Oncotarget

Research Papers:

The histone demethylase KDM3A regulates the transcriptional program of the androgen receptor in prostate cancer cells

Stephen Wilson _, Lingling Fan, Natasha Sahgal, Jianfei Qi and Fabian V. Filipp

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:30328-30343. https://doi.org/10.18632/oncotarget.15681

Metrics: PDF 4520 views  |   HTML 13847 views  |   ?  


Abstract

Stephen Wilson1, Lingling Fan2, Natasha Sahgal3, Jianfei Qi2, Fabian V. Filipp1

1Systems Biology and Cancer Metabolism, Program for Quantitative Systems Biology, University of California Merced, Merced, CA, USA

2Department of Biochemistry and Molecular Biology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA

3Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom

Correspondence to:

Fabian V. Filipp, email: [email protected]

Keywords: cancer systems biology, epigenomics, ChIP-Seq, oncogene, prostate cancer

Received: July 18, 2016    Accepted: September 09, 2016    Published: March 03, 2017

ABSTRACT

The lysine demethylase 3A (KDM3A, JMJD1A or JHDM2A) controls transcriptional networks in a variety of biological processes such as spermatogenesis, metabolism, stem cell activity, and tumor progression. We matched transcriptomic and ChIP-Seq profiles to decipher a genome-wide regulatory network of epigenetic control by KDM3A in prostate cancer cells. ChIP-Seq experiments monitoring histone 3 lysine 9 (H3K9) methylation marks show global histone demethylation effects of KDM3A. Combined assessment of histone demethylation events and gene expression changes presented major transcriptional activation suggesting that distinct oncogenic regulators may synergize with the epigenetic patterns by KDM3A. Pathway enrichment analysis of cells with KDM3A knockdown prioritized androgen signaling indicating that KDM3A plays a key role in regulating androgen receptor activity. Matched ChIP-Seq and knockdown experiments of KDM3A in combination with ChIP-Seq of the androgen receptor resulted in a gain of H3K9 methylation marks around androgen receptor binding sites of selected transcriptional targets in androgen signaling including positive regulation of KRT19, NKX3-1, KLK3, NDRG1, MAF, CREB3L4, MYC, INPP4B, PTK2B, MAPK1, MAP2K1, IGF1, E2F1, HSP90AA1, HIF1A, and ACSL3. The cancer systems biology analysis of KDM3A-dependent genes identifies an epigenetic and transcriptional network in androgen response, hypoxia, glycolysis, and lipid metabolism. Genome-wide ChIP-Seq data highlights specific gene targets and the ability of epigenetic master regulators to control oncogenic pathways and cancer progression.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 15681