Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jasmin Svinka is active.

Publication


Featured researches published by Jasmin Svinka.


Nature Communications | 2015

Disruption of STAT3 signalling promotes KRAS-induced lung tumorigenesis

Beatrice Grabner; Daniel Schramek; Kristina M. Mueller; Herwig P. Moll; Jasmin Svinka; Thomas Hoffmann; Eva Bauer; Leander Blaas; Natascha Hruschka; Katalin Zboray; Patricia Stiedl; Harini Nivarthi; Edith Bogner; Wolfgang Gruber; Thomas Mohr; Ralf Harun Zwick; Lukas Kenner; Valeria Poli; Fritz Aberger; Dagmar Stoiber; Gerda Egger; Harald Esterbauer; Johannes Zuber; Richard Moriggl; Robert Eferl; Balazs Gyorffy; Josef M. Penninger; Helmut Popper; Emilio Casanova

STAT3 is considered to play an oncogenic role in several malignancies including lung cancer; consequently, targeting STAT3 is currently proposed as therapeutic intervention. Here we demonstrate that STAT3 plays an unexpected tumour-suppressive role in KRAS mutant lung adenocarcinoma (AC). Indeed, lung tissue-specific inactivation of Stat3 in mice results in increased KrasG12D-driven AC initiation and malignant progression leading to markedly reduced survival. Knockdown of STAT3 in xenografted human AC cells increases tumour growth. Clinically, low STAT3 expression levels correlate with poor survival and advanced malignancy in human lung AC patients with smoking history, which are prone to KRAS mutations. Consistently, KRAS mutant lung tumours exhibit reduced STAT3 levels. Mechanistically, we demonstrate that STAT3 controls NF-κB-induced IL-8 expression by sequestering NF-κB within the cytoplasm, thereby inhibiting IL-8-mediated myeloid tumour infiltration and tumour vascularization and hence tumour progression. These results elucidate a novel STAT3–NF-κB–IL-8 axis in KRAS mutant AC with therapeutic and prognostic relevance.


Hepatology | 2015

Growth hormone resistance exacerbates cholestasis-induced murine liver fibrosis

Patricia Stiedl; Robert McMahon; Leander Blaas; Victoria Stanek; Jasmin Svinka; Beatrice Grabner; Gernot Zollner; Sonja M. Kessler; Thierry Claudel; Mathias Müller; Wolfgang Mikulits; Martin Bilban; Harald Esterbauer; Robert Eferl; Johannes Haybaeck; Michael Trauner; Emilio Casanova

Growth hormone (GH) resistance has been associated with liver cirrhosis in humans but its contribution to the disease remains controversial. In order to elucidate whether GH resistance plays a causal role in the establishment and development of liver fibrosis, or rather represents a major consequence thereof, we challenged mice lacking the GH receptor gene (Ghr–/–, a model for GH resistance) by crossing them with Mdr2 knockout mice (Mdr2–/–), a mouse model of inflammatory cholestasis and liver fibrosis. Ghr–/–;Mdr2–/– mice showed elevated serum markers associated with liver damage and cholestasis, extensive bile duct proliferation, and increased collagen deposition relative to Mdr2–/– mice, thus suggesting a more severe liver fibrosis phenotype. Additionally, Ghr–/–;Mdr2–/– mice had a pronounced down‐regulation of hepatoprotective genes Hnf6, Egfr, and Igf‐1, and significantly increased levels of reactive oxygen species (ROS) and apoptosis in hepatocytes, compared to control mice. Moreover, single knockout mice (Ghr–/–) fed with a diet containing 1% cholic acid displayed an increase in hepatocyte ROS production, hepatocyte apoptosis, and bile infarcts compared to their wild‐type littermates, indicating that loss of Ghr renders hepatocytes more susceptible to toxic bile acid accumulation. Surprisingly, and despite their severe fibrotic phenotype, Ghr–/–;Mdr2–/– mice displayed a significant decrease in tumor incidence compared to Mdr2–/– mice, indicating that loss of Ghr signaling may slow the progression from fibrosis/cirrhosis to cancer in the liver. Conclusion: GH resistance dramatically exacerbates liver fibrosis in a mouse model of inflammatory cholestasis, therefore suggesting that GH resistance plays a causal role in the disease and provides a novel target for the development of liver fibrosis treatments. (Hepatology 2015;61:613‐626)


Methods of Molecular Biology | 2015

Induction of Colorectal Cancer in Mice and Histomorphometric Evaluation of Tumors

Ilija Crnčec; Paulina Pathria; Jasmin Svinka; Robert Eferl

Colorectal cancer (CRC) originates from the epithelial cells lining the colon or rectum of the gastrointestinal tract and represents the third most common form of cancer worldwide. CRC is frequently associated with Colitis Ulcerosa or Crohns Disease demonstrating the tumor-promoting role of inflammation. Colorectal tumor cells establish heterotypic interactions with inflammatory cells and cancer-associated fibroblasts in the tumor stroma that support tumor angiogenesis and are essential for tumor progression. Therefore, establishment of suitable mouse models mimicking the inflammatory etiology of CRC is important. Here we describe methods to induce CRC in mice, to quantify tumor parameters (multiplicity, tumor load, mean tumor size), and to analyze the cellular composition of the CRC tumor stroma.


Hepatic oncology | 2014

STAT3 in hepatocellular carcinoma: new perspectives

Jasmin Svinka; Wolfgang Mikulits; Robert Eferl

Chronic liver damage and inflammation are strong promoters of hepatocellular carcinoma (HCC) formation. HCC cells communicate with inflammatory and stromal cells via cytokine/chemokine signals. These heterotypic interactions inhibit immunologic anticancer activities and promote protumorigenic activities, such as angiogenesis or invasiveness. STAT3 mediates several reciprocal interactions between liver cancer cells and stromal cells and modulates preconditions of tumor formation such as chronic inflammation. Therefore, activation of STAT3 is considered as a tumor-promoting event in HCC formation. However, the oncogenic role of STAT3 in cancers has been challenged by several reports that suggest a tumor-suppressive activity. Here we discuss tumor-promoting and tumor-suppressive effects of cytokine-activated STAT3 in HCC.


OncoImmunology | 2015

Myeloid STAT3 promotes formation of colitis-associated colorectal cancer in mice

Paulina Pathria; Dagmar Gotthardt; Michaela Prchal-Murphy; Eva-Maria Putz; Martin Holcmann; Michaela Schlederer; Beatrice Grabner; Ilija Crnčec; Jasmin Svinka; Monica Musteanu; Thomas Hoffmann; Martin Filipits; Walter Berger; Valeria Poli; Lukas Kenner; Martin Bilban; Emilio Casanova; Mathias Müller; Birgit Strobl; Editha Bayer; Thomas Mohr; Veronika Sexl; Robert Eferl

Myeloid cells lacking STAT3 promote antitumor responses of NK and T cells but it is unknown if this crosstalk affects development of autochthonous tumors. We deleted STAT3 in murine myeloid cells (STAT3Δm) and examined the effect on the development of autochthonous colorectal cancers (CRCs). Formation of Azoxymethane/Dextransulfate (AOM/DSS)-induced CRCs was strongly suppressed in STAT3Δm mice. Gene expression profiling showed strong activation of T cells in the stroma of STAT3Δm CRCs. Moreover, STAT3Δm host mice were better able to control the growth of transplanted MC38 colorectal tumor cells which are known to be killed in a T cell-dependent manner. These data suggest that myeloid cells lacking STAT3 control formation of CRCs mainly via cross activation of T cells. Interestingly, the few CRCs that formed in STAT3Δm mice displayed enhanced stromalization but appeared normal in size indicating that they have acquired ways to escape enhanced tumor surveillance. We found that CRCs in STAT3Δm mice consistently activate STAT3 signaling which is implicated in immune evasion and might be a target to prevent tumor relapse.


Journal of Molecular Medicine | 2017

Epidermal growth factor signaling protects from cholestatic liver injury and fibrosis

Jasmin Svinka; Sandra Pflügler; Markus Mair; Hanns-Ulrich Marschall; Jan G. Hengstler; Patricia Stiedl; Valeria Poli; Emilio Casanova; Gerald Timelthaler; Maria Sibilia; Robert Eferl

We have demonstrated that the signal transducer and activator of transcription 3 (STAT3) protects from cholestatic liver injury. Specific ablation of STAT3 in hepatocytes and cholangiocytes (STAT3∆hc) aggravated liver damage and fibrosis in the Mdr2−/− (multidrug resistance 2) mouse model for cholestatic disease. Upregulation of bile acid biosynthesis genes and downregulation of epidermal growth factor receptor (EGFR) expression were observed in STAT3∆hc Mdr2−/− mice but the functional consequences of these processes in cholestatic liver injury remained unclear. Here, we show normal canalicular architecture and bile flow but increased amounts of bile acids in the bile of STAT3∆hc Mdr2−/− mice. Moreover, STAT3-deficient hepatocytes displayed increased sensitivity to bile acid-induced apoptosis in vitro. Since EGFR signaling has been reported to protect hepatocytes from bile acid-induced apoptosis, we generated mice with hepatocyte/cholangiocyte-specific ablation of EGFR (EGFR∆hc) and crossed them to Mdr2−/− mice. Importantly, deletion of EGFR phenocopied deletion of STAT3 and led to aggravated liver damage, liver fibrosis, and hyperproliferation of K19+ cholangiocytes. Our data demonstrate hepatoprotective functions of the STAT3-EGFR signaling axis in cholestatic liver disease.Key messageSTAT3 is a negative regulator of bile acid biosynthesis.STAT3 protects from bile acid-induced apoptosis and regulates EGFR expression.EGFR signaling protects from cholestatic liver injury and fibrosis.


Molecular Oncology | 2018

STAT1 is a sex‐specific tumor suppressor in colitis‐associated colorectal cancer

Ilija Crnčec; Madhura Modak; Claire Gordziel; Jasmin Svinka; Irene Scharf; Stefan Moritsch; Paulina Pathria; Michaela Schlederer; Lukas Kenner; Gerald Timelthaler; Mathias Müller; Birgit Strobl; Emilio Casanova; Editha Bayer; Thomas Mohr; Johannes Stöckl; Karlheinz Friedrich; Robert Eferl

The interferon‐inducible transcription factor STAT1 is a tumor suppressor in various malignancies. We investigated sex‐specific STAT1 functions in colitis and colitis‐associated colorectal cancer (CRC) using mice with specific STAT1 deletion in intestinal epithelial cells (STAT1∆IEC). Male but not female STAT1∆IEC mice were more resistant to DSS‐induced colitis than sex‐matched STAT1flox/flox controls and displayed reduced intraepithelial infiltration of CD8+ TCRαβ+ granzyme B+ T cells. Moreover, DSS treatment failed to induce expression of T‐cell‐attracting chemokines in intestinal epithelial cells of male but not of female STAT1∆IEC mice. Application of the AOM‐DSS protocol for induction of colitis‐associated CRC resulted in increased intestinal tumor load in male but not in female STAT1∆IEC mice. A sex‐specific stratification of human CRC patients corroborated the data obtained in mice and revealed that reduced tumor cell‐intrinsic nuclear STAT1 protein expression is a poor prognostic factor in men but not in women. These data demonstrate that epithelial STAT1 is a male‐specific tumor suppressor in CRC of mice and humans.


Leukemia | 2018

Dependency on the TYK2/STAT1/MCL1 axis in anaplastic large cell lymphoma

Nicole Prutsch; Elisabeth Gurnhofer; Tobias Suske; Huan Chang Liang; Michaela Schlederer; Simone Roos; Lawren C. Wu; Ingrid Simonitsch-Klupp; Andrea Alvarez-Hernandez; Christoph Kornauth; Dario A. Leone; Jasmin Svinka; Robert Eferl; Tanja Limberger; Astrid Aufinger; Nitesh Shirsath; Peter Wolf; Thomas Hielscher; Fritz Aberger; Johannes Schmoellerl; Dagmar Stoiber; Birgit Strobl; Ulrich Jäger; Philipp B. Staber; Florian Grebien; Richard Moriggl; Mathias Müller; Giorgio Inghirami; Takaomi Sanda; A. Thomas Look

TYK2 is a member of the JAK family of tyrosine kinases that is involved in chromosomal translocation-induced fusion proteins found in anaplastic large cell lymphomas (ALCL) that lack rearrangements activating the anaplastic lymphoma kinase (ALK). Here we demonstrate that TYK2 is highly expressed in all cases of human ALCL, and that in a mouse model of NPM-ALK-induced lymphoma, genetic disruption of Tyk2 delays the onset of tumors and prolongs survival of the mice. Lymphomas in this model lacking Tyk2 have reduced STAT1 and STAT3 phosphorylation and reduced expression of Mcl1, a pro-survival member of the BCL2 family. These findings in mice are mirrored in human ALCL cell lines, in which TYK2 is activated by autocrine production of IL-10 and IL-22 and by interaction with specific receptors expressed by the cells. Activated TYK2 leads to STAT1 and STAT3 phosphorylation, activated expression of MCL1 and aberrant ALCL cell survival. Moreover, TYK2 inhibitors are able to induce apoptosis in ALCL cells, regardless of the presence or absence of an ALK-fusion. Thus, TYK2 is a dependency that is required for ALCL cell survival through activation of MCL1 expression. TYK2 represents an attractive drug target due to its essential enzymatic domain, and TYK2-specific inhibitors show promise as novel targeted inhibitors for ALCL.


Cancer Research | 2014

Abstract 5349: The role of growth hormone receptor in liver fibrosis and cancer

Patricia Stiedl; Leander Blaas; Viktoria Stanek; Jasmin Svinka; Robert M. Mahon; Gernot Zollner; Thierry Claudel; Mathias Mueller; Wolfgang Mikulits; Harald Esterbauer; Robert Eferl; Johannes Haybaeck; Michael Trauner; Emilio Casanova

Liver fibrosis is defined as the pathological excessive deposition of extracellular matrix proteins which contributes to a disruption of normal liver architecture. Fibrosis can progress to cirrhosis, which can eventually result in hepatocellular carcinoma. Over the past decades our understanding of the cellular and molecular mechanisms of liver fibrosis has advanced greatly. Activated hepatic stellate cells, myofibroblasts of bone marrow origin and portal fibroblasts have all been identified as major contributors to fibrosis. Recently, growth hormone resistance and low serum levels of IGF-1 have also been associated with liver cirrhosis in humans, indicating a role for growth hormone receptor, which itself controls various cellular functions including the transcription of IGF-1 through Stat 5 signaling. Furthermore, supplementation of recombinant IGF-1 in cirrhotic animal models was shown to reduce disease severity. In order to elucidate whether the GHR plays an active role in the establishment of fibrotic liver diseases or rather represents a major consequence of this illness, we crossed mice lacking the Ghr/bp gene (Ghr -/-) with Mdr2 knockout mice (Mdr2-/-), a mouse model of inflammatory cholestasis and liver fibrosis. Our results show that Ghr-/-;Mdr2-/- mice display deregulated bile acid homeostasis and increased serum markers associated with inflammation and fibrosis. Bile duct proliferation and extensive collagen deposition were also observed in Ghr-/-;Mdr2-/- relative to Mdr2-/- mice. Additionally, compared to control mice a pronounced down regulation of the hepato-protective genes Hnf6, Egfr and Igf-1 was seen in Ghr-/-;Mdr2-/- animals, which also displayed significantly increased apoptosis. Moreover, single knockout mice (Ghr-/-) developed bile infacts when fed with 1% cholic acid compared to their wildtype littermates, indicating that loss of GHR renders hepatocytes more susceptible to toxic bile acid accumulation. Surprisingly and despite their severe fibrotic phenotype Ghr -/-; Mdr2 -/- mice showed a significant decrease in tumor incidence compared to Mdr2 -/- mice, indicating that loss of GHR signaling may slow the progression from fibrosis/cirrhosis to cancer in the liver. Our findings suggest that loss of GHR signaling results in increased liver injury in the Mdr2 -/- mouse model of inflammatory cholestasis and liver fibrosis, signifying the possible therapeutic value of this pathway in the development of liver fibrosis treatments. Citation Format: Patricia Stiedl, Leander Blaas, Viktoria Stanek, Jasmin Svinka, Robert Mc Mahon, Gernot Zollner, Thierry Claudel, Mathias Mueller, Wolfgang Mikulits, Harald Esterbauer, Robert Eferl, Johannes Haybaeck, Michael Trauner, Emilio Casanova. The role of growth hormone receptor in liver fibrosis and cancer. [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 5349. doi:10.1158/1538-7445.AM2014-5349


Nature Methods | 2012

A mouse model to identify cooperating signaling pathways in cancer

Monica Musteanu; Leander Blaas; Rainer Zenz; Jasmin Svinka; Thomas Hoffmann; Beatrice Grabner; Daniel Schramek; Hans-Peter Kantner; Mathias Müller; Thomas Kolbe; Thomas Rülicke; Richard Moriggl; Lukas Kenner; Dagmar Stoiber; Josef M. Penninger; Helmut Popper; Emilio Casanova; Robert Eferl

Collaboration


Dive into the Jasmin Svinka's collaboration.

Top Co-Authors

Avatar

Robert Eferl

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Emilio Casanova

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Mathias Müller

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Harald Esterbauer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Mikulits

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beatrice Grabner

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Lukas Kenner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Michael Trauner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Thierry Claudel

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge