Nuclear mTOR acts as a transcriptional integrator of the androgen signaling pathway in prostate cancer

  1. Vincent Giguère1,8,9,10
  1. 1Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A3, Canada;
  2. 2Urologic Oncology Research Group, Cancer Research Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, Québec H4A 3J1, Canada;
  3. 3Génome Québec Innovation Centre, McGill University, Montréal, Québec H3A 0G1, Canada;
  4. 4Department of Human Genetics, McGill University, Montréal, Québec H3A 1A3, Canada;
  5. 5Department of Pathology, McGill University and MUHC, Montréal, Québec H4A 3J1, Canada;
  6. 6Department of Surgery (Urology), McGill University and MUHC, Montréal, Québec H4A 3J1, Canada;
  7. 7Department of Oncology, McGill University and MUHC, Montréal, Québec H4A 3J1, Canada;
  8. 8Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada;
  9. 9Department of Oncology, McGill University, Montréal, Québec H3G 1Y6, Canada;
  10. 10Department of Biochemistry, McGill University, Montréal, Québec H3G 1Y6, Canada
  1. Corresponding author: vincent.giguere{at}mcgill.ca

Abstract

Androgen receptor (AR) signaling reprograms cellular metabolism to support prostate cancer (PCa) growth and survival. Another key regulator of cellular metabolism is mTOR, a kinase found in diverse protein complexes and cellular localizations, including the nucleus. However, whether nuclear mTOR plays a role in PCa progression and participates in direct transcriptional cross-talk with the AR is unknown. Here, via the intersection of gene expression, genomic, and metabolic studies, we reveal the existence of a nuclear mTOR–AR transcriptional axis integral to the metabolic rewiring of PCa cells. Androgens reprogram mTOR–chromatin associations in an AR-dependent manner in which activation of mTOR-dependent metabolic gene networks is essential for androgen-induced aerobic glycolysis and mitochondrial respiration. In models of castration-resistant PCa cells, mTOR was capable of transcriptionally regulating metabolic gene programs in the absence of androgens, highlighting a potential novel castration resistance mechanism to sustain cell metabolism even without a functional AR. Remarkably, we demonstrate that increased mTOR nuclear localization is indicative of poor prognosis in patients, with the highest levels detected in castration-resistant PCa tumors and metastases. Identification of a functional mTOR targeted multigene signature robustly discriminates between normal prostate tissues, primary tumors, and hormone refractory metastatic samples but is also predictive of cancer recurrence. This study thus underscores a paradigm shift from AR to nuclear mTOR as being the master transcriptional regulator of metabolism in PCa.

Keywords

Footnotes

  • Received April 3, 2017.
  • Accepted June 19, 2017.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance