Network


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

Hotspot


Dive into the research topics where Tahereh Javaheri is active.

Publication


Featured researches published by Tahereh Javaheri.


Nature Communications | 2015

STAT3 regulated ARF expression suppresses prostate cancer metastasis.

Jan Pencik; Michaela Schlederer; Wolfgang Gruber; Christine Unger; Steven M. Walker; Athena Chalaris; I. Marie; Melanie R. Hassler; Tahereh Javaheri; Osman Aksoy; Jaine K. Blayney; Nicole Prutsch; Anna Skucha; Merima Herac; Oliver H. Krämer; Peter R. Mazal; Florian Grebien; Gerda Egger; Valeria Poli; Wolfgang Mikulits; Robert Eferl; Harald Esterbauer; Richard D. Kennedy; Falko Fend; Marcus Scharpf; Martin Braun; Sven Perner; David E. Levy; Timothy Ian Malcolm; Suzanne D. Turner

Prostate cancer (PCa) is the most prevalent cancer in men. Hyperactive STAT3 is thought to be oncogenic in PCa. However, targeting of the IL-6/STAT3 axis in PCa patients has failed to provide therapeutic benefit. Here we show that genetic inactivation of Stat3 or IL-6 signalling in a Pten-deficient PCa mouse model accelerates cancer progression leading to metastasis. Mechanistically, we identify p19ARF as a direct Stat3 target. Loss of Stat3 signalling disrupts the ARF–Mdm2–p53 tumour suppressor axis bypassing senescence. Strikingly, we also identify STAT3 and CDKN2A mutations in primary human PCa. STAT3 and CDKN2A deletions co-occurred with high frequency in PCa metastases. In accordance, loss of STAT3 and p14ARF expression in patient tumours correlates with increased risk of disease recurrence and metastatic PCa. Thus, STAT3 and ARF may be prognostic markers to stratify high from low risk PCa patients. Our findings challenge the current discussion on therapeutic benefit or risk of IL-6/STAT3 inhibition.


Cytokine | 2016

JAK-STAT signaling in cancer: From cytokines to non-coding genome

Jan Pencik; Ha Thi Thanh Pham; Johannes Schmoellerl; Tahereh Javaheri; Michaela Schlederer; Zoran Culig; Olaf Merkel; Richard Moriggl; Florian Grebien; Lukas Kenner

In the past decades, studies of the Janus kinases (JAKs) and signal transducers and activators of transcription (STATs) signaling have uncovered highly conserved programs linking cytokine signaling to the regulation of essential cellular mechanisms such as proliferation, invasion, survival, inflammation and immunity. Inhibitors of the JAK/STAT pathway are used for treatment of autoimmune diseases, such as rheumatoid arthritis or psoriasis. Aberrant JAK/STAT signaling has been identified to contribute to cancer progression and metastatic development. Targeting of JAK/STAT pathway is currently one of the most promising therapeutic strategies in prostate cancer (PCa), hematopoietic malignancies and sarcomas. Notably, newly identified regulators of JAK/STAT signaling, the non-coding RNAs transcripts and their role as important targets and potential clinical biomarkers are highlighted in this review. In addition to the established role of the JAK/STAT signaling pathway in traditional cytokine signaling the non-coding RNAs add yet another layer of hidden regulation and function. Understanding the crosstalk of non-coding RNA with JAK/STAT signaling in cancer is of critical importance and may result in better patient stratification not only in terms of prognosis but also in the context of therapy.


Oncotarget | 2017

Combined experience of six independent laboratories attempting to create an Ewing sarcoma mouse model

Tsion Zewdu Minas; Didier Surdez; Tahereh Javaheri; Miwa Tanaka; Michelle Marques Howarth; Hong-Jun Kang; Jenny Han; Zhi-Yan Han; Barbara Sax; Barbara E. Kream; Sung-Hyeok Hong; Haydar Çelik; Franck Tirode; Jan Tuckermann; Jeffrey A. Toretsky; Lukas Kenner; Heinrich Kovar; Sean Lee; E. Alejandro Sweet-Cordero; Takuro Nakamura; Richard Moriggl; Olivier Delattre; Aykut Üren

Ewing sarcoma (ES) involves a tumor-specific chromosomal translocation that produces the EWS-FLI1 protein, which is required for the growth of ES cells both in vitro and in vivo. However, an EWS-FLI1-driven transgenic mouse model is not currently available. Here, we present data from six independent laboratories seeking an alternative approach to express EWS-FLI1 in different murine tissues. We used the Runx2, Col1a2.3, Col1a3.6, Prx1, CAG, Nse, NEFL, Dermo1, P0, Sox9 and Osterix promoters to target EWS-FLI1 or Cre expression. Additional approaches included the induction of an endogenous chromosomal translocation, in utero knock-in, and the injection of Cre-expressing adenovirus to induce EWS-FLI1 expression locally in multiple lineages. Most models resulted in embryonic lethality or developmental defects. EWS-FLI1-induced apoptosis, promoter leakiness, the lack of potential cofactors, and the difficulty of expressing EWS-FLI1 in specific sites were considered the primary reasons for the failed attempts to create a transgenic mouse model of ES.


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.


Cell Death and Disease | 2016

Increased survival and cell cycle progression pathways are required for EWS/FLI1-induced malignant transformation

Tahereh Javaheri; Zahra Kazemi; Jan Pencik; Ha T. T. Pham; Maximilian Kauer; Rahil Noorizadeh; Barbara Sax; Harini Nivarthi; Michaela Schlederer; Barbara Maurer; Maximillian Hofbauer; Dave N. T. Aryee; Marc Wiedner; Eleni M. Tomazou; Malcolm Logan; Christine Hartmann; Jan Tuckermann; Lukas Kenner; Mario Mikula; Helmut Dolznig; Aykut Üren; Günther H. Richter; Florian Grebien; Heinrich Kovar; Richard Moriggl

Ewing sarcoma (ES) is the second most frequent childhood bone cancer driven by the EWS/FLI1 (EF) fusion protein. Genetically defined ES models are needed to understand how EF expression changes bone precursor cell differentiation, how ES arises and through which mechanisms of inhibition it can be targeted. We used mesenchymal Prx1-directed conditional EF expression in mice to study bone development and to establish a reliable sarcoma model. EF expression arrested early chondrocyte and osteoblast differentiation due to changed signaling pathways such as hedgehog, WNT or growth factor signaling. Mesenchymal stem cells (MSCs) expressing EF showed high self-renewal capacity and maintained an undifferentiated state despite high apoptosis. Blocking apoptosis through enforced BCL2 family member expression in MSCs promoted efficient and rapid sarcoma formation when transplanted to immunocompromised mice. Mechanistically, high BCL2 family member and CDK4, but low P53 and INK4A protein expression synergized in Ewing-like sarcoma development. Functionally, knockdown of Mcl1 or Cdk4 or their combined pharmacologic inhibition resulted in growth arrest and apoptosis in both established human ES cell lines and EF-transformed mouse MSCs. Combinatorial targeting of survival and cell cycle progression pathways could counteract this aggressive childhood cancer.


Scientific Reports | 2017

RGD delivery of truncated coagulase to tumor vasculature affords local thrombotic activity to induce infarction of tumors in mice

Rana Jahanban-Esfahlan; Khaled Seidi; Hassan Monhemi; Amir Daei Farshchi Adli; Babak Minofar; Peyman Zare; Davoud Farajzadeh; Safar Farajnia; Ramezan Behzadi; Mehran Mesgari Abbasi; Nosratollah Zarghami; Tahereh Javaheri

Induction of thrombosis in tumor vasculature represents an appealing strategy for combating cancer. Herein, we combined unique intrinsic coagulation properties of staphylocoagulase with new acquired functional potentials introduced by genetic engineering, to generate a novel bi-functional fusion protein consisting of truncated coagulase (tCoa) bearing an RGD motif on its C-terminus for cancer therapy. We demonstrated that free coagulase failed to elicit any significant thrombotic activity. Conversely, RGD delivery of coagulase retained coagulase activity and afforded favorable interaction of fusion proteins with prothrombin and αvβ3 endothelial cell receptors, as verified by in silico, in vitro, and in vivo experiments. Although free coagulase elicited robust coagulase activity in vitro, only targeted coagulase (tCoa-RGD) was capable of producing extensive thrombosis, and subsequent infarction and massive necrosis of CT26 mouse colon, 4T1 mouse mammary and SKOV3 human ovarian tumors in mice. Additionally, systemic injections of lower doses of tCoa-RGD produced striking tumor growth inhibition of CT26, 4T1 and SKOV3 solid tumors in animals. Altogether, the nontoxic nature, unique shortcut mechanism, minimal effective dose, wide therapeutic window, efficient induction of thrombosis, local effects and susceptibility of human blood to coagulase suggest tCoa-RGD fusion proteins as a novel and promising anticancer therapy for human trials.


Journal of Clinical Investigation | 2018

STAT5BN642H is a driver mutation for T cell neoplasia

Ha Thi Thanh Pham; Barbara Maurer; Michaela Prchal-Murphy; Reinhard Grausenburger; Eva Grundschober; Tahereh Javaheri; Harini Nivarthi; Auke Boersma; Thomas Kolbe; Mohamed Elabd; Florian Halbritter; Jan Pencik; Zahra Kazemi; Florian Grebien; Markus Hengstschläger; Lukas Kenner; Stefan Kubicek; Matthias Farlik; Christoph Bock; Peter Valent; Mathias Müller; Thomas Rülicke; Veronika Sexl; Richard Moriggl

STAT5B is often mutated in hematopoietic malignancies. The most frequent STAT5B mutation, Asp642His (N642H), has been found in over 90 leukemia and lymphoma patients. Here, we used the Vav1 promoter to generate transgenic mouse models that expressed either human STAT5B or STAT5BN642H in the hematopoietic compartment. While STAT5B-expressing mice lacked a hematopoietic phenotype, the STAT5BN642H-expressing mice rapidly developed T cell neoplasms. Neoplasia manifested as transplantable CD8+ lymphoma or leukemia, indicating that the STAT5BN642H mutation drives cancer development. Persistent and enhanced levels of STAT5BN642H tyrosine phosphorylation in transformed CD8+ T cells led to profound changes in gene expression that were accompanied by alterations in DNA methylation at potential histone methyltransferase EZH2-binding sites. Aurora kinase genes were enriched in STAT5BN642H-expressing CD8+ T cells, which were exquisitely sensitive to JAK and Aurora kinase inhibitors. Together, our data suggest that JAK and Aurora kinase inhibitors should be further explored as potential therapeutics for lymphoma and leukemia patients with the STAT5BN642H mutation who respond poorly to conventional chemotherapy.


Molecular Pharmacology | 2016

Repurposing treprostinil for enhancing hematopoietic progenitor cell transplantation

Zahra Kazemi; Christian Bergmayr; Michaela Prchal-Murphy; Tahereh Javaheri; Madeleine Themanns; Ha T. T. Pham; Wolfgang Strohmaier; Veronika Sexl; Michael Freissmuth; Eva Zebedin-Brandl

Activation of Gs-coupled receptors enhances engraftment of hematopoietic stem and progenitor cells (HSPCs). We tested the hypothesis that treprostinil, a prostacyclin analog approved for the treatment of pulmonary hypertension, can be repurposed to improve hematopoietic stem cell transplantation. Murine and human HSPCs were isolated from bone marrow and umbilical cord blood, respectively. Prostanoid receptor agonists and the combination thereof with forskolin were tested for their capacity to stimulate [3H]cAMP accumulation in HSPCs. Three independent approaches were employed to verify the ability of agonist-activated HSPCs to reconstitute the bone marrow in lethally irradiated recipient mice. The underlying mechanism was explored in cellular migration assays and by blocking C-X-C motif chemokine receptor 4 (CXCR4). Among several prostanoid agonists tested in combination with forskolin, treprostinil was most efficacious in raising intracellular cAMP levels in murine and human HPSCs. Injection of murine and human HSPCs, which had been pretreated with treprostinil and forskolin, enhanced survival of lethally irradiated recipient mice. Survival was further improved if recipient mice were subcutaneously administered treprostinil (0.15 mg kg−1 8 h−1) for 10 days. This regimen also reduced the number of HSPCs required to rescue lethally irradiated mice. Enhanced survival of recipient mice was causally related to treprostinil-enhanced CXCR4-dependent migration of HSPCs. Treprostinil stimulates the engraftment of human and murine hematopoietic stem cells without impairing their capacity for self-renewal. The investigated dose range corresponds to the dose approved for human use. Hence, these findings may be readily translated into a clinical application.


Nature Communications | 2015

Erratum: STAT3 regulated ARF expression suppresses prostate cancer metastasis.

Jan Pencik; Michaela Schlederer; Wolfgang Gruber; Christine Unger; Steven M. Walker; Athena Chalaris; I. Marie; Melanie R. Hassler; Tahereh Javaheri; Osman Aksoy; Jaine K. Blayney; Nicole Prutsch; Anna Skucha; Merima Herac; Oliver H. Krämer; Peter R. Mazal; Florian Grebien; Gerda Egger; Valeria Poli; Wolfgang Mikulits; Robert Eferl; Harald Esterbauer; Richard D. Kennedy; Falko Fend; Marcus Scharpf; Martin Braun; Sven Perner; David E. Levy; Tim Malcolm; Suzanne D. Turner

Nature Communications 6: Article number:7736 (2015); Published: 22 July 2015; Updated: 26 October 2015 The affiliation details for Jan Pencik are incorrect in this Article. The correct affiliation details for this author are given below: Ludwig Boltzmann Institute for Cancer Research, Waehringerstrasse 13A, 1090 Vienna, Austria.


Journal of Controlled Release | 2018

Tumor target amplification: Implications for nano drug delivery systems

Khaled Seidi; Heidi A. Neubauer; Richard Moriggl; Rana Jahanban-Esfahlan; Tahereh Javaheri

&NA; Tumor cells overexpress surface markers which are absent from normal cells. These tumor‐restricted antigenic signatures are a fundamental basis for distinguishing on‐target from off‐target cells for ligand‐directed targeting of cancer cells. Unfortunately, tumor heterogeneity impedes the establishment of a solid expression pattern for a given target marker, leading to drastic changes in quality (availability) and quantity (number) of the target. Consequently, a subset of cancer cells remains untargeted during the course of treatment, which subsequently promotes drug‐resistance and cancer relapse. Since target inefficiency is only problematic for cancer treatment and not for treatment of other pathological conditions such as viral/bacterial infections, target amplification or the generation of novel targets is key to providing eligible antigenic markers for effective targeted therapy. This review summarizes the limitations of current ligand‐directed targeting strategies and provides a comprehensive overview of tumor target amplification strategies, including self‐amplifying systems, dual targeting, artificial markers and peptide modification. We also discuss the therapeutic and diagnostic potential of these approaches, the underlying mechanism(s) and established methodologies, mostly in the context of different nanodelivery systems, to facilitate more effective ligand‐directed cancer cell monitoring and targeting. Graphical abstract Figure. No Caption available.

Collaboration


Dive into the Tahereh Javaheri's collaboration.

Top Co-Authors

Avatar

Richard Moriggl

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar

Michaela Schlederer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Jan Pencik

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

Florian Grebien

Austrian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Heinrich Kovar

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Zahra Kazemi

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christine Unger

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Gerda Egger

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge