22 research outputs found

    Cell cycle progression or translation control is not essential for vesicular stomatitis virus oncolysis of hepatocellular carcinoma.

    Get PDF
    The intrinsic oncolytic specificity of vesicular stomatitis virus (VSV) is currently being exploited to develop alternative therapeutic strategies for hepatocellular carcinoma (HCC). Identifying key regulators in diverse transduction pathways that define VSV oncolysis in cancer cells represents a fundamental prerequisite to engineering more effective oncolytic viral vectors and adjusting combination therapies. After having identified defects in the signalling cascade of type I interferon induction, responsible for attenuated antiviral responses in human HCC cell lines, we have now investigated the role of cell proliferation and translation initiation. Cell cycle progression and translation initiation factors eIF4E and eIF2Bepsilon have been recently identified as key regulators of VSV permissiveness in T-lymphocytes and immortalized mouse embryonic fibroblasts, respectively. Here, we show that in HCC, decrease of cell proliferation by cell cycle inhibitors or siRNA-mediated reduction of G(1) cyclin-dependent kinase activities (CDK4) or cyclin D1 protein expression, do not significantly alter viral growth. Additionally, we demonstrate that translation initiation factors eIF4E and eIF2Bepsilon are negligible in sustaining VSV replication in HCC. Taken together, these results indicate that cellular proliferation and the initiation phase of cellular protein synthesis are not essential for successful VSV oncolysis of HCC. Moreover, our observations indicate the importance of cell-type specificity for VSV oncolysis, an important aspect to be considered in virotherapy applications in the future

    Microsatellite instability, KRAS mutations and cellular distribution of TRAIL-receptors in early stage colorectal cancer.

    Get PDF
    Thus, we evaluated the immunofluorescence pattern of TRAIL-receptors and E-cadherin to assess the fraction of membrane-bound TRAIL-receptors in 231 selected patients with early-stage CRC undergoing surgical treatment only. Moreover, we investigated whether membrane staining for TRAIL-receptors as well as the presence of KRAS mutations or of microsatellite instability (MSI) had an effect on survival and thus a prognostic effect. The fact that the receptors for the TNF-related apoptosis inducing ligand (TRAIL) are almost invariably expressed in colorectal cancer (CRC) represents the rationale for the employment of TRAIL-receptors targeting compounds for the therapy of patients affected by this tumor. Yet, first reports on the use of these bioactive agents provided disappointing results. We therefore hypothesized that loss of membrane-bound TRAIL-R might be a feature of some CRC and that the evaluation of membrane staining rather than that of the overall expression of TRAIL-R might predict the response to TRAIL-R targeting compounds in this tumor. As expected, almost all CRC samples stained positive for TRAIL-R1 and 2. Instead, membrane staining for these receptors was positive in only 71% and 16% of samples respectively. No correlation between KRAS mutation status or MSI-phenotype and prognosis could be detected. TRAIL-R1 staining intensity correlated with survival in univariate analysis, but only membranous staining of TRAIL-R1 and TRAIL-R2 on cell membranes was an independent predictor of survival (cox multivariate analysis: TRAIL-R1: p = 0.019, RR 2.06[1.12-3.77]; TRAIL-R2: p = 0.033, RR 3.63[1.11-11.84]). In contrast to the current assumptions, loss of membrane staining for TRAIL-receptors is a common feature of early stage CRC which supersedes the prognostic significance of their staining intensity. Failure to achieve therapeutic effects in recent clinical trials using TRAIL-receptors targeting compounds might be due to insufficient selection of patients bearing tumors with membrane-bound TRAIL-receptors

    JNK inhibition sensitises hepatocellular carcinoma cells but not normal hepatocytes to the TNF-related apoptosis-inducing ligand.

    Get PDF
    Background: cJun terminal kinase (JNK) is constitutively activated in most hepatocellular carcinomas (HCCs), yet its exact role in carcinogenesis remains controversial. While tumour necrosis factor (TNF)-related apoptosisinducing ligand (TRAIL) is known as a major mediator of acquired immune tumour surveillance, and is currently being tested in clinical trials as a novel cancer therapy, the resistance of many tumours to TRAIL and concerns about its toxicity in vivo represent obstacles to its clinical application. In this study we investigated whether JNK activity in HCC could contribute to the resistance to apoptosis in these tumours. Methods: The effect of JNK/Jun inhibition on receptormediated apoptosis was analysed by pharmacological inhibition or RNA interference in cancer cells and nontumour cells isolated from human liver or transgenic mice lacking a phosphorylation site for Jun. Results: JNK inhibition caused cell cycle arrest, enhanced caspase recruitment, and greatly sensitised HCC cells but not normal hepatocytes to TRAIL. TRAILinduced activation of JNK could be effectively interrupted by administration of the JNK inhibitor SP600125. Conclusions: Expression and TRAIL-dependent feedback activation of JNK likely represent a mechanism by which cancer cells escape TRAIL-mediated tumour surveillance. JNK inhibition might represent a novel strategy for specifically sensitising HCC cells to TRAIL thus opening promising therapeutic perspectives for safe and effective use of TRAIL in cancer treatment

    Genetic inactivation of the Fanconi anemia gene FANCC identified in the hepatocellular carcinoma cell line HuH-7 confers sensitivity towards DNA-interstrand crosslinking agents

    Get PDF
    Background: Inactivation of the Fanconi anemia (FA) pathway through defects in one of 13 FA genes occurs at low frequency in various solid cancer entities among the general population. As FA pathway inactivation confers a distinct hypersensitivity towards DNA interstrand-crosslinking (ICL)-agents, FA defects represent rational targets for individualized therapeutic strategies. Except for pancreatic cancer, however, the prevalence of FA defects in gastrointestinal (GI) tumors has not yet been systematically explored. Results: A panel of GI cancer cell lines was screened for FA pathway inactivation applying FANCD2 monoubiquitination and FANCD2/RAD51 nuclear focus formation and a newly identified FA pathway-deficient cell line was functionally characterized. The hepatocellular carcinoma (HCC) line HuH-7 was defective in FANCD2 monoubiquitination and FANCD2 nuclear focus formation but proficient in RAD51 focus formation. Gene complementation studies revealed that this proximal FA pathway inactivation was attributable to defective FANCC function in HuH-7 cells. Accordingly, a homozygous inactivating FANCC nonsense mutation (c.553C > T, p.R185X) was identified in HuH-7, resulting in partial transcriptional skipping of exon 6 and leading to the classic cellular FA hypersensitivity phenotype; HuH-7 cells exhibited a strongly reduced proliferation rate and a pronounced G2 cell cycle arrest at distinctly lower concentrations of ICL-agents than a panel of non-isogenic, FA pathway-proficient HCC cell lines. Upon retroviral transduction of HuH-7 cells with FANCC cDNA, FA pathway functions were restored and ICL-hypersensitivity abrogated. Analyses of 18 surgical HCC specimens yielded no further examples for genetic or epigenetic inactivation of FANCC, FANCF, or FANCG in HCC, suggesting a low prevalence of proximal FA pathway inactivation in this tumor type. Conclusions: As the majority of HCC are chemoresistant, assessment of FA pathway function in HCC could identify small subpopulations of patients expected to predictably benefit from individualized treatment protocols using ICL-agents

    RNA interference assay for eIF2B epsilon (eIF2Bε).

    No full text
    <p><b>A)</b> Western blot analysis of mock-infected cultures was performed for each experiment to assess the efficiency of RNA silencing. <b>B)</b> HepG2, Huh-7 and PH5CH8 cells were transfected with siRNA for eIF2B<b>ε</b> at a concentration of 100 nM. As controls, cells were transfected in parallel with control siRNA or mock-transfected. At 72 hours post-transfection, cells were infected with VSV-wt using an MOI of 0.1 for 8 hours. Results are the average of three independent experiments and error bars indicate the standard deviation.</p

    Rapamycin activity on VSV replication.

    No full text
    <p><b>A)</b> HepG2, Huh-7 and PH5CH8 cells were incubated overnight with increasing concentrations of rapamycin (20, 100 and 500 nM), or in the case of the controls, DMSO was added. VSV infection was performed at an MOI of 0.1 for 24 hours in the presence of fresh inhibitor. The viral titers shown are the average of three independent experiments. <b>B)</b> All cell lines were treated with 50 nM of rapamycin as described above and infected with VSV-wt at an MOI of 0.1. Viral titers were determined at 8 hours post-infection. The data represent at least two independent experiments ± standard deviation. <b>C)</b> Cell lysates of mock- and rapamycin-treated cells were analysed by Western blot for detection of the phosphorylated forms of kinase p70S6k and eIF4E and their corresponding base-line expression. <b>D)</b> MTT proliferation assay in mock-treated (DMSO) and rapamycin-treated (RAPA) cultures. Data represent the mean of at least three independent experiments ± standard deviation (* p< 0.05; *** p<0.001).</p

    Immortalized human hepatocytes, PH5CH8 cell line.

    No full text
    <p><b>A)</b> VSV growth in immortalized non-neoplastic hepatocytes (PH5CH8) was compared to HCC cell lines and primary human hepatocytes (PHH). Cells were infected with VSV-wt and the IFN-inducer mutant VSV-M51R at an MOI of 0.001 and titers were determined at different time-points post-infection as indicated. Data shown are the average of three independent experiments and error bars represent standard deviation. Significance of viral titers in PHH was calculated by comparison with titers in Huh-7 (** p<0.01). <b>B)</b> Proliferation of the cell lines: HepG2, Huh-7 and PH5CH8. Cells were plated at the concentration of 5Ă—10<sup>3</sup> cells per well, and their numbers were determined up to four days after plating by MTT proliferation assay. Data are representative of three independent experiments. <b>C)</b> Western blot showing the expression levels of cyclin D1 and CDK4 in HepG2, Huh-7 and PH5CH8 cells compared to PHH.</p
    corecore