Erwin Tomasich
Medical University of Vienna
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Erwin Tomasich.
Scientific Reports | 2015
Peter Horak; Erwin Tomasich; Petr Vaňhara; Kateřina Kratochvílová; Mariam Anees; Maximilian Marhold; Christof E. Lemberger; Marion Gerschpacher; Reinhard Horvat; Maria Sibilia; Dietmar Pils; Michael Krainer
Prostate cancer is the most prevalent cancer in males in developed countries. Tumor suppressor candidate 3 (TUSC3) has been identified as a putative tumor suppressor gene in prostate cancer, though its function has not been characterized. TUSC3 shares homologies with the yeast oligosaccharyltransferase (OST) complex subunit Ost3p, suggesting a role in protein glycosylation. We provide evidence that TUSC3 is part of the OST complex and affects N-linked glycosylation in mammalian cells. Loss of TUSC3 expression in DU145 and PC3 prostate cancer cell lines leads to increased proliferation, migration and invasion as well as accelerated xenograft growth in a PTEN negative background. TUSC3 downregulation also affects endoplasmic reticulum (ER) structure and stress response, which results in increased Akt signaling. Together, our findings provide first mechanistic insight in TUSC3 function in prostate carcinogenesis in general and N-glycosylation in particular.
International Journal of Cancer | 2015
Kateřina Kratochvílová; Peter Horak; Milan Ešner; Karel Souček; Dietmar Pils; Mariam Anees; Erwin Tomasich; František Dráfi; Veronika Jurtíková; Aleš Hampl; Michael Krainer; Petr Vaňhara
Ovarian cancer is one of the most common malignancies in women and contributes greatly to cancer‐related deaths. Tumor suppressor candidate 3 (TUSC3) is a putative tumor suppressor gene located at chromosomal region 8p22, which is often lost in epithelial cancers. Epigenetic silencing of TUSC3 has been associated with poor prognosis, and hypermethylation of its promoter provides an independent biomarker of overall and disease‐free survival in ovarian cancer patients. TUSC3 is localized to the endoplasmic reticulum in an oligosaccharyl tranferase complex responsible for the N‐glycosylation of proteins. However, the precise molecular role of TUSC3 in ovarian cancer remains unclear. In this study, we establish TUSC3 as a novel ovarian cancer tumor suppressor using a xenograft mouse model and demonstrate that loss of TUSC3 alters the molecular response to endoplasmic reticulum stress and induces hallmarks of the epithelial‐to‐mesenchymal transition in ovarian cancer cells. In summary, we have confirmed the tumor‐suppressive function of TUSC3 and identified the possible mechanism driving TUSC3‐deficient ovarian cancer cells toward a malignant phenotype.
Molecular Cancer Research | 2015
Maximilian Marhold; Erwin Tomasich; Ahmed El-Gazzar; Gerwin Heller; Andreas Spittler; Reinhard Horvat; Michael Krainer; Peter Horak
Tumor-initiating subpopulations of cancer cells, also known as cancer stem cells (CSC), were recently identified and characterized in prostate cancer. A well-characterized murine model of prostate cancer was used to investigate the regulation of hypoxia-inducible factor 1α (HIF1A/HIF1α) in CSCs and a basal stem cell subpopulation (Lin−/Sca-1+/CD49f+) was identified, in primary prostate tumors of mice, with elevated HIF1α expression. To further analyze the consequences of hypoxic upregulation on stem cell proliferation and HIF1α signaling, CSC subpopulations from murine TRAMP-C1 cells (Sca-1+/CD49f+) as well as from a human prostate cancer cell line (CD44+/CD49f+) were isolated and characterized. HIF1α levels and HIF target gene expression were elevated in hypoxic CSC-like cells, and upregulation of AKT occurred through a mechanism involving an mTOR/S6K/IRS-1 feedback loop. Interestingly, resistance of prostate CSCs to selective mTOR inhibitors was observed because of HIF1α upregulation. Thus, prostate CSCs show a hypoxic deactivation of a feedback inhibition of AKT signaling through IRS-1. In light of these results, we propose that deregulation of the PI3K/AKT/mTOR pathway through HIF1α is critical for CSC quiescence and maintenance by attenuating CSC metabolism and growth via mTOR and promoting survival by AKT signaling. We also propose that prostate CSCs can exhibit primary drug resistance to selective mTOR inhibitors. Implications: This work contributes to a deeper understanding of hypoxic regulatory mechanisms in CSCs and will help devise novel therapies against prostate cancer. Mol Cancer Res; 13(3); 556–64. ©2014 AACR.
Journal of Hematology & Oncology | 2015
Gerwin Heller; Anna Rommer; Katarina Steinleitner; Julia Etzler; Hubert Hackl; Petra Heffeter; Erwin Tomasich; Martin Filipits; Birgit Steinmetz; Thais Topakian; Simone Klingenbrunner; Barbara Ziegler; Andreas Spittler; Sabine Zöchbauer-Müller; Walter Berger; Rotraud Wieser
BackgroundThe transcription factor Ecotropic Virus Integration site 1 (EVI1) regulates cellular proliferation, differentiation, and apoptosis, and its overexpression contributes to an aggressive course of disease in myeloid leukemias and other malignancies. Notwithstanding, knowledge about the target genes mediating its biological and pathological functions remains limited. We therefore aimed to identify and characterize novel EVI1 target genes in human myeloid cells.MethodsU937T_EVI1, a human myeloid cell line expressing EVI1 in a tetracycline regulable manner, was subjected to gene expression profiling. qRT-PCR was used to confirm the regulation of membrane-spanning-4-domains subfamily-A member-3 (MS4A3) by EVI1. Reporter constructs containing various parts of the MS4A3 upstream region were employed in luciferase assays, and binding of EVI1 to the MS4A3 promoter was investigated by chromatin immunoprecipitation. U937 derivative cell lines experimentally expressing EVI1 and/or MS4A3 were generated by retroviral transduction, and tested for their tumorigenicity by subcutaneous injection into severe combined immunodeficient mice.ResultsGene expression microarray analysis identified 27 unique genes that were up-regulated, and 29 unique genes that were down-regulated, in response to EVI1 induction in the human myeloid cell line U937T. The most strongly repressed gene was MS4A3, and its down-regulation by EVI1 was confirmed by qRT-PCR in additional, independent experimental model systems. MS4A3 mRNA levels were also negatively correlated with those of EVI1 in several published AML data sets. Reporter gene assays and chromatin immunoprecipitation showed that EVI1 regulated MS4A3 via direct binding to a promoter proximal region. Experimental re-expression of MS4A3 in an EVI1 overexpressing cell line counteracted the tumor promoting effect of EVI1 in a murine xenograft model by increasing the rate of apoptosis.ConclusionsOur data reveal MS4A3 as a novel direct target of EVI1 in human myeloid cells, and show that its repression plays a role in EVI1 mediated tumor aggressiveness.
Cancer Research | 2012
Peter Horak; Erwin Tomasich; Mariam Anees; Maximilian Marhold; Marion Gerschpacher; Dietmar Pils; Michael Krainer
Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Prostate cancer is the most prevalent cancer in males in developed countries. Molecular aberrations in the pathogenesis of this disease are numerous, though losses on the short arm of chromosome 8 are highly prevalent. In our previous studies, hypermethylation and diminished TUSC3 expression turned out to be significantly associated with poor progression free and overall survival in ovarian cancer. Additionally, expression profiling of TUSC3 negative cancer cell lines revealed deregulation of genes involved in ‘cell structure and motility’ and ‘development processes’, further supporting our hypothesis of TUSC3 as a tumor suppressor. In silico prediction of TUSC3 shows a high homology of TUSC3 to the Ost3p subunit of the oligosaccharyltransferase (OST) complex in yeast, hinting to its role in protein glycosylation. In this project, we characterized TUSC3 (N33), a putative tumor suppressor gene on 8p, in prostate cancer. To study the prognostic role of TUSC3 in prostate cancer we used a comprehensive prostate cancer tissue microarray, comprised of samples from 143 prostate cancer patients. Serum DNA methylation was studied in an additional cohort of 67 prostate cancer patients. Statistical analysis demonstrated low or missing TUSC3 protein expression in 56.6% of prostate cancer patients based on immunohistochemical staining. Methylation of the TUSC3 promoter was observed in 38.8% of prostate cancer patients’ serum DNA, confirming our previous findings of TUSC3 promoter hypermethylation as a possible epigenetic regulatory mechanism. In our prostate cancer cohorts, however, neither expression nor hypermethylation of TUSC3 had a significant effect on overall survival, possibly due to the relatively short follow-up period. Next, we studied the function of TUSC3 in N-glycosylation and in particular its influence on carcinogenesis using a cell culture model. We downregulated TUSC3 expression in prostate cancer cell lines DU145 and PC3 using shRNA mediated knockdown and analyzed them for their proliferative and carcinogenic properties in vitro. We could show that loss of TUSC3 expression confers growth advantage of prostate cancer cell lines, in particular under conditions of serum starvation. We also observed increased migratory properties of prostate cancer cell lines upon TUSC3 knockdown. Further, influence of TUSC3 downregulation on several putative targets of N-linked glycosylation has been analyzed in vitro. In our study, we characterized TUSC3 expression and/or methylation in prostate cancer patients and obtained a first mechanistic insight into its function in N-glycosylation and carcinogenesis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3997. doi:1538-7445.AM2012-3997
The Journal of Pathology | 2018
Gerwin Heller; Corinna Altenberger; Irene Steiner; Thais Topakian; Barbara Ziegler; Erwin Tomasich; György Lang; Adelheid End-Pfützenreuter; Sonja Zehetmayer; Balazs Dome; Britt Madeleine Arns; Walter Klepetko; Christoph Zielinski; Sabine Zöchbauer-Müller
Deregulated DNA methylation leading to transcriptional inactivation of certain genes occurs frequently in non‐small‐cell lung cancers (NSCLCs). As well as protein‐coding genes, microRNA (miRNA)‐coding genes may be targets for methylation in NSCLCs; however, the number of known methylated miRNA genes is still small. Thus, we investigated methylation of miRNA genes in primary tumour (TU) samples and corresponding non‐malignant lung tissue (NL) samples of 50 NSCLC patients by using methylated DNA immunoprecipitation followed by custom‐designed tiling microarray analyses (MeDIP‐chip), and 252 differentially methylated probes between TU samples and NL samples were identified. These probes were annotated, which resulted in the identification of 34 miRNA genes with increased methylation in TU samples. Some of these miRNA genes were already known to be methylated in NSCLCs (e.g. those encoding miR‐9‐3 and miR‐124), but methylation of the vast majority of them was previously unknown. We selected six miRNA genes (those encoding miR‐10b, miR‐1179, miR‐137, miR‐572, miR‐3150b, and miR‐129‐2) for gene‐specific methylation analyses in TU samples and corresponding NL samples of 104 NSCLC patients, and observed a statistically significant increase in methylation of these genes in TU samples (p < 0.0001). In silico target prediction of the six miRNAs identified several oncogenic/cell proliferation‐promoting factors (e.g. CCNE1 as an miR‐1179 target). To investigate whether miR‐1179 indeed targets CCNE1, we transfected miR‐1179 gene mimics into CCNE1‐expressing NSCLC cells, and observed downregulated CCNE1 mRNA expression in these cells as compared with control cells. Similar effects on cyclin E1 expression were seen in western blot analyses. In addition, we found a statistically significant reduction in the growth of NSCLC cells transfected with miR‐1179 mimics as compared with control cells. In conclusion, we identified many methylated miRNA genes in NSCLC patients, and found that the miR‐1179 gene is a potential tumour cell growth suppressor in NSCLCs. Overall, our findings emphasize the impact of miRNA gene methylation on the pathogenesis of NSCLCs.
Cancer Research | 2016
Michael Schwarz; Erwin Tomasich; Maximilian Marhold; Andreas Heinzel; Paul Perco; Peter Horak; Bernd Mayer; Michael Krainer
Synthetic lethality describes an interdependent relationship between two genes, where the loss of either one alone can be compensated, while the simultaneous loss of both genes causes a non-viable phenotype. In recent years, first therapeutics based on this concept entered the clinic, most notably the PARP inhibitors for BRCA1/2-mutated cancers. In the present study, we analyzed synthetic lethal interactions in yeast to identify new and potentially synergistic drug combinations for breast cancer therapy. We were able to confirm significantly enhanced cytotoxicity for predicted drug pairs in breast cancer cell lines in vitro. First, a predictor built from publicly available yeast genetic interactions in the Data Repository of Yeast Genetic INteraction (DRYGIN) was used to predict potential synthetic lethal genetic interactions in human. Independently, a data set containing all pharmacological approaches, targeted or cytostatic, in breast cancer therapy was created. These drug combinations and their respective targets were then analyzed regarding their coverage of predicted synthetic lethal interactions. New drug combinations, previously unused in breast cancer therapy, were identified in silico by combining drugs already in use for breast cancer therapy (individually or in other combinations) in such a manner that their combination covers one or more potential synthetic lethal gene pairs. From this set of new drug combination and their synthetic lethal gene pairs we further pursued the predicted interdependencies between farnesyl diphosphate synthase (FDPS) and tubulin, beta 1 (TUBB1) and between FDPS and phosphoinositide-dependent kinase-1 (PDPK1) as well as prostaglandin-endoperoxide synthase 2 (PTGS2). Drugs targeting these genes are celecoxib (PDPK1, PTGS2), an anti-inflammatory drug, and zoledronic acid (FDPS), a bone degradation inhibitor, as well as the cytotoxic agent paclitaxel (TUBB1). We performed cell viability and cytotoxicity assays to determine therapeutic effects of celecoxib, zoledronic acid and paclitaxel alone and in combination on selected breast cancer cell lines. Our results showed statistically significant decreases in cell viability for the combinational treatment with zoledronic acid and paclitaxel as well as with the per se non-cytotoxic combination of zoledronic acid and celecoxib when compared to single agent treatment. In conclusion, we present a bioinformatics approach to predict potentially synergistic gene interactions based on synthetic lethality found in yeast and a strategy for utilizing these interactions for identifying new potentially synergistic drug combinations. Citation Format: Michael Schwarz, Erwin Tomasich, Maximilian Marhold, Andreas Heinzel, Paul Perco, Peter Horak, Bernd Mayer, Michael Krainer. Drug combinations for breast cancer based on synthetic lethality screens in yeast. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2697.
Cancer Research | 2016
Maximilian Marhold; Erwin Tomasich; Simon Udovica; Gerwin Heller; Corinna Altenberger; Andreas Spittler; Reinhard Horvat; Peter Horak; Michael Krainer
Neuroendocrine differentiation of prostate cancer (PCa) occurs frequently during the development of castration resistance and rarely in primary tumors. Using the transgenic mouse model of prostate cancer (TRAMP) in a B6/C57 background, we were able to detect, isolate and further characterize basal, luminal and neuroendocrine subsets of cancer cells. We performed allograft experiments in NSG mice to ensure cellular stem and progenitor properties as well as metastatic potential, and thus created a murine model of neuroendocrine prostate cancer. Whilst all three previously described cell populations were present in approximately three quarters of primary TRAMP tumors, some tumors lacked basal stem cells and showed a more aggressive phenotype. These tumors, mainly consisting of small cancer cells, expressed markers of neuroendocrine differentiation such as synaptophysin and chromogranin A as shown by immunohistochemistry and are further referred to as neuroendocrine carcinomas (NECs), as ruled by an experienced uropathologist of our institution. Adenocarcinoma-like tumors (ACs), in contrast, showed high expression of cytokeratins and retained glandular histology. Using fluorescence-activated cell sorting (FACS) against newly discovered NEC markers within the TRAMP model, we found a relative increase of neuroendocrine progenitors in prostate NECs compared to ACs (approximately 75 vs. 33%, respectively). In ACs, on the other hand, luminal progenitors were found to be the predominant drivers of cancer progression. To further evaluate this, we transplanted single-cell-suspensions into NSG mice without androgen supplementation and observed successful engraftment of both non-basal cell populations. Additionally, we were able to passage the resulted tumors for at least two generations and observed maintenance of histology and biological features for tumors of both luminal and neuroendocrine origin. Based on a RNA sequencing, we were able to define gene signatures for neuroendocrine and luminal progenitors, uncovering a number of novel potential therapeutic targets. Ultimately, we were able to evaluate the prognostic value of the signatures obtained from mice in the human disease by in-silico analyses of publically available gene expression profile databases. In conclusion, we created and characterized a murine model of neuroendocrine prostate cancer using flow cytometry and murine allografts. Further, we established gene expression signatures of luminal and neuroendocrine progenitors and translated them to the human disease. Our findings foster the understanding of neuroendocrine differentiation in prostate cancer and may help in developing new targeted approaches in this entity. Citation Format: Maximilian Marhold, Erwin Tomasich, Simon Udovica, Gerwin Heller, Corinna Altenberger, Andreas Spittler, Reinhard Horvat, Peter Horak, Michael Krainer. Neuroendocrine and luminal progenitors drive cancer progression in prostate cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2407.
Cancer Research | 2016
Corinna Altenberger; Gerwin Heller; Barbara Ziegler; Erwin Tomasich; Maximilian Marhold; Leonhard Müllauer; György Lang; Adelheid End-Pfützenreuter; Balazs Dome; Britt-Madeleine Arns; Walter Klepetko; Christoph Zielinski; Sabine Zöchbauer-Müller
DNA methylation is one of the major epigenetic mechanisms regulating transcriptional activity and is involved in the pathogenesis of non-small cell lung cancers (NSCLC). In a recent study we identified a large number of tumor-specifically methylated genes in NSCLCs (Heller et al., Carcinogenesis 2013). For further detailed analyses we selected the genes SPAG6 (Sperm Associated Antigen 6) and L1TD1 (LINE-1 Type Transposase Domain Containing 1). By analysing publically available IlluminaHiSeq RNA-seq data we observed frequent downregulation of SPAG6 and L1TD1 mRNA expression in primary tumor (TU) samples compared to corresponding non-malignant lung tissue (NL) samples of NSCLC patients. In addition, we investigated SPAG6 and L1TD1 mRNA expression in 5 NSCLC cell lines and found SPAG6 as well as L1TD1 mRNA expression frequently downregulated in all of these cell lines compared to normal human bronchial epithelial cells (NHBECs). Subsequently, we treated cells of NSCLC cell lines which did not express SPAG6 or L1TD1 with the epigenetically active drugs 5-aza-2′-deoxycytidine and Trichostatin A and observed re-expression of both genes suggesting that transcriptional regulation of SPAG6 and L1TD1 is mediated by DNA methylation in NSCLCs. Bisulfite genomic sequencing of parts of the 5′ region of SPAG6 and L1TD1 revealed that the vast majority of CpG sites indeed are methylated in NSCLC cells in contrast to NHBECs. Moreover, we analysed SPAG6 and L1TD1 methylation in TU and NL samples of 147 stage I-III NSCLC patients using the gene-specific approach methylation-sensitive high resolution melt analysis (MS-HRM). Differences in SPAG6 as well as in L1TD1 methylation between TU and NL samples were statistically significant for both genes and confirmed that SPAG6 and L1TD1 are tumor-specifically methylated in NSCLCs. Additionally, we investigated SPAG6 and L1TD1 protein expression in TU and NL samples of 35 NSCLC patients by immunohistochemistry. In the vast majority of SPAG6 or L1TD1 methylated TU samples, protein expression of these genes was downregulated in tumor cells. Moreover, we performed cell proliferation, cell viability and colony formation assays in vitro and observed that ectopic expression of L1TD1 but not of SPAG6 reduced tumor cell proliferation, viability and the ability of NSCLC cells to form colonies. To further investigate the role of L1TD1 in the pathogenesis of NSCLCs, in vivo studies are being carried out. Overall, our results demonstrate that DNA methylation is the major mechanism for frequent downregulation of SPAG6 and L1TD1 expression in NSCLCs. Citation Format: Corinna Altenberger, Gerwin Heller, Barbara Ziegler, Erwin Tomasich, Maximilian Marhold, Leonhard Mullauer, Gyorgy Lang, Adelheid End-Pfutzenreuter, Balazs Dome, Britt-Madeleine Arns, Walter Klepetko, Christoph C. Zielinski, Sabine Zochbauer-Muller. SPAG6 and L1TD1 are transcriptionally regulated by DNA methylation in non-small cell lung cancers. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2772.
Cancer Research | 2014
Maximilian Marhold; Erwin Tomasich; Zuzana Pernicová; Radek Fedr; Karel Souček; Andreas Spittler; Michael Krainer; Peter Horak
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Tumor initiating subpopulations of cancer cells, also known as cancer progenitor or cancer stem cells (CSCs) were recently identified and characterized in prostate cancer. Emerging evidence suggests that hypoxia-inducible factor 1 alpha (HIF1A) contributes to CSC maintenance and influences the PI3K/AKT/mTOR axis, a pathway which is frequently altered in prostate cancer. In our study, we assessed the effects of hypoxia on PI3K/AKT/mTOR signaling in a subset of basal prostate cancer stem cells in a murine model of prostate cancer. First, we successfully isolated Lin-/Sca-1+/CD49f+ basal stem cell subpopulations from primary prostate tumors of TRAMP (transgenic adenocarcinoma of mouse prostate) mice. We observed a consistent decrease in S6 phosphorylation within the CSC subpopulation, in agreement with an mTOR pathway inhibition. However, AKT phosphorylation in the CSC subpopulation was simultaneously elevated, thus prompting further investigations. To analyze the molecular pathways leading to this deregulation, we used a cell line model and isolated Sca-1+/CD49f+ CSC-like subpopulations from the murine prostate cancer cell line TRAMP-C1 and CD44+/CD49f+ CSC-like subpopulations from the androgen independent human prostate cancer cell line DU145. We used sphere formation assays and expression of Notch 1 and Oct 3/4 to confirm the stem and progenitor cell properties of the sorted subpopulations. We could confirm the decrease in mTOR/S6 signaling and identify the upregulation of AKT through IRS-1 mediated feedback loop in prostate CSC-like cells under hypoxic conditions. This AKT/mTOR deregulation was promptly reversed in normoxia, suggestive of the involvement of hypoxia inducible factors. We found elevated HIF1A protein levels as well as increased target gene expression in CSC subpopulations in vitro and in vivo. Further, lentiviral knockdown of HIF1A restored AKT/mTOR activity in CSC-like cells. We could show that CSC-like prostate cancer cells with elevated expression of basal stem cell markers, such as CD44, Sca-1 or CD49f, show a hypoxic deactivation of feedback inhibition on AKT signaling through IRS-1. In light of these results, we propose that deregulation of the PI3K/AKT/mTOR pathway via HIF1A may be critical for cancer stem cell quiescence and maintenance, slowing down cancer stem cell metabolism and growth via mTOR and promoting their survival via AKT signaling. Citation Format: Maximilian Marhold, Erwin Tomasich, Zuzana Pernicova, Radek Fedr, Karel Soucek, Andreas Spittler, Michael Krainer, Peter Horak. Hypoxia leads to deregulation of PI3K/AKT/mTOR signaling in prostate cancer stem cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3047. doi:10.1158/1538-7445.AM2014-3047