Network


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

Hotspot


Dive into the research topics where Boris Kovacic is active.

Publication


Featured researches published by Boris Kovacic.


Embo Molecular Medicine | 2010

Stat5 is indispensable for the maintenance of bcr/abl-positive leukaemia

Andrea Hoelbl; Christian Schuster; Boris Kovacic; Bing-Mei Zhu; Mark C. Wickre; Maria A. Hoelzl; Sabine Fajmann; Florian Grebien; Wolfgang Warsch; Gabriele Stengl; Lothar Hennighausen; Valeria Poli; Hartmut Beug; Richard Moriggl; Veronika Sexl

Tumourigenesis caused by the Bcr/Abl oncoprotein is a multi‐step process proceeding from initial to tumour‐maintaining events and finally results in a complex tumour‐supporting network. A key to successful cancer therapy is the identification of critical functional nodes in an oncogenic network required for disease maintenance. So far, the transcription factors Stat3 and Stat5a/b have been implicated in bcr/abl‐induced initial transformation. However, to qualify as a potential drug target, a signalling pathway must be required for the maintenance of the leukaemic state. Data on the roles of Stat3 or Stat5a/b in leukaemia maintenance are elusive. Here, we show that both, Stat3 and Stat5 are necessary for initial transformation. However, Stat5‐ but not Stat3‐deletion induces G0/G1 cell cycle arrest and apoptosis of imatinib‐sensitive and imatinib‐resistant stable leukaemic cells in vitro. Accordingly, Stat5‐abrogation led to effective elimination of myeloid and lymphoid leukaemia maintenance in vivo. Hence, we identified Stat5 as a vulnerable point in the oncogenic network downstream of Bcr/Abl representing a case of non‐oncogene addiction (NOA).


Cell | 2011

Targeting the SH2-kinase interface in Bcr-Abl inhibits leukemogenesis.

Florian Grebien; Oliver Hantschel; John Wojcik; Ines Kaupe; Boris Kovacic; Arkadiusz M. Wyrzucki; Gerald Gish; Sabine Cerny-Reiterer; Akiko Koide; Hartmut Beug; Tony Pawson; Peter Valent; Shohei Koide; Giulio Superti-Furga

Summary Chronic myelogenous leukemia (CML) is caused by the constitutively active tyrosine kinase Bcr-Abl and treated with the tyrosine kinase inhibitor (TKI) imatinib. However, emerging TKI resistance prevents complete cure. Therefore, alternative strategies targeting regulatory modules of Bcr-Abl in addition to the kinase active site are strongly desirable. Here, we show that an intramolecular interaction between the SH2 and kinase domains in Bcr-Abl is both necessary and sufficient for high catalytic activity of the enzyme. Disruption of this interface led to inhibition of downstream events critical for CML signaling and, importantly, completely abolished leukemia formation in mice. Furthermore, disruption of the SH2-kinase interface increased sensitivity of imatinib-resistant Bcr-Abl mutants to TKI inhibition. An engineered Abl SH2-binding fibronectin type III monobody inhibited Bcr-Abl kinase activity both in vitro and in primary CML cells, where it induced apoptosis. This work validates the SH2-kinase interface as an allosteric target for therapeutic intervention. PaperFlick


Blood | 2008

Stat5 regulates cellular iron uptake of erythroid cells via IRP-2 and TfR-1

Marc Kerenyi; Florian Grebien; Helmuth Gehart; Manfred Schifrer; Matthias Artaker; Boris Kovacic; Hartmut Beug; Richard Moriggl; Ernst W. Müllner

Erythropoiesis strictly depends on signal transduction through the erythropoietin receptor (EpoR)-Janus kinase 2 (Jak2)-signal transducer and activator of transcription 5 (Stat5) axis, regulating proliferation, differentiation, and survival. The exact role of the transcription factor Stat5 in erythropoiesis remained puzzling, however, since the first Stat5-deficient mice carried a hypomorphic Stat5 allele, impeding full phenotypical analysis. Using mice completely lacking Stat5--displaying early lethality--we demonstrate that these animals suffer from microcytic anemia due to reduced expression of the antiapoptotic proteins Bcl-x(L) and Mcl-1 followed by enhanced apoptosis. Moreover, transferrin receptor-1 (TfR-1) cell surface levels on erythroid cells were decreased more than 2-fold on erythroid cells of Stat5(-/-) animals. This reduction could be attributed to reduced transcription of TfR-1 mRNA and iron regulatory protein 2 (IRP-2), the major translational regulator of TfR-1 mRNA stability in erythroid cells. Both genes were demonstrated to be direct transcriptional targets of Stat5. This establishes an unexpected mechanistic link between EpoR/Jak/Stat signaling and iron metabolism, processes absolutely essential for erythropoiesis and life.


Blood | 2008

Stat5 activation enables erythropoiesis in the absence of EpoR and Jak2

Florian Grebien; Marc Kerenyi; Boris Kovacic; Thomas Kolbe; Verena Becker; Helmut Dolznig; Klaus Pfeffer; Ursula Klingmüller; Mathias Müller; Hartmut Beug; Ernst W. Müllner; Richard Moriggl

Erythropoiesis requires erythropoietin (Epo) and stem cell factor (SCF) signaling via their receptors EpoR and c-Kit. EpoR, like many other receptors involved in hematopoiesis, acts via the kinase Jak2. Deletion of EpoR or Janus kinase 2 (Jak2) causes embryonic lethality as a result of defective erythropoiesis. The contribution of distinct EpoR/Jak2-induced signaling pathways (mitogen-activated protein kinase, phosphatidylinositol 3-kinase, signal transducer and activator of transcription 5 [Stat5]) to functional erythropoiesis is incompletely understood. Here we demonstrate that expression of a constitutively activated Stat5a mutant (cS5) was sufficient to relieve the proliferation defect of Jak2(-/-) and EpoR(-/-) cells in an Epo-independent manner. In addition, tamoxifen-induced DNA binding of a Stat5a-estrogen receptor (ER)* fusion construct enabled erythropoiesis in the absence of Epo. Furthermore, c-Kit was able to enhance signaling through the Jak2-Stat5 axis, particularly in lymphoid and myeloid progenitors. Although abundance of hematopoietic stem cells was 2.5-fold reduced in Jak2(-/-) fetal livers, transplantation of Jak2(-/-)-cS5 fetal liver cells into irradiated mice gave rise to mature erythroid and myeloid cells of donor origin up to 6 months after transplantation. Cytokine- and c-Kit pathways do not function independently of each other in hematopoiesis but cooperate to attain full Jak2/Stat5 activation. In conclusion, activated Stat5 is a critical downstream effector of Jak2 in erythropoiesis/myelopoiesis, and Jak2 functionally links cytokine- with c-Kit-receptor tyrosine kinase signaling.


Journal of Clinical Investigation | 2004

TYK2 is a key regulator of the surveillance of B lymphoid tumors

Dagmar Stoiber; Boris Kovacic; Christian Schuster; Carola Schellack; Marina Karaghiosoff; Rita Kreibich; Eva Weisz; Michaela Artwohl; Olaf C. Kleine; Mathias Müller; Sabina Baumgartner-Parzer; Jacques Ghysdael; Michael Freissmuth; Veronika Sexl

Aberrant activation of the JAK-STAT pathway has been implicated in tumor formation; for example, constitutive activation of JAK2 kinase or the enforced expression of STAT5 induces leukemia in mice. We show here that the Janus kinase TYK2 serves an opposite function. Mice deficient in TYK2 developed Abelson-induced B lymphoid leukemia/lymphoma as well as TEL-JAK2-induced T lymphoid leukemia with a higher incidence and shortened latency compared with WT controls. The cell-autonomous properties of Abelson murine leukemia virus-transformed (A-MuLV-transformed) TYK2(-/-) cells were unaltered, but the high susceptibility of TYK2(-/-) mice resulted from an impaired tumor surveillance, and accordingly, TYK2(-/-) A-MuLV-induced lymphomas were easily rejected after transplantation into WT hosts. The increased rate of leukemia/lymphoma formation was linked to a decreased in vitro cytotoxic capacity of TYK2(-/-) NK and NKT cells toward tumor-derived cells. RAG2/TYK2 double-knockout mice succumbed to A-MuLV-induced leukemia/lymphoma faster than RAG2(-/-)TYK2(+/-) mice. This defines NK cells as key players in tumor surveillance in Abelson-induced malignancies. Our observations provide compelling evidence that TYK2 is an important regulator of lymphoid tumor surveillance.


Frontiers in Bioscience | 2008

The different functions of Stat5 and chromatin alteration through Stat5 proteins.

Jan-Wilhelm Kornfeld; Florian Grebien; Marc Kerenyi; Katrin Friedbichler; Boris Kovacic; Zankl B; Hoelbl A; Nivarti H; Hartmut Beug; Sexl; Mathias Müller; Lukas Kenner; Ernst W. Müllner; Fabrice Gouilleux; Richard Moriggl

Stat5 proteins modulate gene transcription upon cytokine- and growth factor action. Stat5a and Stat5b proteins alone are weak activators of transcription. They can modify chromatin organization through oligomerization and they act predominantly in co-operation and interaction with other proteins. The conservative view of exclusively nuclear functions of Stat5 was challenged by the observation of additional Stat5 effects in the cytoplasm, resulting in activation of the PI3K-Akt pathway. We summarize biological consequences of mutations in conserved domains of Stat5 or of deletions in the N- or C-terminal domains with impact on target gene transcription. Formation of higher-order oligomers is dramatically changed upon amino- or carboxyterminal deletions in Stat5 proteins. Mutations in or deletion of the Stat5 N-terminus leads to diminished leukemogenic potential of oncogenic Stat5, probably due to the inability to form Stat5 tetramers. The Stat5 N-terminal domain prevents persistent activation and can act as a DNA-docking platform for the glucocorticoid receptor (GR). The corresponding protocols should facilitate follow-up studies on Stat5 proteins and their contribution to normal physiological versus pathological processes through differential chromatin binding.


Nature Immunology | 2017

Chronic signaling via the metabolic checkpoint kinase mTORC1 induces macrophage granuloma formation and marks sarcoidosis progression

Monika Linke; Ha Thi Thanh Pham; Karl Katholnig; Thomas Schnöller; Anne Miller; Florian Demel; Birgit Schütz; Margit Rosner; Boris Kovacic; Nyamdelger Sukhbaatar; Birgit Niederreiter; Stephan Blüml; Peter Kuess; Veronika Sexl; Mathias Müller; Mario Mikula; Wolfram Weckwerth; Arvand Haschemi; Martin Susani; Markus Hengstschläger; Michael J Gambello; Thomas Weichhart

The aggregation of hypertrophic macrophages constitutes the basis of all granulomatous diseases, such as tuberculosis or sarcoidosis, and is decisive for disease pathogenesis. However, macrophage-intrinsic pathways driving granuloma initiation and maintenance remain elusive. We found that activation of the metabolic checkpoint kinase mTORC1 in macrophages by deletion of the gene encoding tuberous sclerosis 2 (Tsc2) was sufficient to induce hypertrophy and proliferation, resulting in excessive granuloma formation in vivo. TSC2-deficient macrophages formed mTORC1-dependent granulomatous structures in vitro and showed constitutive proliferation that was mediated by the neo-expression of cyclin-dependent kinase 4 (CDK4). Moreover, mTORC1 promoted metabolic reprogramming via CDK4 toward increased glycolysis while simultaneously inhibiting NF-κB signaling and apoptosis. Inhibition of mTORC1 induced apoptosis and completely resolved granulomas in myeloid TSC2-deficient mice. In human sarcoidosis patients, mTORC1 activation, macrophage proliferation and glycolysis were identified as hallmarks that correlated with clinical disease progression. Collectively, TSC2 maintains macrophage quiescence and prevents mTORC1-dependent granulomatous disease with clinical implications for sarcoidosis.


Embo Molecular Medicine | 2012

Diverging fates of cells of origin in acute and chronic leukaemia

Boris Kovacic; Andrea Hoelbl; Gabriele Litos; Memetcan Alacakaptan; Christian Schuster; Katrin M. Fischhuber; Marc A. Kerenyi; Gabriele Stengl; Richard Moriggl; Veronika Sexl; Hartmut Beug

The large difference in phenotypes among tumour populations may stem from the stochastic origin of tumours from distinct cells – tumour cells are assumed to retain the phenotypes of the cells from which they derive. Yet, functional studies addressing the cellular origin of leukaemia are lacking. Here we show that the cells of origin of both, BCR/ABL‐induced chronic myeloid (CML) and B‐cell acute lymphoid leukaemia (B‐ALL), resemble long‐term haematopoietic stem cells (LT‐HSCs). During disease‐maintenance, CML LT‐HSCs persist to function as cancer stem cells (CSCs) that maintain leukaemia and require signalling by the transcription factor STAT5. In contrast, B‐ALL LT‐HSCs differentiate into CSCs that correspond to pro‐B cells. This transition step requires a transient IL‐7 signal and is lost in IL‐7Rα‐deficient cells. Thus, in BCR/ABLp185+ B‐ALL and BCR/ABLp210+ CML, the final phenotype of the tumour as well as the abundance of CSCs is dictated by diverging differentiation fates of their common cells of origin.


PLOS ONE | 2012

Authentication of Primordial Characteristics of the CLBL-1 Cell Line Prove the Integrity of a Canine B-Cell Lymphoma in a Murine In Vivo Model

Barbara C. Rütgen; Saskia Willenbrock; Nicola Reimann-Berg; Ingrid Walter; Andrea Fuchs-Baumgartinger; Siegfried Wagner; Boris Kovacic; Sabine E. Essler; Ilse Schwendenwein; Ingo Nolte; Armin Saalmüller; Hugo Murua Escobar

Cell lines are key tools in cancer research allowing the generation of neoplasias in animal models resembling the initial tumours able to mimic the original neoplasias closely in vivo. Canine lymphoma is the major hematopoietic malignancy in dogs and considered as a valuable spontaneous large animal model for human Non-Hodgkins Lymphoma (NHL). Herein we describe the establishment and characterisation of an in vivo model using the canine B-cell lymphoma cell line CLBL-1 analysing the stability of the induced tumours and the ability to resemble the original material. CLBL-1 was injected into Rag2−/−γc −/− mice. The generated tumor material was analysed by immunophenotyping and histopathology and used to establish the cell line CLBL-1M. Both cell lines were karyotyped for detection of chromosomal aberrations. Additionally, CLBL-1 was stimulated with IL-2 and DSP30 as described for primary canine B-cell lymphomas and NHL to examine the stimulatory effect on cell proliferation. CLBL-1 in vivo application resulted in lymphoma-like disease and tumor formation. Immunophenotypic analysis of tumorous material showed expression of CD45+, MHCII+, CD11a+ and CD79αcy+. PARR analysis showed positivity for IgH indicating a monoclonal character. These cytogenetic, molecular, immunophenotypical and histological characterisations of the in vivo model reveal that the induced tumours and thereof generated cell line resemble closely the original material. After DSP30 and IL-2 stimulation, CLBL-1 showed to respond in the same way as primary material. The herein described CLBL-1 in vivo model provides a highly stable tool for B-cell lymphoma research in veterinary and human medicine allowing various further in vivo studies.


Archives of Toxicology | 2016

Full biological characterization of human pluripotent stem cells will open the door to translational research.

Nina Kramer; Margit Rosner; Boris Kovacic; Markus Hengstschläger

Since the discovery of human embryonic stem cells (hESC) and human-induced pluripotent stem cells (hiPSC), great hopes were held for their therapeutic application including disease modeling, drug discovery screenings, toxicological screenings and regenerative therapy. hESC and hiPSC have the advantage of indefinite self-renewal, thereby generating an inexhaustible pool of cells with, e.g., specific genotype for developing putative treatments; they can differentiate into derivatives of all three germ layers enabling autologous transplantation, and via donor-selection they can express various genotypes of interest for better disease modeling. Furthermore, drug screenings and toxicological screenings in hESC and hiPSC are more pertinent to identify drugs or chemical compounds that are harmful for human, than a mouse model could predict. Despite continuing research in the wide field of therapeutic applications, further understanding of the underlying basic mechanisms of stem cell function is necessary. Here, we summarize current knowledge concerning pluripotency, self-renewal, apoptosis, motility, epithelial-to-mesenchymal transition and differentiation of pluripotent stem cells.

Collaboration


Dive into the Boris Kovacic's collaboration.

Top Co-Authors

Avatar

Richard Moriggl

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Hartmut Beug

Research Institute of Molecular Pathology

View shared research outputs
Top Co-Authors

Avatar

Veronika Sexl

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Florian Grebien

Austrian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Mathias Müller

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Christian Schuster

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Marc Kerenyi

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Andrea Hoelbl

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Dagmar Stoiber

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Margit Rosner

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge