Oncotarget

Research Papers:

The BRG1 chromatin remodeling enzyme links cancer cell metabolism and proliferation

Qiong Wu, Pasil Madany, Jason R. Dobson, Jake M. Schnabl, Soni Sharma, Tara C. Smith, Andre J. van Wijnen, Janet L. Stein, Jane B. Lian, Gary S. Stein, Rohini Muthuswami, Anthony N. Imbalzano _ and Jeffrey A. Nickerson

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:38270-38281. https://doi.org/10.18632/oncotarget.9505

Metrics: PDF 2624 views  |   HTML 2684 views  |   ?  


Abstract

Qiong Wu1, Pasil Madany1, Jason R. Dobson1, Jake M. Schnabl1, Soni Sharma2, Tara C. Smith1 Andre J. van Wijnen3, Janet L. Stein4, Jane B. Lian4, Gary S. Stein4, Rohini Muthuswami2, Anthony N. Imbalzano1, Jeffrey A. Nickerson1

1Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA

2School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India

3Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA

4Department of Biochemistry and Vermont Cancer Center for Basic and Translational Research, University of Vermont College of Medicine, Burlington, WA, USA

Correspondence to:

Anthony N. Imbalzano, email: [email protected]

Jeffrey A. Nickerson, email: [email protected]

Keywords: breast cancer, metabolism, lipogenesis, gene regulation, BRG1

Received: January 07, 2016     Accepted: May 01, 2016     Published: May 20, 2016

ABSTRACT

Cancer cells reprogram cellular metabolism to meet the demands of growth. Identification of the regulatory machinery that regulates cancer-specific metabolic changes may open new avenues for anti-cancer therapeutics. The epigenetic regulator BRG1 is a catalytic ATPase for some mammalian SWI/SNF chromatin remodeling enzymes. BRG1 is a well-characterized tumor suppressor in some human cancers, but is frequently overexpressed without mutation in other cancers, including breast cancer. Here we demonstrate that BRG1 upregulates de novo lipogenesis and that this is crucial for cancer cell proliferation. Knockdown of BRG1 attenuates lipid synthesis by impairing the transcription of enzymes catalyzing fatty acid and lipid synthesis. Remarkably, exogenous addition of palmitate, the key intermediate in fatty acid synthesis, rescued the cancer cell proliferation defect caused by BRG1 knockdown. Our work suggests that targeting BRG1 to reduce lipid metabolism and, thereby, to reduce proliferation, has promise for epigenetic therapy in triple negative breast cancer.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 9505