Oncotarget

Research Papers:

Sumoylation of HDAC2 promotes NF-κB-dependent gene expression

Tobias Wagner, Nicole Kiweler, Katharina Wolff, Shirley K. Knauer, André Brandl, Peter Hemmerich, Jan-Hermen Dannenberg, Thorsten Heinzel, Günter Schneider and Oliver H. Krämer _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:7123-7135. https://doi.org/10.18632/oncotarget.3344

Metrics: PDF 2729 views  |   HTML 3305 views  |   ?  


Abstract

Tobias Wagner1, Nicole Kiweler2, Katharina Wolff1, Shirley K. Knauer3, André Brandl1, Peter Hemmerich4, Jan-Hermen Dannenberg5, Thorsten Heinzel1, Günter Schneider6 and Oliver H. Krämer2

1 Centre for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Jena, Germany

2 Department of Toxicology, University Medical Center, Mainz, Germany

3 Centre for Medical Biotechnology, Molecular Biology II, University of Duisburg-Essen, Essen, Germany

4 Leibniz-Institute for Age Research, Fritz-Lipmann-Institute, Jena, Germany

5 Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, The Netherlands

6 Klinikum rechts der Isar, II. Medizinische Klinik, Technische Universität München, München, Germany

Correspondence:

Oliver H. Krämer, email:

Keywords: Histone deacetylase 2, p53, p65, RSK1, SUMO

Received: January 02, 2015 Accepted: January 04, 2015 Published: January 21, 2015

Abstract

The transcription factor nuclear factor-κB (NF-κB) is crucial for the maintenance of homeostasis. It is incompletely understood how nuclear NF-κB and the crosstalk of NF-κB with other transcription factors are controlled. Here, we demonstrate that the epigenetic regulator histone deacetylase 2 (HDAC2) activates NF-κB in transformed and primary cells. This function depends on both, the catalytic activity and an intact HDAC2 sumoylation motif. Several mechanisms account for the induction of NF-κB through HDAC2. The expression of wild-type HDAC2 can increase the nuclear presence of NF-κB. In addition, the ribosomal S6 kinase 1 (RSK1) and the tumor suppressor p53 contribute to the regulation of NF-κB by HDAC2. Moreover, TP53 mRNA expression is positively regulated by wild-type HDAC2 but not by sumoylation-deficient HDAC2. Thus, sumoylation of HDAC2 integrates NF-κB signaling involving p53 and RSK1. Since HDAC2-dependent NF-κB activity protects colon cancer cells from genotoxic stress, our data also suggest that high HDAC2 levels, which are frequently found in tumors, are linked to chemoresistance. Accordingly, inhibitors of NF-κB and of the NF-κB/p53-regulated anti-apoptotic protein survivin significantly sensitize colon carcinoma cells expressing wild-type HDAC2 to apoptosis induced by the genotoxin doxorubicin. Hence, the HDAC2-dependent signaling node we describe here may offer an interesting therapeutic option.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 3344