77 research outputs found

    The oncogenic properties of EWS/WT1 of desmoplastic small round cell tumors are unmasked by loss of p53 in murine embryonic fibroblasts

    Full text link
    BACKGROUND: Desmoplastic small round cell tumor (DSRCT) is characterized by the presence of a fusion protein EWS/WT1, arising from the t (11;22) (p13;q12) translocation. Here we examine the oncogenic properties of two splice variants of EWS/WT1, EWS/WT1-KTS and EWS/WT1 + KTS. METHODS: We over-expressed both EWS/WT1 variants in murine embryonic fibroblasts (MEFs) of wild-type, p53+/- and p53-/- backgrounds and measured effects on cell-proliferation, anchorage-independent growth, clonogenicity after serum withdrawal, and sensitivity to cytotoxic drugs and gamma irradiation in comparison to control cells. We examined gene expression profiles in cells expressing EWS/WT1. Finally we validated our key findings in a small series of DSRCT. RESULTS: Neither isoform of EWS/WT1 was sufficient to transform wild-type MEFs however the oncogenic potential of both was unmasked by p53 loss. Expression of EWS/WT1 in MEFs lacking at least one allele of p53 enhanced cell-proliferation, clonogenic survival and anchorage-independent growth. EWS/WT1 expression in wild-type MEFs conferred resistance to cell-cycle arrest after irradiation and daunorubicin induced apoptosis. We show DSRCT commonly have nuclear localization of p53, and copy-number amplification of MDM2/MDMX. Expression of either isoform of EWS/WT1 induced characteristic mRNA expression profiles. Gene-set enrichment analysis demonstrated enrichment of WNT pathway signatures in MEFs expressing EWS/WT1 + KTS. Wnt-activation was validated in cell lines with over-expression of EWS/WT1 and in DSRCT. CONCLUSION: In conclusion, we show both isoforms of EWS/WT1 have oncogenic potential in MEFs with loss of p53. In addition we provide the first link between EWS/WT1 and Wnt-pathway signaling. These data provide novel insights into the function of the EWS/WT1 fusion protein which characterize DSRCT

    Epigenetic targeting of Hedgehog pathway transcriptional output through BET bromodomain inhibition

    Get PDF
    Hedgehog signaling drives oncogenesis in several cancers and strategies targeting this pathway have been developed, most notably through inhibition of Smoothened. However, resistance to Smoothened inhibitors occurs via genetic changes of Smoothened or other downstream Hedgehog components. Here, we overcome these resistance mechanisms by modulating GLI transcription via inhibition of BET bromodomain proteins. We show the BET bromodomain protein, BRD4, regulates GLI transcription downstream of SMO and SUFU and chromatin immunoprecipitation studies reveal BRD4 directly occupies GLI1 and GLI2 promoters, with a substantial decrease in engagement of these sites upon treatment with JQ1, a small molecule inhibitor targeting BRD4. Globally, genes associated with medulloblastoma-specific GLI1 binding sites are downregulated in response to JQ1 treatment, supporting direct regulation of GLI activity by BRD4. Notably, patient- and GEMM-derived Hedgehog-driven tumors (basal cell carcinoma, medulloblastoma and atypical teratoid/rhabdoid tumor) respond to JQ1 even when harboring genetic lesions rendering them resistant to Smoothened antagonists

    Retrospective evaluation of whole exome and genome mutation calls in 746 cancer samples

    No full text
    Funder: NCI U24CA211006Abstract: The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) curated consensus somatic mutation calls using whole exome sequencing (WES) and whole genome sequencing (WGS), respectively. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2,658 cancers across 38 tumour types, we compare WES and WGS side-by-side from 746 TCGA samples, finding that ~80% of mutations overlap in covered exonic regions. We estimate that low variant allele fraction (VAF < 15%) and clonal heterogeneity contribute up to 68% of private WGS mutations and 71% of private WES mutations. We observe that ~30% of private WGS mutations trace to mutations identified by a single variant caller in WES consensus efforts. WGS captures both ~50% more variation in exonic regions and un-observed mutations in loci with variable GC-content. Together, our analysis highlights technological divergences between two reproducible somatic variant detection efforts

    The role of the EWS/WT1 fusion protein in the pathogenesis of desmoplastic small round cell tumor

    No full text
    © 2012 Dr. Pratiti BandopadhayayDesmoplastic small round cell tumor (DSRCT) is an aggressive tumor characterised by the t(11;22) translocation which results in the fusion of exons 1-VII of the Ewing’s sarcoma gene (EWS) to exons VIII-X of the Wilms tumor gene (WT1). Little is understood in regards to the role of the fusion protein in the pathogenesis of this tumor. Two splice variants of the fusion protein are described, which result in the insertion (+KTS) or absence (-KTS) of a three amino acid sequence in the WT1 zinc binding fingers. Using inducible lentiviral expression vectors, EWS/WT1 was expressed in primary murine embryonic fibroblasts (MEFs) derived from E14.5 embryos of C57BL6 or p53 knock-out mice. EWS/WT1-KTS or EWS/WT1+KTS were insufficient to transform wild type MEFs, however in p53 knock-out MEFs both isoforms of EWS/WT1 were able to transform the cells. Primary MEFs expressing EWS/WT1-KTS or EWS/WT1+KTS demonstrated attenuated p53 mediated responses. Microarray analysis demonstrated different expression profiles of cells expressing EWS/WT1 compared to eGFP controls and pathway analysis shows differential expression of genes involved Wnt signaling. Analysis of a series of DSRCT samples revealed both transcripts of EWS/WT1 are present with the EWS/WT1+KTS transcript being more abundant. There was no evidence of p53 mutations in exons five to eight however there was a high incidence of MDM2/MDM4 amplification in a series of 15 tumors examined, representing a mechanism by which p53 function may be suppressed in the tumours. -catenin immunohistochemistry in the tumors revealed nuclear immunoreactivity of -catenin, suggesting up-regulation of Wnt signaling and this was validated with real time PCR. These results provide novel insights into the oncogenic properties of EWS/WT1
    • …
    corecore