gy and 1.695 Gy ; Dq : 2.977 Gy , 1.199 Gy and 2.312 Gy in turn ) .\n\nThe radiosensitization of p53 was associated with the increase of MDM2 and P21 expression .\n\nThe ionizing radiation ( IR)-induced apoptosis was significant in H1299-P53 compared with in H1299 and H199-175H ( p<0.05 ) by flow cytometry , and the expression of cleaved-caspase3 was increased in H1299-P53 cells .\n\nWhile the IR-induced autophagy was significant in H1299 cells ( p<0.01 ) and decreased in H1299-P53 and H1299-175H cells ( p<0.01 ) by MDC staining , the expression of MAPLC3II and Beclin-1 increased in H1299 , but not in H1299-p53 and H199-175H cells .\n\nThe IR-induced cell survival was significantly increased by Z-VAD-FMK and decreased by 3MA in H1299-P53 cells ; IR- induced autophagy was significantly increased by Z-VAD-FMK in H1299-P53 cells ( p<0.01 ) , but not changed in H1299 cells .\n\nConclusion : p53 could regulate radiosensitivity by inhibiting autophagy and activating apoptosis ; autophagy provides a prosurvival mechanism , and p53 potently abrogated the IR-induced autophagy , while mutant 175H shown no effect on radiosensitivity , suggesting that individual treatment strategies should be based on p53 status in patients .", "output": "Resisting cell death" }, { "input": "Sunitinib is an oral multitargeted receptor tyrosine kinase inhibitor with antiangiogenic and antitumor activity that mainly targets vascular endothelial growth factor receptors , and recently , it has been shown to be an active agent for the treatment of malignant pheochromocytomas .\n\nPreviously , we demonstrated that sunitinib directly inhibited mTORC1 signaling in rat pheochromocytoma PC12 cells .\n\nAlthough autophagy is a highly regulated cellular process , its relevance to cancer seems to be complicated .\n\nIt is of note that inhibition of mTORC1 is a prerequisite for autophagy induction .\n\nIndeed , direct mTORC1 inhibition initiates ULK1/2 autophosphorylation and subsequent Atg13 and FIP200 phosphorylation , inducing autophagy .\n\nHere , we demonstrated that sunitinib significantly increased the levels of LC3-II , concomitant with a decrease of p62 in PC12 cells .\n\nFollowing sunitinib treatment , immunofluorescent imaging revealed a marked increased punctate LC3-II distribution .\n\nFurthermore , Atg13 knockdown significantly reduced its protein level , which in turn abolished sunitinib-induced autophagy .\n\nMoreover , inhibition of autophagy by siRNAs targeting Atg13 or by pharmacological inhibition with ammonium chloride , enhanced both sunitinib-induced apoptosis and anti-proliferation .\n\nThus , sunitinib-induced autophagy is dependent on the suppression of mTORC1 signaling and the formation of ULK1/2-Atg13-FIP200 complexes .\n\nInhibition of autophagy may be a promising therapeutic option for improving the anti-tumor effect of sunitinib .", "output": "Resisting cell death" }, { "input": "ABSTRACT : BACKGROUND : The ubiquitin-proteasome system and macroautophagy ( hereafter referred to autophagy ) are two complementary pathways for protein degradation .\n\nEmerging evidence suggests that proteasome inhibition might be a promising approach for tumor therapy .\n\nAccumulating data suggest that autophagy is activated as a compensatory mechanism upon proteasome activity is impaired .\n\nMETHOD : Autophagy activation was measured using acridine orange staining and LC3 transition .\n\nCell viability and apoptosis were measured using MTT assay and flow cytometry , respectively .\n\nBeclin 1 expression vectors or shRNA against Beclin 1 ( shBeclin 1 ) were transfected to investigate the role of Beclin 1 in autophagy activation and cytotoxicity of ovarian cancer cells induced by proteasome inhibitors .\n\nRESULTS : Proteasome inhibitors suppressed proliferation and induced autophagy in ovarian cancer cells .\n\nNeither phosphoinositide 3-kinase ( PI3K ) inhibitors nor shRNA against Beclin 1 could abolish the formation of acidic vacuoles and the processing of LC3 induced by proteasome inhibitors .\n\nMoreover , Beclin 1 overexpression enhanced anti-proliferative effects of proteasome inhibitors in ovarian cancer cells .\n\nCONCLUSIONS : For the first time , the current study demonstrated that proteasome inhibitors induced PI3K and Beclin 1-independent autophagy in ovarian cancer cells .\n\nIn addition , this study revealed autophagy-independent tumor suppressive effects of Beclin 1 in ovarian cancer cells .", "output": "Resisting cell death" }, { "input": "Recently , our group reported the discovery of three new withanolides , physangulidines A-C , from Physalis angulata .\n\nIn this study , the biological effects of physangulidine A ( 1 ) , which was the most active and abundant of the three new constituents , are described .\n\nIt was found that 1 significantly reduces survival in clonogenic assays for two hormone-independent prostate cancer cell lines .\n\nFlow cytometry and confocal microscopy studies in DU145 human prostate cancer cells indicated that 1 induces cell cycle arrest in the G(2)/M phase and causes defective mitosis .\n\nIt was determined also that 1 produces programed cell death by apoptosis , as evidenced by biochemical markers and distinct changes in cell morphology .\n\nThese results imply that the antimitotic and proapoptotic effects of 1 may contribute significantly to the biological activities and potential medicinal properties of its plant of origin .", "output": "Sustaining proliferative signaling, Evading growth suppressors, Resisting cell death" }, { "input": "The evidence that androgen blockade-resistant prostate cancer , termed castration resistant , remains androgen receptor ( AR ) dependent is compelling .\n\nAR is re-activated through multiple mechanisms including expression of constitutively active splice variants that lack hormone binding domains ( HBDs ) .\n\nThis highlights need to develop therapies that target regions other than the HBD .\n\nBecause the p160 coactivators interact most strongly with the amino-terminus of AR , we examined the consequences of disrupting this interaction .\n\nWe identified two overlapping SRC-1 peptides that interact with AR , but not with progesterone receptor .\n\nThese peptides reduce AR and AR variant AR-V7 dependent induction of an AR responsive reporter .\n\nUsing mammalian two hybrid assays , we found that the peptides interrupt the AR/SRC-1 , AR/SRC-2 and AR N/C interactions , but not SRC-1/CARM-1 interactions .\n\nConsistent with the SRC-1 dependence of induced , but not repressed genes , in LNCaP cells , the peptides inhibited hormone dependent induction of endogenous target genes including PSA and TMPRSS2 , but did not block AR dependent repression of UGT2B17 or inhibit vitamin D receptor activity .\n\nSimultaneous detection of SRC-1 peptides and PSA by double immunofluorescence in transfected LNCaP cells clearly demonstrated a strong reduction in PSA levels in cells expressing the peptides .\n\nThe peptides also inhibited the AR dependent expression of PSA in castration resistant C4-2 cells .\n\nMoreover they inhibited androgen dependent proliferation of LNCaP cells and proliferation of C4-2 cells in androgen depleted medium without affecting AR negative PC-3 cells .\n\nThus , the p160 coactivator binding site is a novel potential therapeutic target to inhibit AR activity .", "output": "Sustaining proliferative signaling" }, { "input": "AIM : To explore the inhibitory effect of sulfated polysaccharide from Masson pine ( Pinus massoniana ) pollen ( SPPM60 ) on G(2)/M phase of human liver cancer HepG2 cells and its mechanism .\n\nMETHODS : The proliferation rate of HepG2 cells was evaluated by methyl thiazolyl tetrazolium ( MTT ) colorimetric assay .\n\nThe cycles of HepG2 cells were measured by flow cytometry when 200\u03bcg/ml concentration of SPPM60 was adopted , the expression of the genes related to cell cycle was detected by real-time PCR .\n\nRESULTS : SPPM60 inhibited the proliferation of HepG2 cells and the inhibition rate was elevated with increase of SPPM60 concentration .\n\nAfter treatment with 200\u03bcg/ml of SPPM60 , the percentage of S phase cells was decreased , but that of G(2)/M phase was significantly increased ( 72h vs control : 32.96\ufffd0.33% vs 18.59\ufffd0.04% , 3.44\ufffd0.05% vs 18.30\ufffd0.08% , P<0.01 ) .\n\nThe results of real-time PCR showed that SPPM60 could down-regulate the mRNA levels of CDK1 and CyclinB ( P<0.01 ) , and up-regulate the expression of p53 and p21 ( P<0.05 ) .\n\nCONCLUSION : SPPM60 causes arrest of HepG2 cells at G(2)/M phase , and the mechanism is related to the down-regulation of CDK1 and CyclinB and up-regulation of p53 and p21 expression .", "output": "Sustaining proliferative signaling, Evading growth suppressors" } ] }