Network


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

Hotspot


Dive into the research topics where Robert Eferl is active.

Publication


Featured researches published by Robert Eferl.


Arthritis Research & Therapy | 2007

Activator protein 1 (Fos/Jun) functions in inflammatory bone and skin disease

Rainer Zenz; Robert Eferl; Clemens Scheinecker; Kurt Redlich; Josef S Smolen; Helia B. Schonthaler; Lukas Kenner; Erwin Tschachler; Erwin F. Wagner

Activator protein 1 (AP-1) (Fos/Jun) is a transcriptional regulator composed of members of the Fos and Jun families of DNA binding proteins. The functions of AP-1 were initially studied in mouse development as well as in the whole organism through conventional transgenic approaches, but also by gene targeting using knockout strategies. The importance of AP-1 proteins in disease pathways including the inflammatory response became fully apparent through conditional mutagenesis in mice, in particular when employing gene inactivation in a tissue-specific and inducible fashion. Besides the well-documented roles of Fos and Jun proteins in oncogenesis, where these genes can function both as tumor promoters or tumor suppressors, AP-1 proteins are being recognized as regulators of bone and immune cells, a research area termed osteoimmunology. In the present article, we review recent data regarding the functions of AP-1 as a regulator of cytokine expression and an important modulator in inflammatory diseases such as rheumatoid arthritis, psoriasis and psoriatic arthritis. These new data provide a better molecular understanding of disease pathways and should pave the road for the discovery of new targets for therapeutic applications.


PLOS ONE | 2012

NADPH oxidase NOX4 mediates stellate cell activation and hepatocyte cell death during liver fibrosis development.

Patricia Sancho; Jèssica Mainez; Eva Crosas-Molist; Cesar Roncero; Conrado M. Fernández-Rodríguez; Fernando Pinedo; Heidemarie Huber; Robert Eferl; Wolfgang Mikulits; Isabel Fabregat

A role for the NADPH oxidases NOX1 and NOX2 in liver fibrosis has been proposed, but the implication of NOX4 is poorly understood yet. The aim of this work was to study the functional role of NOX4 in different cell populations implicated in liver fibrosis: hepatic stellate cells (HSC), myofibroblats (MFBs) and hepatocytes. Two different mice models that develop spontaneous fibrosis (Mdr2−/−/p19ARF−/−, Stat3Δhc/Mdr2−/−) and a model of experimental induced fibrosis (CCl4) were used. In addition, gene expression in biopsies from chronic hepatitis C virus (HCV) patients or non-fibrotic liver samples was analyzed. Results have indicated that NOX4 expression was increased in the livers of all animal models, concomitantly with fibrosis development and TGF-β pathway activation. In vitro TGF-β-treated HSC increased NOX4 expression correlating with transdifferentiation to MFBs. Knockdown experiments revealed that NOX4 downstream TGF-β is necessary for HSC activation as well as for the maintenance of the MFB phenotype. NOX4 was not necessary for TGF-β-induced epithelial-mesenchymal transition (EMT), but was required for TGF-β-induced apoptosis in hepatocytes. Finally, NOX4 expression was elevated in patients with hepatitis C virus (HCV)-derived fibrosis, increasing along the fibrosis degree. In summary, fibrosis progression both in vitro and in vivo (animal models and patients) is accompanied by increased NOX4 expression, which mediates acquisition and maintenance of the MFB phenotype, as well as TGF-β-induced death of hepatocytes.


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 | 2012

Antagonistic effects of selenium and lipid peroxides on growth control in early hepatocellular carcinoma.

Nataliya Rohr-Udilova; Wolfgang Sieghart; Robert Eferl; Dagmar Stoiber; Linda Björkhem-Bergman; Lennart C. Eriksson; Klaus Stolze; Hubert Hayden; Bernhard K. Keppler; Sandra Sagmeister; Bettina Grasl-Kraupp; Rolf Schulte-Hermann; Markus Peck-Radosavljevic

Activation of the activator protein 1 (AP‐1) transcription factor as well as increased serum levels of vascular endothelial growth factor (VEGF) and interleukin (IL)‐8 predict poor prognosis of patients with hepatocellular carcinomas (HCCs). Moreover, HCC patients display reduced selenium levels, which may cause lipid peroxidation and oxidative stress because selenium is an essential component of antioxidative glutathione peroxidases (GPx). We hypothesized that selenium‐lipid peroxide antagonism controls the above prognostic markers and tumor growth. (1) In human HCC cell lines (HCC‐1.2, HCC‐3, and SNU398) linoleic acid peroxide (LOOH) and other prooxidants enhanced the expression of VEGF and IL‐8. LOOH up‐regulated AP‐1 activation. Selenium inhibited these effects. This inhibition was mediated by glutathione peroxidase 4 (GPx4), which preferentially degrades lipid peroxides. Selenium enhanced GPx4 expression and total GPx activity, while knock‐down of GPx4 by small interfering RNA (siRNA) increased VEGF, and IL‐8 expression. (2) These results were confirmed in a rat hepatocarcinogenesis model. Selenium treatment during tumor promotion increased hepatic GPx4 expression and reduced the expression of VEGF and of the AP‐1 component c‐fos as well as nodule growth. (3) In HCC patients, increased levels of LOOH‐related antibodies (LOOH‐Ab) were found, suggesting enhanced LOOH formation. LOOH‐Ab correlated with serum VEGF and IL‐8 and with AP‐1 activation in HCC tissue. In contrast, selenium inversely correlated with VEGF, IL‐8, and HCC size (the latter only for tumors smaller than 3 cm). Conclusion: Reduced selenium levels result in accumulation of lipid peroxides. This leads to enhanced AP‐1 activation and consequently to elevated expression of VEGF and IL‐8, which accelerate the growth of HCC. Selenium supplementation could be considered for investigation as a strategy for chemoprevention or additional therapy of early HCC in patients with low selenium levels. (HEPATOLOGY 2012)


American Journal of Hematology | 2014

Iron deficiency alters megakaryopoiesis and platelet phenotype independent of thrombopoietin

Rayko Evstatiev; Adam Bukaty; Kristine Jimenez; Stefanie Kulnigg-Dabsch; Lidia Surman; Werner Schmid; Robert Eferl; Kathrin Lippert; Barbara Scheiber-Mojdehkar; Hans Michael Kvasnicka; Vineeta Khare; Christoph Gasche

Iron deficiency is a common cause of reactive thrombocytosis, however, the exact pathways have not been revealed. Here we aimed to study the mechanisms behind iron deficiency‐induced thrombocytosis. Within few weeks, iron‐depleted diet caused iron deficiency in young Sprague–Dawley rats, as reflected by a drop in hemoglobin, mean corpuscular volume, hepatic iron content and hepcidin mRNA in the liver. Thrombocytosis established in parallel. Moreover, platelets produced in iron deficient animals displayed a higher mean platelet volume and increased aggregation. Bone marrow studies revealed subtle alterations that are suggestive of expansion of megakaryocyte progenitors, an increase in megakaryocyte ploidy and accelerated megakaryocyte differentiation. Iron deficiency did not alter the production of hematopoietic growth factors such as thrombopoietin, interleukin 6 or interleukin 11. Megakaryocytic cell lines grown in iron‐depleted conditions exhibited reduced proliferation but increased ploidy and cell size. Our data suggest that iron deficiency increases megakaryopoietic differentiation and alters platelet phenotype without changes in megakaryocyte growth factors, specifically TPO. Iron deficiency‐induced thrombocytosis may have evolved to maintain or increase the coagulation capacity in conditions with chronic bleeding. Am. J. Hematol. 89:524–529, 2014.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Acquisition of an immunosuppressive protumorigenic macrophage phenotype depending on c-Jun phosphorylation

Simona Hefetz-Sela; Ilan Stein; Yair Klieger; Rinnat M. Porat; Moshe Sade-Feldman; Farid Zreik; Arnon Nagler; Orit Pappo; Luca Quagliata; Eva Dazert; Robert Eferl; Luigi Terracciano; Erwin F. Wagner; Yinon Ben-Neriah; Michal Baniyash; Eli Pikarsky

Significance Tumor-associated macrophages are often associated with poor prognosis, but the molecular cues that impart the change in macrophage phenotypes are mostly unclear. Our findings identify c-Jun N-terminal phosphorylation as a key mediator of macrophage education and point to a recruitment of immunosuppressive regulatory T cells as a possible effector protumorigenic mechanism. This cross-talk between adaptive and innate immune responses occurs before overt tumorigenesis, reversing the classic paradigm of initiation and promotion. Our findings raise the possibility that targeting immune checkpoints could be effective for tumor prevention in chronic inflammatory diseases. The inflamed tumor microenvironment plays a critical role in tumorigenesis. However, the mechanisms through which immune cells, particularly macrophages, promote tumorigenesis have only been partially elucidated, and the full scope of signaling pathways supplying macrophages with protumorigenic phenotypes still remain largely unknown. Here we report that germ-line absence of c-Jun N-terminal phosphorylation at serines 63 and 73 impedes inflammation-associated hepatocarcinogenesis, yet deleting c-Jun only in hepatocytes does not inhibit hepatocellular carcinoma (HCC) formation. Moreover, in human HCC-bearing livers, c-Jun phosphorylation is found in inflammatory cells, whereas it is mostly absent from malignant hepatocytes. Interestingly, macrophages in livers of mice with chronic hepatitis gradually switch their phenotype along the course of disease. Macrophage phenotype and density are dictated by c-Jun phosphorylation, in vitro and in vivo. Transition of macrophage phenotype, from antitumorigenic to protumorigenic, occurs before tumorigenesis, resulting in the production of various chemokines, including chemokine (C-C motif) ligand 17 (CCL17) and CCL22. Such signals, emanating from the liver microenvironment, direct the recruitment of regulatory T cells, which are known to facilitate HCC growth. Our findings identify c-Jun phosphorylation as a key mediator of macrophage education and point to the recruitment of immunosuppressive regulatory T cells as a possible protumorigenic mechanism.


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)


Gastroenterology | 2017

EGFR in Tumor-Associated Myeloid Cells Promotes Development of Colorectal Cancer in Mice and Associates With Outcomes of Patients

Sriram Srivatsa; Mariel C. Paul; C. Cardone; Martin Holcmann; Nicole Amberg; Paulina Pathria; Michaela A. Diamanti; Markus Linder; Gerald Timelthaler; Hans Peter Dienes; Lukas Kenner; Fritz Wrba; Gerald W. Prager; Stefan Rose-John; Robert Eferl; Giuseppina Liguori; Gerardo Botti; Erika Martinelli; Florian R. Greten; Fortunato Ciardiello; Maria Sibilia

Background & Aims Inhibitors of the epidermal growth factor receptor (EGFR) are the first-line therapy for patients with metastatic colorectal tumors without RAS mutations. However, EGFR inhibitors are ineffective in these patients, and tumor level of EGFR does not associate with response to therapy. We screened human colorectal tumors for EGFR-positive myeloid cells and investigated their association with patient outcome. We also performed studies in mice to evaluate how EGFR expression in tumor cells and myeloid cells contributes to development of colitis-associated cancer and ApcMin-dependent intestinal tumorigenesis. Methods We performed immunohistochemical and immunofluorescent analyses of 116 colorectal tumor biopsies to determine levels of EGFR in tumor and stroma; we also collected information on tumor stage and patient features and outcomes. We used the Mann-Whitney U and Kruskal-Wallis tests to correlate tumor levels of EGFR with tumor stage, and the Kaplan-Meier method to estimate patients’ median survival time. We performed experiments in mice lacking EGFR in intestinal epithelial cells (Villin-Cre; Egfrf/f and Villin-CreERT2; Egfrf/f mice) or myeloid cells (LysM-Cre; Egfrf/f mice) on a mixed background. These mice were bred with ApcMin/+ mice; colitis-associated cancer and colitis were induced by administration of dextran sodium sulfate (DSS), with or without azoxymethane (AOM), respectively. Villin-CreERT2 was activated in developed tumors by administration of tamoxifen to mice. Littermates that expressed full-length EGFR were used as controls. Intestinal tissues were collected; severity of colitis, numbers and size of tumors, and intestinal barrier integrity were assessed by histologic, immunohistochemical, quantitative reverse transcription polymerase chain reaction, and flow cytometry analyses. Results We detected EGFR in myeloid cells in the stroma of human colorectal tumors; myeloid cell expression of EGFR associated with tumor metastasis and shorter patient survival time. Mice with deletion of EGFR from myeloid cells formed significantly fewer and smaller tumors than the respective EGFR-expressing controls in an ApcMin/+ background as well as after administration of AOM and DSS. Deletion of EGFR from intestinal epithelial cells did not affect tumor growth. Furthermore, tamoxifen-induced deletion of EGFR from epithelial cells of established intestinal tumors in mice given AOM and DSS did not reduce tumor size. EGFR signaling in myeloid cells promoted activation of STAT3 and expression of survivin in intestinal tumor cells. Mice with deletion of EGFR from myeloid cells developed more severe colitis after DSS administration, characterized by increased intestinal inflammation and intestinal barrier disruption, than control mice or mice with deletion of EGFR from intestinal epithelial cells. EGFR-deficient myeloid cells in the colon of DSS-treated LysM-Cre; Egfrf/f mice had reduced expression of interleukin 6 (IL6), and epithelial STAT3 activation was reduced compared with controls. Administration of recombinant IL6 to LysM-Cre; Egfrf/f mice given DSS protected them from weight loss and restored epithelial proliferation and STAT3 activation, compared with administration of DSS alone to these mice. Conclusions Increased expression of EGFR in myeloid cells from the colorectal tumor stroma associates with tumor progression and reduced survival time of patients with metastatic colorectal cancer. Deletion of EGFR from myeloid cells, but not intestinal epithelial cells, protects mice from colitis-induced intestinal cancer and ApcMin-dependent intestinal tumorigenesis. Myeloid cell expression of EGFR increases activation of STAT3 and expression of survivin in intestinal epithelial cells and expression of IL6 in colon tissues. These findings indicate that expression of EGFR by myeloid cells of the colorectal tumor stroma, rather than the cancer cells themselves, contributes to tumor development.


PLOS ONE | 2014

Inducible, Dose-Adjustable and Time-Restricted Reconstitution of Stat1 Deficiency In Vivo

Nicole R. Leitner; Caroline Lassnig; Rita Rom; Susanne Heider; Zsuzsanna Bago-Horvath; Robert Eferl; Simone Müller; Thomas Kolbe; Lukas Kenner; Thomas Rülicke; Birgit Strobl; Mathias Müller

Signal transducer and activator of transcription (STAT) 1 is a key player in interferon (IFN) signaling, essential in mediating host defense against viruses and other pathogens. STAT1 levels are tightly regulated and loss- or gain-of-function mutations in mice and men lead to severe diseases. We have generated a doxycycline (dox) -inducible, FLAG-tagged Stat1 expression system in mice lacking endogenous STAT1 (i.e. Stat1ind mice). We show that STAT1 expression depends on the time and dose of dox treatment in primary cells and a variety of organs isolated from Stat1ind mice. In bone marrow-derived macrophages, a fraction of the amount of STAT1 present in WT cells is sufficient for full expression of IFN-induced genes. Dox-induced STAT1 established protection against virus infections in primary cells and mice. The availability of the Stat1ind mouse model will enable an examination of the consequences of variable amounts of STAT1. The model will also permit the study of STAT1 dose-dependent and reversible functions as well as of STAT1s contributions to the development, progression and resolution of disease.


Scientific Reports | 2018

Deviations of the immune cell landscape between healthy liver and hepatocellular carcinoma

Nataliya Rohr-Udilova; Florian Klinglmüller; Rolf Schulte-Hermann; Judith Stift; Merima Herac; Martina Salzmann; Francesca Finotello; Gerald Timelthaler; Georg Oberhuber; Matthias Pinter; Thomas Reiberger; Erika Jensen-Jarolim; Robert Eferl; Michael Trauner

Tumor-infiltrating immune cells are highly relevant for prognosis and identification of immunotherapy targets in hepatocellular carcinoma (HCC). The recently developed CIBERSORT method allows immune cell profiling by deconvolution of gene expression microarray data. By applying CIBERSORT, we assessed the relative proportions of immune cells in 41 healthy human livers, 305 HCC samples and 82 HCC adjacent tissues. The obtained immune cell profiles provided enumeration and activation status of 22 immune cell subtypes. Mast cells were evaluated by immunohistochemistry in ten HCC patients. Activated mast cells, monocytes and plasma cells were decreased in HCC, while resting mast cells, total and naïve B cells, CD4+ memory resting and CD8+ T cells were increased when compared to healthy livers. Previously described S1, S2 and S3 molecular HCC subclasses demonstrated increased M1-polarized macrophages in the S3 subclass with good prognosis. Strong total immune cell infiltration into HCC correlated with total B cells, memory B cells, T follicular helper cells and M1 macrophages, whereas weak infiltration was linked to resting NK cells, neutrophils and resting mast cells. Immunohistochemical analysis of patient samples confirmed the reduced frequency of mast cells in human HCC tumor tissue as compared to tumor adjacent tissue. Our data demonstrate that deconvolution of gene expression data by CIBERSORT provides valuable information about immune cell composition of HCC patients.

Collaboration


Dive into the Robert Eferl's collaboration.

Top Co-Authors

Avatar

Emilio Casanova

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Jasmin Svinka

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

Dagmar Stoiber

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Harald Esterbauer

Medical University of 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

Richard Moriggl

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Mikulits

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge