Oncotarget

Research Papers:

Caveolin-1 regulates cancer cell metabolism via scavenging Nrf2 and suppressing MnSOD-driven glycolysis

Peter C. Hart _, Bianca A. Ratti, Mao Mao, Kristine Ansenberger-Fricano, Ayesha N. Shajahan-Haq, Angela L. Tyner, Richard D. Minshall and Marcelo G. Bonini

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:308-322. https://doi.org/10.18632/oncotarget.5687

Metrics: PDF 3167 views  |   HTML 4309 views  |   ?  


Abstract

Peter C. Hart1,2, Bianca A. Ratti1,7, Mao Mao1, Kristine Ansenberger-Fricano1, Ayesha N. Shajahan-Haq6, Angela L. Tyner3, Richard D. Minshall4,5, Marcelo G. Bonini1,2,4,7

1Department of Medicine, University of Illinois College of Medicine at Chicago, Chicago, IL, USA

2Department of Pathology, University of Illinois College of Medicine at Chicago, Chicago, IL, USA

3Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine at Chicago, Chicago, IL, USA

4Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, USA

5Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, USA

6Department of Oncology, Georgetown University, Medical Center, Washington, D.C., USA

7Programa de Biociencias Aplicadas a Farmacia (PBF) Universidade Estadual de Maringa, Maringa, PR, Brazil

Correspondence to:

Marcelo G. Bonini, e-mail: [email protected]

Keywords: Caveolin-1, MnSOD, Nrf2, breast cancer, tumor progression

Received: March 10, 2015     Accepted: October 16, 2015     Published: October 26, 2015

ABSTRACT

Aerobic glycolysis is an indispensable component of aggressive cancer cell metabolism. It also distinguishes cancer cells from most healthy cell types in the body. Particularly for this reason, targeting the metabolism to improve treatment outcomes has long been perceived as a potentially valuable strategy. In practice, however, our limited knowledge of why and how metabolic reprogramming occurs has prevented progress towards therapeutic interventions that exploit the metabolic peculiarities of tumors. We recently described that in breast cancer, MnSOD upregulation is both necessary and sufficient to activate glycolysis. Here, we focused on determining the molecular mechanisms of MnSOD upregulation. We found that Caveolin-1 (Cav-1) is a central component of this mechanism due to its suppressive effects of NF-E2-related factor 2 (Nrf2), a transcription factor upstream of MnSOD. In transformed MCF10A(Er/Src) cells, Cav-1 loss preceded the activation of Nrf2 and its induction of MnSOD expression. Consistently, with previous observations, MnSOD expression secondary to Nrf2 activation led to an increase in the glycolytic rate dependent on mtH2O2 production and the activation of AMPK. Moreover, rescue of Cav-1 expression in a breast cancer cell line (MCF7) suppressed Nrf2 and reduced MnSOD expression. Experimental data were reinforced by epidemiologic nested case-control studies showing that Cav-1 and MnSOD are inversely expressed in cases of invasive ductal carcinoma, with low Cav-1 and high MnSOD expression being associated with lower 5-year survival rates and molecular subtypes with poorest prognosis.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 5687