Network


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

Hotspot


Dive into the research topics where Kristina M. Mueller is active.

Publication


Featured researches published by Kristina M. Mueller.


Hepatology | 2011

Impairment of hepatic growth hormone and glucocorticoid receptor signaling causes steatosis and hepatocellular carcinoma in mice

Kristina M. Mueller; Jan-Wilhelm Kornfeld; Katrin Friedbichler; Leander Blaas; Gerda Egger; Harald Esterbauer; Peter Hasselblatt; Michaela Schlederer; Susanne Haindl; Kay Uwe Wagner; David Engblom; Guenter Haemmerle; Dagmar Kratky; Veronika Sexl; Lukas Kenner; Andrey V. Kozlov; Luigi Terracciano; Rudolf Zechner; Guenther Schuetz; Emilio Casanova; J. Andrew Pospisilik; Markus H. Heim; Richard Moriggl

Growth hormone (GH)‐activated signal transducer and activator of transcription 5 (STAT5) and the glucocorticoid (GC)‐responsive glucocorticoid receptor (GR) are important signal integrators in the liver during metabolic and physiologic stress. Their deregulation has been implicated in the development of metabolic liver diseases, such as steatosis and progression to fibrosis. Using liver‐specific STAT5 and GR knockout mice, we addressed their role in metabolism and liver cancer onset. STAT5 single and STAT5/GR double mutants developed steatosis, but only double‐mutant mice progressed to liver cancer. Mechanistically, STAT5 deficiency led to the up‐regulation of prolipogenic sterol regulatory element binding protein 1 (SREBP‐1) and peroxisome proliferator activated receptor gamma (PPAR‐γ) signaling. Combined loss of STAT5/GR resulted in GH resistance and hypercortisolism. The combination of both induced expression of adipose tissue lipases, adipose tissue lipid mobilization, and lipid flux to the liver, thereby aggravating STAT5‐dependent steatosis. The metabolic dysfunctions in STAT5/GR compound knockout animals led to the development of hepatic dysplasia at 9 months of age. At 12 months, 35% of STAT5/GR‐deficient livers harbored dysplastic nodules and ∼60% hepatocellular carcinomas (HCCs). HCC development was associated with GH and insulin resistance, enhanced tumor necrosis factor alpha (TNF‐α) expression, high reactive oxygen species levels, and augmented liver and DNA damage parameters. Moreover, activation of the c‐Jun N‐terminal kinase 1 (JNK1) and STAT3 was prominent. Conclusion: Hepatic STAT5/GR signaling is crucial for the maintenance of systemic lipid homeostasis. Impairment of both signaling cascades causes severe metabolic liver disease and promotes spontaneous hepatic tumorigenesis. (HEPATOLOGY 2011;54:1398–1409)


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.


Molecular and Cellular Endocrinology | 2012

Hepatic growth hormone and glucocorticoid receptor signaling in body growth, steatosis and metabolic liver cancer development

Kristina M. Mueller; Madeleine Themanns; Katrin Friedbichler; Jan-Wilhelm Kornfeld; Harald Esterbauer; Jan Tuckermann; Richard Moriggl

Highlights ► Conditional mouse models illuminated the role of hepatic GH-STAT5 and GC-GR signaling in liver function. ► We provide an overview of overlapping and distinct functions of hepatic GH-STAT5 and GC-GR signaling in growth/metabolism. ► Impaired hepatic GH-STAT5 signaling sensitizes hepatocytes to injury and tumorigenic transformation. ► Loss of hepatic GR function causes chronic stress, thereby aggravating liver cancer formation upon impaired STAT5 function.


Hepatology | 2012

Growth-hormone-induced signal transducer and activator of transcription 5 signaling causes gigantism, inflammation, and premature death but protects mice from aggressive liver cancer.

Katrin Friedbichler; Madeleine Themanns; Kristina M. Mueller; Michaela Schlederer; Jan-Wilhelm Kornfeld; Luigi Terracciano; Andrey V. Kozlov; Susanne Haindl; Lukas Kenner; Thomas Kolbe; Mathias Mueller; Kenneth J. Snibson; Markus H. Heim; Richard Moriggl

Persistently high levels of growth hormone (GH) can cause liver cancer. GH activates multiple signal‐transduction pathways, among them janus kinase (JAK) 2‐signal transducer and activator of transcription (STAT) 5 (signal transducer and activator of transcription 5). Both hyperactivation and deletion of STAT5 in hepatocytes have been implicated in the development of hepatocellular carcinoma (HCC); nevertheless, the role of STAT5 in the development of HCC as a result of high GH levels remains enigmatic. Thus, we crossed a mouse model of gigantism and inflammatory liver cancer caused by hyperactivated GH signaling (GHtg) to mice with hepatic deletion of STAT5 (STAT5Δhep). Unlike GHtg mice, GHtgSTAT5Δhep animals did not display gigantism. Moreover, the premature mortality, which was associated with chronic inflammation, as well as the pathologic alterations of hepatocytes observed in GHtg mice, were not observed in GHtg animals lacking STAT5. Strikingly, loss of hepatic STAT5 proteins led to enhanced HCC development in GHtg mice. Despite reduced chronic inflammation, GHtgSTAT5Δhep mice displayed earlier and more advanced HCC than GHtg animals. This may be attributed to the combination of increased peripheral lipolysis, hepatic lipid synthesis, loss of hepatoprotective mediators accompanied by aberrant activation of tumor‐promoting c‐JUN and STAT3 signaling cascades, and accumulation of DNA damage secondary to loss of cell‐cycle control. Thus, HCC was never observed in STAT5Δhep mice. Conclusion: As a result of their hepatoprotective functions, STAT5 proteins prevent progressive fatty liver disease and the formation of aggressive HCC in the setting of hyperactivated GH signaling. At the same time, they play a key role in controlling systemic inflammation and regulating organ and body size. (Hepatology 2012)


PLOS ONE | 2014

Reliable Quantification of Protein Expression and Cellular Localization in Histological Sections

Michaela Schlederer; Kristina M. Mueller; Johannes Haybaeck; Susanne Heider; Nicole Huttary; Margit Rosner; Markus Hengstschläger; Richard Moriggl; Helmut Dolznig; Lukas Kenner

In targeted therapy, patient tumors are analyzed for aberrant activations of core cancer pathways, monitored based on biomarker expression, to ensure efficient treatment. Thus, diagnosis and therapeutic decisions are often based on the status of biomarkers determined by immunohistochemistry in combination with other clinical parameters. Standard evaluation of cancer specimen by immunohistochemistry is frequently impeded by its dependence on subjective interpretation, showing considerable intra- and inter-observer variability. To make treatment decisions more reliable, automated image analysis is an attractive possibility to reproducibly quantify biomarker expression in patient tissue samples. We tested whether image analysis could detect subtle differences in protein expression levels. Gene dosage effects generate well-graded expression patterns for most gene-products, which vary by a factor of two between wildtype and haploinsufficient cells lacking one allele. We used conditional mouse models with deletion of the transcription factors Stat5ab in the liver as well Junb deletion in a T-cell lymphoma model. We quantified the expression of total or activated STAT5AB or JUNB protein in normal (Stat5ab+/+ or JunB+/+), hemizygous (Stat5ab+/Δ or JunB+/Δ) or knockout (Stat5abΔ/Δ or JunBΔ/Δ) settings. Image analysis was able to accurately detect hemizygosity at the protein level. Moreover, nuclear signals were distinguished from cytoplasmic expression and translocation of the transcription factors from the cytoplasm to the nucleus was reliably detected and quantified using image analysis. We demonstrate that image analysis supported pathologists to score nuclear STAT5AB expression levels in immunohistologically stained human hepatocellular patient samples and decreased inter-observer variability.


Oncotarget | 2015

YK-4-279 effectively antagonizes EWS-FLI1 induced leukemia in a transgenic mouse model

Tsion Zewdu Minas; Jenny Han; Tahereh Javaheri; Sung-Hyeok Hong; Michaela Schlederer; Yasemin Saygideğer-Kont; Haydar Çelik; Kristina M. Mueller; Idil Temel; Metin Ozdemirli; Heinrich Kovar; Hayriye V. Erkizan; Jeffrey A. Toretsky; Lukas Kenner; Richard Moriggl; Aykut Üren

Ewing sarcoma is an aggressive tumor of bone and soft tissue affecting predominantly children and young adults. Tumor-specific chromosomal translocations create EWS-FLI1 and similar aberrant ETS fusion proteins that drive sarcoma development in patients. ETS family fusion proteins and over-expressed ETS proteins are also found in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. Transgenic expression of EWS-FLI1 in mice promotes high penetrance erythroid leukemia with dense hepatic and splenic infiltrations. We identified a small molecule, YK-4-279, that directly binds to EWS-FLI1 and inhibits its oncogenic activity in Ewing sarcoma cell lines and xenograft mouse models. Herein, we tested in vivo therapeutic efficacy and potential side effects of YK-4-279 in the transgenic mouse model with EWS-FLI1 induced leukemia. A two-week course of treatment with YK-4-279 significantly reduced white blood cell count, nucleated erythroblasts in the peripheral blood, splenomegaly, and hepatomegaly of erythroleukemic mice. YK-4-279 inhibited EWS-FLI1 target gene expression in neoplastic cells. Treated animals showed significantly better overall survival compared to control mice that rapidly succumbed to leukemia. YK-4-279 treated mice did not show overt toxicity in liver, spleen, or bone marrow. In conclusion, this in vivo study highlights the efficacy of YK-4-279 to treat EWS-FLI1 expressing neoplasms and support its therapeutic potential for patients with Ewing sarcoma and other ETS-driven malignancies.


Diabetes | 2017

Adipocyte Glucocorticoid Receptor Deficiency Attenuates Aging- and Hfd-Induced Obesity, and Impairs the Feeding-Fasting Transition.

Kristina M. Mueller; Kerstin Hartmann; Doris Kaltenecker; Sabine Vettorazzi; Mandy Bauer; Lea Mauser; Sabine Amann; Sigrid Jall; Katrin Fischer; Harald Esterbauer; Timo D. Müller; Matthias H. Tschöp; Christoph Magnes; Johannes Haybaeck; Thomas Scherer; Natalie Bordag; Jan Tuckermann; Richard Moriggl

Glucocorticoids (GCs) are important regulators of systemic energy metabolism, and aberrant GC action is linked to metabolic dysfunctions. Yet, the extent to which normal and pathophysiological energy metabolism depend on the GC receptor (GR) in adipocytes remains unclear. Here, we demonstrate that adipocyte GR deficiency in mice significantly impacts systemic metabolism in different energetic states. Plasma metabolomics and biochemical analyses revealed a marked global effect of GR deficiency on systemic metabolite abundance and, thus, substrate partitioning in fed and fasted states. This correlated with a decreased lipolytic capacity of GR-deficient adipocytes under postabsorptive and fasting conditions, resulting from impaired signal transduction from β-adrenergic receptors to adenylate cyclase. Upon prolonged fasting, the impaired lipolytic response resulted in abnormal substrate utilization and lean mass wasting. Conversely, GR deficiency attenuated aging-/diet-associated obesity, adipocyte hypertrophy, and liver steatosis. Systemic glucose tolerance was improved in obese GR-deficient mice, which was associated with increased insulin signaling in muscle and adipose tissue. We conclude that the GR in adipocytes exerts central but diverging roles in the regulation of metabolic homeostasis depending on the energetic state. The adipocyte GR is indispensable for the feeding-fasting transition but also promotes adiposity and associated metabolic disorders in fat-fed and aged mice.


Translational Oncology | 2015

Lung Adenocarcinomas and Lung Cancer Cell Lines Show Association of MMP-1 Expression With STAT3 Activation

Alexander Schütz; Katrin Röser; Jana Klitzsch; Franziska Lieder; Fritz Aberger; Wolfgang Gruber; Kristina M. Mueller; Alexander Pupyshev; Richard Moriggl; Karlheinz Friedrich

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in the majority of lung cancer. This study aims at defining connections between STAT3 function and the malignant properties of non–small cell lung carcinoma (NSCLC) cells. To address possible mechanisms by which STAT3 influences invasiveness, the expression of matrix metalloproteinase-1 (MMP-1) was analyzed and correlated with the STAT3 activity status. Studies on both surgical biopsies and on lung cancer cell lines revealed a coincidence of STAT3 activation and strong expression of MMP-1. MMP-1 and tyrosine-phosphorylated activated STAT3 were found co-localized in cancer tissues, most pronounced in tumor fronts, and in particular in adenocarcinomas. STAT3 activity was constitutive, although to different degrees, in the lung cancer cell lines investigated. Three cell lines (BEN, KNS62, and A549) were identified in which STAT3 activitation was inducible by Interleukin-6 (IL-6). In A549 cells, STAT3 activity enhanced the level of MMP-1 mRNA and stimulated transcription from the MMP-1 promoter in IL-6–stimulated A549 cells. STAT3 specificity of this effect was confirmed by STAT3 knockdown through RNA interference. Our results link aberrant activity of STAT3 in lung cancer cells to malignant tumor progression through up-regulation of expression of invasiveness-associated MMPs.


Diabetologia | 2017

Adipocyte STAT5 deficiency promotes adiposity and impairs lipid mobilisation in mice.

Doris Kaltenecker; Kristina M. Mueller; Pia Benedikt; Ursula Feiler; Madeleine Themanns; Michaela Schlederer; Lukas Kenner; Martina Schweiger; Guenter Haemmerle; Richard Moriggl

Aims/hypothesisDysfunction of lipid metabolism in white adipose tissue can substantially interfere with health and quality of life, for example in obesity and associated metabolic diseases. Therefore, it is important to characterise pathways that regulate lipid handling in adipocytes and determine how they affect metabolic homeostasis. Components of the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway are involved in adipocyte physiology and pathophysiology. However, the exact physiological importance of the STAT family member STAT5 in white adipose tissue is yet to be determined. Here, we aimed to delineate adipocyte STAT5 functions in the context of lipid metabolism in white adipose tissue.MethodsWe generated an adipocyte specific knockout of Stat5 in mice using the Adipoq-Cre recombinase transgene followed by in vivo and in vitro biochemical and molecular studies.ResultsAdipocyte-specific deletion of Stat5 resulted in increased adiposity, while insulin resistance and gluconeogenic capacity was decreased, indicating that glucose metabolism can be improved by interfering with adipose STAT5 function. Basal lipolysis and fasting-induced lipid mobilisation were diminished upon STAT5 deficiency, which coincided with reduced levels of the rate-limiting lipase of triacylglycerol hydrolysis, adipose triglyceride lipase (ATGL, encoded by Pnpla2) and its coactivator comparative gene identification 58 (CGI-58). In a mechanistic analysis, we identified a functional STAT5 response element within the Pnpla2 promoter, indicating that Pnpla2 is transcriptionally regulated by STAT5.Conclusions/interpretationOur findings reveal an essential role for STAT5 in maintaining lipid homeostasis in white adipose tissue and provide a rationale for future studies into the potential of STAT5 manipulation to improve outcomes in metabolic diseases.


Scientific Reports | 2016

Hepatic Deletion of Janus Kinase 2 Counteracts Oxidative Stress in Mice

Madeleine Themanns; Kristina M. Mueller; Sonja M. Kessler; Nicole Golob-Schwarzl; Thomas Mohr; Doris Kaltenecker; Jérôme Bourgeais; Jamile Paier-Pourani; Katrin Friedbichler; Doris Schneller; Michaela Schlederer; Eva Zebedin-Brandl; Luigi Terracciano; Xiaonan Han; Lukas Kenner; Kay Uwe Wagner; Wolfgang Mikulits; Andrey V. Kozlov; Markus H. Heim; Fabrice Gouilleux; Johannes Haybaeck; Richard Moriggl

Genetic deletion of the tyrosine kinase JAK2 or the downstream transcription factor STAT5 in liver impairs growth hormone (GH) signalling and thereby promotes fatty liver disease. Hepatic STAT5 deficiency accelerates liver tumourigenesis in presence of high GH levels. To determine whether the upstream kinase JAK2 exerts similar functions, we crossed mice harbouring a hepatocyte-specific deletion of JAK2 (JAK2Δhep) to GH transgenic mice (GHtg) and compared them to GHtgSTAT5Δhep mice. Similar to GHtgSTAT5Δhep mice, JAK2 deficiency resulted in severe steatosis in the GHtg background. However, in contrast to STAT5 deficiency, loss of JAK2 significantly delayed liver tumourigenesis. This was attributed to: (i) activation of STAT3 in STAT5-deficient mice, which was prevented by JAK2 deficiency and (ii) increased detoxification capacity of JAK2-deficient livers, which diminished oxidative damage as compared to GHtgSTAT5Δhep mice, despite equally severe steatosis and reactive oxygen species (ROS) production. The reduced oxidative damage in JAK2-deficient livers was linked to increased expression and activity of glutathione S-transferases (GSTs). Consistent with genetic deletion of Jak2, pharmacological inhibition and siRNA-mediated knockdown of Jak2 led to significant upregulation of Gst isoforms and to reduced hepatic oxidative DNA damage. Therefore, blocking JAK2 function increases detoxifying GSTs in hepatocytes and protects against oxidative liver damage.

Collaboration


Dive into the Kristina M. Mueller's collaboration.

Top Co-Authors

Avatar

Richard Moriggl

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

Michaela Schlederer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Katrin Friedbichler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Doris Kaltenecker

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

Madeleine Themanns

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Johannes Haybaeck

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge