Network


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

Hotspot


Dive into the research topics where Bernd Groner is active.

Publication


Featured researches published by Bernd Groner.


The EMBO Journal | 2003

The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2

Oliver H. Krämer; Ping Zhu; Heather P. Ostendorff; Martin Golebiewski; Jens Tiefenbach; Marvin A. Peters; Boris Brill; Bernd Groner; Ingolf Bach; Thorsten Heinzel; Martin Göttlicher

Histone‐modifying enzymes play essential roles in physiological and aberrant gene regulation. Since histone deacetylases (HDACs) are promising targets of cancer therapy, it is important to understand the mechanisms of HDAC regulation. Selective modulators of HDAC isoenzymes could serve as efficient and well‐tolerated drugs. We show that HDAC2 undergoes basal turnover by the ubiquitin–proteasome pathway. Valproic acid (VPA), in addition to selectively inhibiting the catalytic activity of class I HDACs, induces proteasomal degradation of HDAC2, in contrast to other inhibitors such as trichostatin A (TSA). Basal and VPA‐induced HDAC2 turnover critically depend on the E2 ubiquitin conjugase Ubc8 and the E3 ubiquitin ligase RLIM. Ubc8 gene expression is induced by both VPA and TSA, whereas only TSA simultaneously reduces RLIM protein levels and therefore fails to induce HDAC2 degradation. Thus, poly‐ubiquitination and proteasomal degradation provide an isoenzyme‐selective mechanism for downregulation of HDAC2.


Immunity | 1997

Phosphatidylinositol 3-Kinase Couples the Interleukin-2 Receptor to the Cell Cycle Regulator E2F

Paul Brennan; Jane W. Babbage; Boudewijn M.T. Burgering; Bernd Groner; Karin Reif; Doreen A. Cantrell

Cell cycle progression initiated by interleukin-2 (IL-2) in T cells is critical for lymphoproliferation and an immune response. Phosphatidyl inositol 3-kinase (PI3K) is activated by IL-2. However, nuclear targets for PI3K are not known. Here we identify the cell cycle regulator E2F as an IL-2 target in T lymphocytes and PI3K as the critical signaling pathway. We eliminate both Stat5 and Raf/MEK pathways from E2F regulation. Protein kinase B (PKB) is activated by IL-2 via PI3K. The expression of an active PKB is sufficient to induce E2F activity. Inhibition of PI3K inhibits phosphorylation of Rb, induction of cyclin D3, and degradation of p27kip1. These results establish a crucial PI3K/PKB-mediated link between the IL-2 teceptor and the cell cycle machinery.


Molecular and Cellular Biology | 1997

Comparison of the transactivation domains of Stat5 and Stat6 in lymphoid cells and mammary epithelial cells.

Richard Moriggl; Susanne Berchtold; Karlheinz Friedrich; Gesche J. R. Standke; Winfried Kammer; Markus Heim; Manuela Wissler; Elisabeth Stöcklin; Fabrice Gouilleux; Bernd Groner

Stat (signal transducers and activators of transcription) and Jak (Janus kinases) proteins are central components in the signal transduction events in hematopoietic and epithelial cells. They are rapidly activated by various cytokines, hormones, and growth factors. Upon ligand binding and cytokine receptor dimerization, Stat proteins are phosphorylated on tyrosine residues by Jak kinases. Activated Stat proteins form homo- or heterodimers, translocate to the nucleus, and induce transcription from responsive genes. Stat5 and Stat6 are transcription factors active in mammary epithelial cells and immune cells. Prolactin activates Stat5, and interleukin-4 (IL-4) activates Stat6. Both cytokines are able to stimulate cell proliferation, differentiation, and survival. We investigated the transactivation potential of Stat6 and found that it is not restricted to lymphocytes. IL-4-dependent activation of Stat6 was also observed in HC11 mammary epithelial cells. In these cells, Stat6 activation led to the induction of the beta-casein gene promoter. The induction of this promoter was confirmed in COS7 cells. The glucocorticoid receptor was able to further enhance IL-4-induced gene transcription through the action of Stat6. Deletion analysis of the carboxyl-terminal region of Stat6 and recombination of this region with a heterologous DNA binding domain allowed the delimitation and characterization of the transactivation domain of Stat6. The potencies of the transactivation domains of Stat5, Stat6, and viral protein VP16 were compared. Stat6 had a transactivation domain which was about 10-fold stronger than that of Stat5. In pre-B cells (Ba/F3), the transactivation domain of Stat6 was IL-4 regulated, independently from its DNA binding function.


Journal of Biological Chemistry | 1996

Participation of JAK and STAT Proteins in Growth Hormone-induced Signaling

Yulong Han; Douglas W. Leaman; Diane Watling; Neil C. Rogers; Bernd Groner; Ian M. Kerr; William I. Wood; George R. Stark

The binding of growth hormone leads to dimerization of its receptor, accompanied by phosphorylation and activation of intracellular tyrosine kinases (JAKs) and the latent cytoplasmic transcriptions factors STAT1, STAT3, and STAT5. Both JAK1 and JAK2 are phosphorylated in response to growth hormone in mouse 3T3 F442A and human HT1080 cells. The roles of JAKs in growth hormone signal transduction were examined by using mutant HT1080 cells missing either JAK1 or JAK2. JAK2 is absolutely required for growth hormone-dependent phosphorylation of the receptor, STAT1 and STAT3, JAK1, and the SH2-containing adaptor molecule Shc. In contrast, JAK1 is not required for any of the above functions. These data indicate that JAK2 is both necessary and sufficient for the growth hormone-dependent phosphorylation events required to couple the receptor both to STAT-dependent signaling pathways and to pathways involving Shc. Furthermore, STAT5 is activated by growth hormone in 3T3 F442A cells, but not in HT1080 cells, revealing that the set of STATs activated by growth hormone can vary, possibly contributing to the specificity of the growth hormone response in different cell types.


Nature Genetics | 2010

miR-212 and miR-132 are required for epithelial stromal interactions necessary for mouse mammary gland development

Ahmet Ucar; Vida Vafaizadeh; Hubertus Jarry; Jan Fiedler; Petra Klemmt; Thomas Thum; Bernd Groner; Kamal Chowdhury

MicroRNAs are small noncoding RNAs that carry out post-transcriptional regulation of the expression of their target genes. However, their roles in mammalian organogenesis are only beginning to be understood. Here we show that the microRNA-212/132 family (which comprises miR-212 and miR-132) is indispensable during the development of the mammary glands in mice, particulary for the regulation of the outgrowth of the epithelial ducts. Mammary transplantation experiments revealed that the function of the miR-212/132 family is required in the stroma but not in the epithelia. Both miR-212 and miR-132 are expressed exclusively in mammary stroma and directly target the matrix metalloproteinase MMP-9. In glands that lack miR-212 and miR-132, MMP-9 expression increases and accumulates around the ducts. This may interfere with collagen deposition and lead to hyperactivation of the tumor growth factor-β signaling pathway, thereby impairing ductal outgrowth. Our results identify the miR-212/132 family as one of the main regulators of the epithelial-stromal interactions that are required for proper pubertal development of the mammary gland.


Science Signaling | 2014

STAT3 Induction of miR-146b Forms a Feedback Loop to Inhibit the NF-κB to IL-6 Signaling Axis and STAT3-Driven Cancer Phenotypes

Michael Xiang; Nicolai Juul Birkbak; Vida Vafaizadeh; Sarah R. Walker; Jennifer E. Yeh; Suhu Liu; Yasmin Kroll; Mark P. Boldin; Konstantin D. Taganov; Bernd Groner; Andrea L. Richardson; David A. Frank

An epigenetic modification prevents the production of a tumor-suppressing and anti-inflammatory microRNA in receptor-negative breast cancers. Micro-Mediated Feedback Chronic inflammation and interleukin-6 (IL-6), which is produced in response to nuclear factor κB (NF-κB) signaling, is a proinflammatory cytokine associated with cancer. Signal transducer and activator of transcription 3 (STAT3) is a transcription factor stimulated in response to IL-6 and its receptor-bound kinases from the Janus kinase (JAK) family. Xiang et al. found that STAT3 stimulated expression of the gene encoding the microRNA miR-146b, which inhibited NF-κB–mediated induction of IL-6 to prevent a proinflammatory response in normal breast epithelial cells. However, promoter methylation reduced miR-146b expression in breast cancer cell lines and patient tissue, and its expression correlated with survival in patients with estrogen receptor– or triple-negative breast cancer. In addition to inhibiting STAT3 activity and cell migration and invasion, introduction of a miR-146b mimic was as cytotoxic as pharmacological inhibition of JAK to triple-negative breast cancer cells in culture, and combination therapy in cells was additive. The findings suggest that therapies reintroducing or stimulating miR-146b production may be beneficial to patients with tumors with high STAT3 activity. Interleukin-6 (IL-6)–mediated activation of signal transducer and activator of transcription 3 (STAT3) is a mechanism by which chronic inflammation can contribute to cancer and is a common oncogenic event. We discovered a pathway, the loss of which is associated with persistent STAT3 activation in human cancer. We found that the gene encoding the tumor suppressor microRNA miR-146b is a direct STAT3 target gene, and its expression was increased in normal breast epithelial cells but decreased in tumor cells. Methylation of the miR-146b promoter, which inhibited STAT3-mediated induction of expression, was increased in primary breast cancers. Moreover, we found that miR-146b inhibited nuclear factor κB (NF-κB)–dependent production of IL-6, subsequent STAT3 activation, and IL-6/STAT3–driven migration and invasion in breast cancer cells, thereby establishing a negative feedback loop. In addition, higher expression of miR-146b was positively correlated with patient survival in breast cancer subtypes with increased IL6 expression and STAT3 phosphorylation. Our results identify an epigenetic mechanism of crosstalk between STAT3 and NF-κB relevant to constitutive STAT3 activation in malignancy and the role of inflammation in oncogenesis.


Journal of Immunology | 2000

Cooperation Among Stat1, Glucocorticoid Receptor, and PU.1 in Transcriptional Activation of the High-Affinity Fcγ Receptor I in Monocytes

Saara Aittomäki; Marko Pesu; Bernd Groner; Olli A. Jänne; Jorma J. Palvimo; Olli Silvennoinen

IFN-γ and glucocorticoids regulate inflammatory and immune responses through Stat1 and glucocorticoid receptor (GR) transcription factors, respectively. The biological responses to these polypeptides are determined by integration of various signaling pathways in a cell-type and promoter-dependent manner. In this study we have characterized the molecular basis for the functional cooperation between IFN-γ and dexamethasone (Dex) in the induction of the high-affinity Fcγ receptor I (FcγRI) in monocytes. Dex did not affect IFN-γ-induced Stat1 DNA binding activity or induce novel DNA-binding complexes to the FcγRI promoter. By using cell systems lacking functional GR or Stat1, we showed that GR stimulated Stat1-dependent transcription in a ligand-dependent manner, while Stat1 did not influence GR-dependent transcription. The cooperation required phosphorylation of Tyr701, DNA binding, and the trans-activation domain of Stat1, but did not involve Ser727 phosphorylation of Stat1 or physical interaction between GR and Stat1. The costimulatory effect of Dex was not dependent on a consensus glucocorticoid response element in the Stat1-responsive promoters, but required the DNA-binding and trans-activation functions of GR, and Dex-induced protein synthesis. GR activated the natural FcγRI promoter construct, and this response required both Stat1 and the Ets family transcription factor PU.1. Previously, physical association between GR and Stat5 has been shown to enhance Stat5-dependent and suppress GR-dependent transcription. The results shown here demonstrate a distinct, indirect mechanism of cross-modulation between cytokine and steroid receptor signaling that integrates Stat1 and GR pathways with cell type-specific PU.1 transcription factor in the regulation of FcγRI gene transcription.


Breast Cancer Research and Treatment | 2003

Regression of cutaneous tumor lesions in patients intratumorally injected with a recombinant single-chain antibody-toxin targeted to ErbB2/HER2

Marc Azemar; Sirus Djahansouzi; Elke Jäger; Christine Solbach; Mathias Schmidt; Alexander B. Maurer; Klaus Mross; Clemens Unger; Gunter von Minckwitz; Peter Dall; Bernd Groner; Winfried S. Wels

ScFv(FRP5)-ETA is a recombinant single-chain antibody-toxin with binding specificity for ErbB2/HER2. Previously potent antitumoral activity of the molecule against ErbB2 overexpressing tumor cells was demonstrated in vitro and in animal models. Here we report on the first application of scFv(FRP5)-ETA in human cancer patients summarizing case reports collected in four different clinical centers. Eleven patients suffering from metastatic breast and colorectal cancers and from malignant melanoma were treated on a compassionate-use basis by intratumoral injection of scFv(FRP5)-ETA into cutaneous lesions once daily for 7–10 days. Total daily doses ranged from 60 to 900 µg, and total doses per treatment cycle ranged from 0.6 to 6.0 mg. Treatment caused injected tumors to shrink in six of the 10 cases evaluated (60%). Complete regression of injected tumor nodules was accomplished in four patients (40%) and partial reduction in tumor size in another two patients (20%). Adverse reactions were restricted to local symptoms such as pain and inflammation at injection sites which were fully reversible. Only in one patient treated at the highest daily doses systemic liver toxicity of grade 2 was observed and treatment was discontinued on day 7. No hematologic, renal, and/or cardiovascular toxicities were noted. Our results demonstrate that local therapy with scFv(FRP5)-ETA can be effective against ErbB2 expressing tumors justifying further clinical development of this reagent.


Journal of Mammary Gland Biology and Neoplasia | 2006

The Biological Functions of the Versatile Transcription Factors STAT3 and STAT5 and New Strategies for their Targeted Inhibition

Sylvane Desrivières; Christian Kunz; Itamar Barash; Vida Vafaizadeh; Corina Borghouts; Bernd Groner

Signal transducers and activators of transcription (STATs) comprise a unique family of transcription factors, which transmit the interactions of cytokines, hormones and growth factors with their cell surface receptors into transcriptional programs. The mechanism of STAT activation has been well-established and comprises tyrosine phosphorylation, dimerization, nuclear translocation, binding to specific DNA response elements, recruitment of co-activators or co-repressors and transcriptional induction or repression of target genes. Gene deletion, microarrays, proteomics and chromatin immunoprecipitation experiments have revealed target genes with a broad range of functions regulated by STAT3 and STAT5. In the mammary gland, STAT5-induced genes contribute mainly to the prolactin dependent lobulo-alveolar development, whereas STAT3 induced genes control apoptosis during involution. Crucial effects have also been observed in other tissues. The germ line deletion of STAT3 or STAT5 causes early embryonal or perinatal lethality in mice. STAT5 is also required for proliferation of T- and B-cells and hematopoietic stem cell self-renewal. Deregulated STAT activity is often found associated with tumorigenesis and activated STATs seem to be limiting components in tumor cells. This review summarizes the functions of STAT3 and STAT5 in different cell types and the strategies that are used to counteract their action in tumor cells.


Stem Cells | 2010

Mammary epithelial reconstitution with gene-modified stem cells assigns roles to Stat5 in luminal alveolar cell fate decisions, differentiation, involution, and mammary tumor formation.

Vida Vafaizadeh; Petra Klemmt; Christian Brendel; Kristoffer Weber; Carmen Doebele; Kara L. Britt; Manuel Grez; Boris Fehse; Sylvane Desrivières; Bernd Groner

The mammary gland represents a unique model system to study gene functions in adult stem cells. Mammary stem cells (MaSCs) can regenerate a functional epithelium on transplantation into cleared fat pads. We studied the consequences of distinct genetic modifications of MaSCs on their repopulation and differentiation ability. The reconstitution of ductal trees was used as a stem cell selection procedure and the nearly quantitative lentiviral infection efficiency of the primary mammary epithelial cells (MECs) rendered the enrichment of MaSCs before their transplantation unnecessary. The repopulation frequency of transduced MaSCs was nearly 100% in immunodeficient recipients and the resulting transgenic ducts homogeneously expressed the virally encoded fluorescent marker proteins. Transplantation of a mixture of MECs, expressing different fluorescent proteins, resulted in a distinct pattern of ductal outgrowths originating from a small number of individually transduced MaSCs. We used genetically modified MECs to define multiple functions of Stat5 during mammary gland development and differentiation. Stat5‐downregulation in MaSCs did not affect primary ductal outgrowth, but impaired side branching and the emergence of mature alveolar cells from luminal progenitors during pregnancy. Conversely, the expression of a constitutively active variant of Stat5 (cS5‐F) caused epithelial hyperproliferation, thickening of the ducts and precocious, functional alveoli formation in virgin mice. Expression of cS5‐F also prevented involution and caused the formation of estrogen and progesterone receptor positive (ER+PR+) adenocarcinomas. The tumors expressed activated Stat5 and Stat3 and contained a small fraction of CD44+ cells, possibly indicative of cancer stem cells. STEM CELLS 2010;28:928–938

Collaboration


Dive into the Bernd Groner's collaboration.

Top Co-Authors

Avatar

Boris Brill

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Corina Borghouts

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Petra Klemmt

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Richard Moriggl

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge