Oncotarget

Priority Research Papers:

This article has been corrected. Correction in: Oncotarget. 2021; 12:2305-2306.

Tumor suppressor RARRES1 links tubulin deglutamylation to mitochondrial metabolism and cell survival

Sara Maimouni, Mi-Hye Lee, You-Me Sung, Michael Hall, Arpita Roy, Chokri Ouaari, Yoo-Seok Hwang, Justin Spivak, Eric Glasgow, Matthew Swift, Jay Patel, Amrita Cheema, Deepak Kumar and Stephen Byers _

PDF  |  Full Text  |  Supplementary Files  |  How to cite

Oncotarget. 2019; 10:1606-1624. https://doi.org/10.18632/oncotarget.26600

Metrics: PDF 1678 views  |   Full Text 2773 views  |   ?  


Abstract

Sara Maimouni2,*, Mi-Hye Lee1,*, You-Me Sung1,*, Michael Hall1, Arpita Roy3, Chokri Ouaari1,3, Yoo-Seok Hwang4, Justin Spivak1, Eric Glasgow1, Matthew Swift1, Jay Patel1, Amrita Cheema1, Deepak Kumar3 and Stephen Byers1,2

1 Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA

2 Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA

3 University of the District of Columbia, Washington, DC, USA

4 Cancer & Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, MD, USA

* These authors have contributed equally to this article

Correspondence to:

Stephen Byers, email: [email protected]

Keywords: retinoic acid signaling; RARRES1; metabolic reprogramming; PTM tubulin; drug resistance

Received: October 09, 2018    Accepted: December 04, 2018    Published: February 26, 2019

Abstract

RARRES1, a retinoic acid regulated carboxypeptidase inhibitor associated with fatty acid metabolism, stem cell differentiation and tumorigenesis is among the most commonly methylated loci in multiple cancers but has no known mechanism of action. Here we show that RARRES1 interaction with cytoplasmic carboxypeptidase 2 (CCP2) inhibits tubulin deglutamylation, which in turn regulates the mitochondrial voltage dependent anion channel (VDAC1), mitochondrial membrane potential, AMPK activation, energy balance and metabolically reprograms cells and zebrafish to a more energetic and anabolic phenotype. Depletion of RARRES1 also increases expression of stem cell markers, promotes anoikis, anchorage independent growth and insensitivity to multiple apoptotic stimuli. As depletion of CCP2 or inhibition of VDAC1 reverses the effects of RARRES1 depletion on energy balance and cell survival we conclude that RARRES1 modulation of CCP2-modulated tubulin-mitochondrial VDAC1 interactions is a fundamental regulator of cancer and stem cell metabolism and survival.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 26600