Network


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

Hotspot


Dive into the research topics where Veronika Sexl is active.

Publication


Featured researches published by Veronika Sexl.


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).


Nature Chemical Biology | 2012

BCR-ABL uncouples canonical JAK2-STAT5 signaling in chronic myeloid leukemia

Oliver Hantschel; Wolfgang Warsch; Eva Eckelhart; Ines Kaupe; Florian Grebien; Kay Uwe Wagner; Giulio Superti-Furga; Veronika Sexl

Constitutive activation of STAT5 is critical for the maintenance of chronic myeloid leukemia (CML) characterized by the BCR-ABL oncoprotein. Tyrosine kinase inhibitors (TKIs) for the STAT5-activating kinase JAK2 have been discussed as a treatment option for CML patients. Using murine leukemia models combined with inducible ablation of JAK2, we show JAK2 dependence for initial lymphoid transformation, which is lost once leukemia is established. In contrast, initial myeloid transformation and leukemia maintenance were independent of JAK2. Nevertheless, several JAK2 TKIs induced apoptosis in BCR-ABL(+) cells irrespective of the presence of JAK2. This is caused by the previously unknown direct off-target inhibition of BCR-ABL. Cellular and enzymatic analyses suggest that BCR-ABL phosphorylates STAT5 directly. Our findings suggest uncoupling of the canonical JAK2-STAT5 module upon BCR-ABL expression, thereby making JAK2 targeting dispensable. Thus, attempts to pharmacologically target STAT5 in BCR-ABL(+) diseases need to focus on STAT5 itself.


Blood | 2011

A novel Ncr1-Cre mouse reveals the essential role of STAT5 for NK-cell survival and development.

Eva Eckelhart; Wolfgang Warsch; Eva Zebedin; Olivia Simma; Dagmar Stoiber; Thomas Kolbe; Thomas Rülicke; Mathias Mueller; Emilio Casanova; Veronika Sexl

We generated a transgenic mouse line that expresses the Cre recombinase under the control of the Ncr1 (p46) promoter. Cre-mediated recombination was tightly restricted to natural killer (NK) cells, as revealed by crossing Ncr1-iCreTg mice to the eGFP-LSLTg reporter strain. Ncr1-iCreTg mice were further used to study NK cell-specific functions of Stat5 (signal transducers and activators of transcription 5) by generating Stat5(f/f) Ncr1-iCreTg animals. Stat5(f/f) Ncr1-iCreTg mice were largely devoid of NK cells in peripheral lymphoid organs. In the bone marrow, NK-cell maturation was abrogated at the NK cell-precursor stage. Moreover, we found that in vitro deletion of Stat5 in interleukin 2-expanded NK cells was incompatible with NK-cell viability. In vivo assays confirmed the complete abrogation of NK cell-mediated tumor control against B16F10-melanoma cells. In contrast, T cell-mediated tumor surveillance against MC38-adenocarcinoma cells was undisturbed. In summary, the results of our study show that STAT5 has a cell-intrinsic role in NK-cell development and that Ncr1-iCreTg mice are a powerful novel tool with which to study NK-cell development, biology, and function.


Journal of Clinical Investigation | 2011

Lipoxygenase mediates invasion of intrametastatic lymphatic vessels and propagates lymph node metastasis of human mammary carcinoma xenografts in mouse

Dontscho Kerjaschki; Zsuzsanna Bago-Horvath; Margaretha Rudas; Veronika Sexl; Christine Schneckenleithner; Susanne Wolbank; Gregor Bartel; Sigurd Krieger; Romana Kalt; Brigitte Hantusch; Thomas Keller; Katalin Nagy-Bojarszky; Nicole Huttary; Ingrid Raab; Karin Lackner; Katharina Krautgasser; Helga Schachner; Klaus Kaserer; Sandra Rezar; Sybille Madlener; Caroline Vonach; Agnes Davidovits; Hitonari Nosaka; Monika Hämmerle; Katharina Viola; Helmut Dolznig; Martin Schreiber; Alexander Nader; Wolfgang Mikulits; Michael Gnant

In individuals with mammary carcinoma, the most relevant prognostic predictor of distant organ metastasis and clinical outcome is the status of axillary lymph node metastasis. Metastases form initially in axillary sentinel lymph nodes and progress via connecting lymphatic vessels into postsentinel lymph nodes. However, the mechanisms of consecutive lymph node colonization are unknown. Through the analysis of human mammary carcinomas and their matching axillary lymph nodes, we show here that intrametastatic lymphatic vessels and bulk tumor cell invasion into these vessels highly correlate with formation of postsentinel metastasis. In an in vitro model of tumor bulk invasion, human mammary carcinoma cells caused circular defects in lymphatic endothelial monolayers. These circular defects were highly reminiscent of defects of the lymphovascular walls at sites of tumor invasion in vivo and were primarily generated by the tumor-derived arachidonic acid metabolite 12S-HETE following 15-lipoxygenase-1 (ALOX15) catalysis. Accordingly, pharmacological inhibition and shRNA knockdown of ALOX15 each repressed formation of circular defects in vitro. Importantly, ALOX15 knockdown antagonized formation of lymph node metastasis in xenografted tumors. Furthermore, expression of lipoxygenase in human sentinel lymph node metastases correlated inversely with metastasis-free survival. These results provide evidence that lipoxygenase serves as a mediator of tumor cell invasion into lymphatic vessels and formation of lymph node metastasis in ductal mammary carcinomas.


Cancer Cell | 2013

A Kinase-Independent Function of CDK6 Links the Cell Cycle to Tumor Angiogenesis.

Karoline Kollmann; Gerwin Heller; Christine Schneckenleithner; Wolfgang Warsch; Ruth Scheicher; Rene G. Ott; Markus Schäfer; Sabine Fajmann; Michaela Schlederer; Ana-Iris Schiefer; Ursula Reichart; Matthias Mayerhofer; Christoph Hoeller; Sabine Zöchbauer-Müller; Dontscho Kerjaschki; Christoph Bock; Lukas Kenner; Gerald Hoefler; Michael Freissmuth; Anthony R. Green; Richard Moriggl; Meinrad Busslinger; Marcos Malumbres; Veronika Sexl

Summary In contrast to its close homolog CDK4, the cell cycle kinase CDK6 is expressed at high levels in lymphoid malignancies. In a model for p185BCR-ABL+ B-acute lymphoid leukemia, we show that CDK6 is part of a transcription complex that induces the expression of the tumor suppressor p16INK4a and the pro-angiogenic factor VEGF-A. This function is independent of CDK6’s kinase activity. High CDK6 expression thus suppresses proliferation by upregulating p16INK4a, providing an internal safeguard. However, in the absence of p16INK4a, CDK6 can exert its full tumor-promoting function by enhancing proliferation and stimulating angiogenesis. The finding that CDK6 connects cell-cycle progression to angiogenesis confirms CDK6’s central role in hematopoietic malignancies and could underlie the selection pressure to upregulate CDK6 and silence p16INK4a.


Journal of Immunology | 2008

Unique Effects of KIT D816V in BaF3 Cells: Induction of Cluster Formation, Histamine Synthesis, and Early Mast Cell Differentiation Antigens

Matthias Mayerhofer; Karoline V. Gleixner; Andrea Hoelbl; Stefan Florian; Gregor Hoermann; Karl J. Aichberger; Martin Bilban; Harald Esterbauer; Maria-Theresa Krauth; Wolfgang R. Sperr; Jack B. Longley; Robert Kralovics; Richard Moriggl; Jacques Zappulla; Roland S. Liblau; Ilse Schwarzinger; Veronika Sexl; Christian Sillaber; Peter Valent

Oncogenic tyrosine kinases (TK) usually convert growth factor-dependent cells to factor independence with autonomous proliferation. However, TK-driven neoplasms often are indolent and characterized by cell differentiation rather than proliferation. A prototype of an indolent TK-driven neoplasm is indolent systemic mastocytosis. We found that the D816V-mutated variant of KIT, a TK detectable in most patients with systemic mastocytosis, induces cluster formation and expression of several mast cell differentiation and adhesion Ags, including microphthalmia transcription factor, IL-4 receptor, histamine, CD63, and ICAM-1 in IL-3-dependent BaF3 cells. By contrast, wild-type KIT did not induce cluster formation or mast cell differentiation Ags. Additionally, KIT D816V, but not wild-type KIT, induced STAT5 activation in BaF3 cells. However, despite these intriguing effects, KIT D816V did not convert BaF3 cells to factor-independent proliferation. Correspondingly, BaF3 cells with conditional expression of KIT D816V did not form tumors in nude mice. Together, the biologic effects of KIT D816V in BaF3 cells match strikingly with the clinical course of indolent systemic mastocytosis and with our recently established transgenic mouse model, in which KIT D816V induces indolent mast cell accumulations but usually does not induce a malignant mast cell disease. Based on all these results, it is hypothesized that KIT D816V as a single hit may be sufficient to cause indolent systemic mastocytosis, whereas additional defects may be required to induce aggressive mast cell disorders.


Blood | 2015

CDK6 as a key regulator of hematopoietic and leukemic stem cell activation.

Ruth Scheicher; Andrea Hoelbl-Kovacic; Florian Bellutti; Anca-Sarmiza Tigan; Michaela Prchal-Murphy; Gerwin Heller; Christine Schneckenleithner; María Salazar-Roa; Sabine Zöchbauer-Müller; Johannes Zuber; Marcos Malumbres; Karoline Kollmann; Veronika Sexl

The cyclin-dependent kinase 6 (CDK6) and CDK4 have redundant functions in regulating cell-cycle progression. We describe a novel role for CDK6 in hematopoietic and leukemic stem cells (hematopoietic stem cells [HSCs] and leukemic stem cells [LSCs]) that exceeds its function as a cell-cycle regulator. Although hematopoiesis appears normal under steady-state conditions, Cdk6(-/-) HSCs do not efficiently repopulate upon competitive transplantation, and Cdk6-deficient mice are significantly more susceptible to 5-fluorouracil treatment. We find that activation of HSCs requires CDK6, which interferes with the transcription of key regulators, including Egr1. Transcriptional profiling of HSCs is consistent with the central role of Egr1. The impaired repopulation capacity extends to BCR-ABL(p210+) LSCs. Transplantation with BCR-ABL(p210+)-infected bone marrow from Cdk6(-/-) mice fails to induce disease, although recipient mice do harbor LSCs. Egr1 knock-down in Cdk6(-/-) BCR-ABL(p210+) LSKs significantly enhances the potential to form colonies, underlining the importance of the CDK6-Egr1 axis. Our findings define CDK6 as an important regulator of stem cell activation and an essential component of a transcriptional complex that suppresses Egr1 in HSCs and LSCs.


Blood | 2008

Leukemic challenge unmasks a requirement for PI3Kδ in NK cell–mediated tumor surveillance

Eva Zebedin; Olivia Simma; Christian Schuster; Eva M Putz; Sabine Fajmann; Wolfgang Warsch; Eva Eckelhart; Dagmar Stoiber; Eva Weisz; Johannes A. Schmid; Winfried F. Pickl; Christian Baumgartner; Peter Valent; Roland P. Piekorz; Michael Freissmuth; Veronika Sexl

Specific inhibitors of PI3K isoforms are currently evaluated for their therapeutic potential in leukemia. We found that BCR/ABL(+) human leukemic cells express PI3Kdelta and therefore explored its impact on leukemia development. Using PI3Kdelta-deficient mice, we define a dual role of PI3Kdelta in leukemia. We observed a growth-promoting effect in tumor cells and an essential function in natural killer (NK) cell-mediated tumor surveillance: Abelson-transformed PI3Kdelta-deficient cells induced leukemia in RAG2-deficient mice with an increased latency, indicating that PI3Kdelta accelerated leukemia progression in vivo. However, the absence of PI3Kdelta also affected NK cell-mediated tumor surveillance. PI3Kdelta-deficient NK cells failed to lyse a large variety of target cells because of defective degranulation, as also documented by capacitance recordings. Accordingly, transplanted leukemic cells killed PI3Kdelta-deficient animals more rapidly. As a net effect, no difference in disease latency in vivo was detected if both leukemic cells and NK cells lack PI3Kdelta. Other tumor models confirmed that PI3Kdelta-deficient mice succumbed more rapidly when challenged with T- or B-lymphoid leukemic or B16 melanoma cells. Thus, the action of PI3Kdelta in the NK compartment is as relevant to survival of the mice as the delayed tumor progression. This dual function must be taken into account when using PI3Kdelta inhibitors as antileukemic agents in clinical trials.


Cancer Discovery | 2016

STAT5 Is a Key Regulator in NK Cells and Acts as a Molecular Switch from Tumor Surveillance to Tumor Promotion

Dagmar Gotthardt; Eva Maria Putz; Eva Grundschober; Michaela Prchal-Murphy; Elisabeth Straka; Petra Kudweis; Gerwin Heller; Zsuzsanna Bago-Horvath; Agnieszka Witalisz-Siepracka; Abbarna A. Cumaraswamy; Patrick T. Gunning; Birgit Strobl; Mathias Müller; Richard Moriggl; Christian Stockmann; Veronika Sexl

UNLABELLEDnNatural killer (NK) cells are tightly regulated by the JAK-STAT signaling pathway and cannot survive in the absence of STAT5. We now report that STAT5-deficient NK cells can be rescued by overexpression of BCL2. Our experiments define STAT5 as a master regulator of NK-cell proliferation and lytic functions. Although NK cells are generally responsible for killing tumor cells, the rescued STAT5-deficient NK cells promote tumor formation by producing enhanced levels of the angiogenic factor VEGFA. The importance of VEGFA produced by NK cells was verified by experiments with a conditional knockout of VEGFA in NK cells. We show that STAT5 normally represses the transcription of VEGFA in NK cells, in both mice and humans. These findings reveal that STAT5-directed therapies may have negative effects: In addition to impairing NK-cell-mediated tumor surveillance, they may even promote tumor growth by enhancing angiogenesis.nnnSIGNIFICANCEnThe importance of the immune system in effective cancer treatment is widely recognized. We show that the new signal interceptors targeting the JAK-STAT5 pathway may have dangerous side effects that must be taken into account in clinical trials: inhibiting JAK-STAT5 has the potential to promote tumor growth by enhancing NK-cell-mediated angiogenesis.


Journal of Pharmacology and Experimental Therapeutics | 2006

Inhibition of Xenograft Tumor Growth and Down-Regulation of ErbB Receptors by an Antibody Directed against Lewis Y Antigen

Hesso Farhan; Christian Schuster; Markus Klinger; Eva Weisz; Guenter Waxenecker; Manfred Schuster; Veronika Sexl; Geert C. Mudde; Michael Freissmuth; Ralf Kircheis

The blood group-related Lewis Y antigen is expressed on the majority of human cancers of epithelial origin with only limited expression on normal tissue. Therefore, the Lewis Y antigen represents an interesting candidate for antibody-based treatment strategies. Previous experiments showed that the humanized Lewis Y-specific monoclonal antibody, IGN311, reduced ErbB-receptor-mediated stimulation of mitogen-activated protein kinase by altering receptor recycling. Here, we tested whether binding of IGN311 to growth factor receptors is relevant also to inhibition of tumor growth in vivo. Prolonged incubation with IGN311 of human tumor cell lines, which express high levels of ErbB1 (A431) or ErbB2 (SK-BR-3), resulted in down-regulation of the receptors and inhibition of cell proliferation. IGN311 inhibited the growth of tumors derived from A431 cells xenografted in nude mice. Treatment with IGN311 was associated with a down-regulation of ErbB1 in the excised tumor tissue. Importantly, these effects of IGN311 were also mimicked by the Fab fragment of IGN311. These data indicate that tumor cell growth inhibition by IGN311 cannot solely be accounted for by invoking cellular and humoral immunological mechanisms. A direct effect on signaling via binding to Lewis Y glycosylated growth factor receptors on tumor cells is also likely to contribute to the therapeutic effect of IGN311 in vivo.

Collaboration


Dive into the Veronika Sexl's collaboration.

Top Co-Authors

Avatar

Christian Schuster

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

Michael Freissmuth

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

Peter Valent

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Warsch

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Gregor Hoermann

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Hartmut Beug

Research Institute of Molecular Pathology

View shared research outputs
Top Co-Authors

Avatar

Boris Kovacic

Research Institute of Molecular Pathology

View shared research outputs
Top Co-Authors

Avatar

Eva Eckelhart

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge