Network


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

Hotspot


Dive into the research topics where Nicole R. Leitner is active.

Publication


Featured researches published by Nicole R. Leitner.


PLOS ONE | 2012

TYK2 kinase activity is required for functional type I interferon responses in vivo.

Michaela Prchal-Murphy; Christian Semper; Caroline Lassnig; Barbara Wallner; Christian Gausterer; Ingeborg Teppner-Klymiuk; Julianna Kobolák; Simone Müller; Thomas Kolbe; Marina Karaghiosoff; Andras Dinnyes; Thomas Rülicke; Nicole R. Leitner; Birgit Strobl; Mathias Müller

Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family and is involved in cytokine signalling. In vitro analyses suggest that TYK2 also has kinase-independent, i.e., non-canonical, functions. We have generated gene-targeted mice harbouring a mutation in the ATP-binding pocket of the kinase domain. The Tyk2 kinase-inactive (Tyk2K923E) mice are viable and show no gross abnormalities. We show that kinase-active TYK2 is required for full-fledged type I interferon- (IFN) induced activation of the transcription factors STAT1-4 and for the in vivo antiviral defence against viruses primarily controlled through type I IFN actions. In addition, TYK2 kinase activity was found to be required for the protein’s stability. An inhibitory function was only observed upon over-expression of TYK2K923E in vitro. Tyk2K923E mice represent the first model for studying the kinase-independent function of a JAK in vivo and for assessing the consequences of side effects of JAK inhibitors.


Transgenic Research | 2012

Generation of mice with a conditional Stat1 null allele

Barbara Wallner; Nicole R. Leitner; Raimund M. Vielnascher; Elisabeth Kernbauer; Thomas Kolbe; Marina Karaghiosoff; Thomas Rülicke; Thomas Decker; Mathias Müller

Interferons (IFNs) are key cytokines in the innate immune response that also bridge the gap to adaptive immunity. Signaling upon stimulation by IFN type I, II and III is mediated by the Jak-Stat pathway. STAT1 is activated by all three IFN receptor complexes and absence of STAT1 from mice increases their susceptibility to pathogens. In addition, depending on the setting, STAT1 can act as tumor suppressor or oncogene. Here we report the generation and detailed functional characterization of a conditional Stat1 knockout mouse. We show the integrity of the conditional Stat1 locus and report successful in vivo deletion by means of a ubiquitous and a tissue-specific Cre recombinase. The conditional Stat1 null allele represents an important tool for identifying novel and cell-autonomous STAT1 functions in infection and cancer.


Molecular and Cellular Biology | 2014

STAT1β is not dominant negative and is capable of contributing to gamma interferon-dependent innate immunity.

Christian Semper; Nicole R. Leitner; Caroline Lassnig; Matthias Parrini; Tanel Mahlakõiv; Michael Rammerstorfer; Karin Lorenz; Doris Rigler; Simone Müller; Thomas Kolbe; Claus Vogl; Thomas Rülicke; Peter Staeheli; Thomas Decker; Mathias Müller; Birgit Strobl

ABSTRACT The transcription factor STAT1 is essential for interferon (IFN)-mediated immunity in humans and mice. STAT1 function is tightly regulated, and both loss- and gain-of-function mutations result in severe immune diseases. The two alternatively spliced isoforms, STAT1α and STAT1β, differ with regard to a C-terminal transactivation domain, which is absent in STAT1β. STAT1β is considered to be transcriptionally inactive and to be a competitive inhibitor of STAT1α. To investigate the functions of the STAT1 isoforms in vivo, we generated mice deficient for either STAT1α or STAT1β. As expected, the functions of STAT1α and STAT1β in IFN-α/β- and IFN-λ-dependent antiviral activity are largely redundant. In contrast to the current dogma, however, we found that STAT1β is transcriptionally active in response to IFN-γ. In the absence of STAT1α, STAT1β shows more prolonged IFN-γ-induced phosphorylation and promoter binding. Both isoforms mediate protective, IFN-γ-dependent immunity against the bacterium Listeria monocytogenes, although with remarkably different efficiencies. Our data shed new light on the potential contributions of the individual STAT1 isoforms to STAT1-dependent immune responses. Knowledge of STAT1βs function will help fine-tune diagnostic approaches and help design more specific strategies to interfere with STAT1 activity.


Journal of Proteomics | 2011

A comparative proteome analysis links tyrosine kinase 2 (Tyk2) to the regulation of cellular glucose and lipid metabolism in response to poly(I:C)

Tom Grunert; Nicole R. Leitner; Martina Marchetti-Deschmann; Ingrid Miller; Barbara Wallner; Marta Radwan; Claus Vogl; Thomas Kolbe; Dagmar Kratky; Manfred Gemeiner; Günter Allmaier; Mathias Müller; Birgit Strobl

Tyrosine kinase 2 (Tyk2) is an integral part of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway which relays intracellular signals of various cytokines. Tyk2 crucially contributes to host defense mechanisms against microbial pathogens and to tumor surveillance but also facilitates immune pathologies. Here we investigated the impact of Tyk2 on the macrophage proteome using the synthetic double-stranded RNA analog polyinosinic acid-polycytidylic acid (poly(I:C)) as a mimicry of viral infections. By means of 2D-DIGE in connection with PMF obtained by MALDI-MS and sequence tag determination by MS/MS we unambiguously identified eighteen protein spots corresponding to sixteen distinct proteins that are regulated by poly(I:C) and differentially expressed between wildtype (WT) and Tyk2-deficient macrophages. The majority of these proteins are functionally assigned to cellular immune responses and to metabolism. We show for selected metabolic enzymes, i.e. triosephosphate isomerase (TIM), ATP-citrate synthase (ACLY) and long-chain-fatty-acid-CoA ligase 4 (ACSL4), that Tyk2 affects protein expression transcriptionally and post-transcriptionally. We furthermore confirm the involvement of Tyk2 in the regulation of lipid and carbohydrate metabolism at the level of metabolites. Taken together, our results provide new evidence for important functions of Tyk2 at the molecular interface between innate immunity and cellular metabolism.


Cytokine | 2017

Tyrosine kinase 2 - Surveillant of tumours and bona fide oncogene.

Nicole R. Leitner; Agnieszka Witalisz-Siepracka; Birgit Strobl; Mathias Müller

Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family, which transduces cytokine and growth factor signalling. Analysis of TYK2 loss-of-function revealed its important role in immunity to infection, (auto-) immunity and (auto-) inflammation. TYK2-deficient patients unravelled high similarity between mice and men with respect to cellular signalling functions and basic immunology. Genome-wide association studies link TYK2 to several autoimmune and inflammatory diseases as well as carcinogenesis. Due to its cytokine signalling functions TYK2 was found to be essential in tumour surveillance. Lately TYK2 activating mutants and fusion proteins were detected in patients diagnosed with leukaemic diseases suggesting that TYK2 is a potent oncogene. Here we review the cell intrinsic and extrinsic functions of TYK2 in the characteristics preventing and enabling carcinogenesis. In addition we describe an unexpected function of kinase-inactive TYK2 in tumour rejection.


Transgenic Research | 2014

Conditional ablation of TYK2 in immunity to viral infection and tumor surveillance.

Raimund M. Vielnascher; Eva Hainzl; Nicole R. Leitner; Michael Rammerstorfer; David Popp; Agnieszka Witalisz; Rita Rom; Marina Karaghiosoff; Thomas Kolbe; Simone Müller; Thomas Rülicke; Caroline Lassnig; Birgit Strobl; Mathias Müller

Tyrosine kinase 2 (TYK2) has a pivotal role in immunity to infection and tumor surveillance. It is associated with several cytokine receptor chains including type I interferon (IFN) receptor 1 (IFNAR1), interleukin- (IL-) 12 receptor beta 1 (IL-12Rb1) and IL-10R2. We have generated a mouse with a conditional Tyk2 null allele and proved integrity of the conditional Tyk2 locus. TYK2 was successfully removed by the use of ubiquitous and tissue-specific Cre-expressing mouse strains. Myeloid TYK2 was found to critically contribute to the defense against murine cytomegalovirus. Ubiquitous TYK2 ablation severely impaired tumor immunosurveillance, while deletion in myeloid, dendritic or T cells alone showed no effect. The conditional Tyk2 mouse strain will be instrumental to further dissect TYK2 functions in infection, inflammation and cancer.


PLOS ONE | 2012

In vivo functional requirement of the mouse Ifitm1 gene for germ cell development, interferon mediated immune response and somitogenesis.

Ingeborg Klymiuk; Lukas Kenner; Thure Adler; Dirk H. Busch; Auke Boersma; Martin Irmler; Valérie Gailus-Durner; Helmut Fuchs; Nicole R. Leitner; Mathias Müller; Ralf Kühn; Michaela Schlederer; Irina Treise; Martin Hrabě de Angelis; Johannes Beckers

The mammalian Interferon induced transmembrane protein 1 (Ifitm1) gene was originally identified as a member of a gene family highly inducible by type I and type II interferons. Based on expression analyses, it was suggested to be required for normal primordial germ cell migration. The knockdown of Ifitm1 in mouse embryos provided evidence for a role in somitogenesis. We generated the first targeted knockin allele of the Ifitm1 gene to systematically reassess all inferred functions. Sperm motility and the fertility of male and female mutant mice are as in wild type littermates. Embryonic somites and the adult vertebral column appear normal in homozygous Ifitm1 knockout mice, demonstrating that Ifitm1 is not essential for normal segmentation of the paraxial mesoderm. Proportions of leucocyte subsets, including granulocytes, monocytes, B-cells, T-cells, NK-cells, and NKT-cells, are unchanged in mutant mice. Based on a normal immune response to Listeria monocytogenes infection, there is no evidence for a dysfunction in downstream IFNγ signaling in Ifitm1 mutant mice. Expression from the Ifitm1 locus from E8.5 to E14.5 is highly dynamic. In contrast, in adult mice, Ifitm1 expression is highly restricted and strong in the bronchial epithelium. Intriguingly, IFITM1 is highly overexpressed in tumor epithelia cells of human squamous cell carcinomas and in adenocarcinomas of NSCLC patients. These analyses underline the general importance of targeted in vivo studies for the functional annotation of the mammalian genome. The first comprehensive description of the Ifitm1 expression pattern provides a rational basis for the further examination of Ifitm1 gene functions. Based on our data, the fact that IFITM1 can function as a negative regulator of cell proliferation, and because the gene maps to chromosome band 11p15.5, previously associated with NSCLC, it is likely that IFITM1 in man has a key role in tumor formation.


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.


Haematologica | 2014

Lactotransferrin-Cre reporter mice trace neutrophils, monocytes/macrophages and distinct subtypes of dendritic cells

Boris Kovacic; Andrea Hoelbl-Kovacic; Katrin M. Fischhuber; Nicole R. Leitner; Dagmar Gotthardt; Emilio Casanova; Veronika Sexl; Mathias Müller

Considerable effort has been expended to identify genes that account for myeloid lineage commitment and development. However, currently available non-invasive mouse models utilize myeloid-specific reporters that are significantly expressed in hematopoietic stem cells as well as lymphoid compartments. Here, we describe a myeloid-specific marker that is not shared by any other lineage. We show that lactotransferrin mRNA is expressed by Gr-1+/CD11b+ cells in the bone marrow, as opposed to hematopoietic stem cells or any peripheral cell population. To follow the progeny of lactotransferrin-expressing bone marrow cells, we generated a mouse model in which a reporter gene is irreversibly activated from the lactotransferrin-promoter. We found that lactotransferrin-reporter labels a majority of neutrophils, monocytes, macrophages and distinct subtypes of dendritic cells, while excluding T, B, natural killer cells, interferon-producing killer dendritic cells, plasmacytoid dendritic cells, erythrocytes and eosinophils. Lactotransferrin-reporter− bone marrow cells retain lymphoid, erythroid and long-term repopulating potential, while lactotransferrin-reporter+ bone marrow cells confer only myeloid, but not lymphoid potential. We conclude that lactotransferrin represents a late stage differentiation marker of neutrophils, macrophages and distinct subtypes of dendritic cells.


BMC Biotechnology | 2006

A time- and dose-dependent STAT1 expression system

Nicole R. Leitner; Birgit Strobl; Marion Bokor; Ronald Painz; Thomas Kolbe; Thomas Rülicke; Mathias Müller; Marina Karaghiosoff

BackgroundThe signal transducer and activator of transcription (STAT) family of transcription factors mediates a variety of cytokine dependent gene regulations. STAT1 has been mainly characterized by its role in interferon (IFN) type I and II signaling and STAT1 deficiency leads to high susceptibility to several pathogens. For fine-tuned analysis of STAT1 function we established a dimerizer-inducible system for STAT1 expression in vitro and in vivo.ResultsThe functionality of the dimerizer-induced STAT1 system is demonstrated in vitro in mouse embryonic fibroblasts and embryonic stem cells. We show that this two-vector based system is highly inducible and does not show any STAT1 expression in the absence of the inducer. Reconstitution of STAT1 deficient cells with inducible STAT1 restores IFNγ-mediated gene induction, antiviral responses and STAT1 activation remains dependent on cytokine stimulation. STAT1 expression is induced rapidly upon addition of dimerizer and expression levels can be regulated in a dose-dependent manner. Furthermore we show that in transgenic mice STAT1 can be induced upon stimulation with the dimerizer, although only at low levels.ConclusionThese results prove that the dimerizer-induced system is a powerful tool for STAT1 analysis in vitro and provide evidence that the system is suitable for the use in transgenic mice. To our knowledge this is the first report for inducible STAT1 expression in a time- and dose-dependent manner.

Collaboration


Dive into the Nicole R. Leitner's collaboration.

Top Co-Authors

Avatar

Mathias Müller

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Birgit Strobl

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Thomas Kolbe

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Thomas Rülicke

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Caroline Lassnig

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Christian Semper

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simone Müller

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Barbara Wallner

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Claus Vogl

University of Veterinary Medicine Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge