Induction of autophagy by ARHI (DIRAS3) alters fundamental metabolic pathways in ovarian cancer models

Argentina Ornelas, Christopher R. McCullough, Zhen Lu, Niki M. Zacharias, Lindsay E. Kelderhouse, Joshua Gray, Hailing Yang, Brian J. Engel, Yan Wang, Weiqun Mao, Margie N. Sutton, Pratip K. Bhattacharya, Robert C. Bast, Steven W. Millward

Research output: Contribution to journalArticlepeer-review

21 Scopus citations

Abstract

Background: Autophagy is a bulk catabolic process that modulates tumorigenesis, therapeutic resistance, and dormancy. The tumor suppressor ARHI (DIRAS3) is a potent inducer of autophagy and its expression results in necroptotic cell death in vitro and tumor dormancy in vivo. ARHI is down-regulated or lost in over 60% of primary ovarian tumors yet is dramatically up-regulated in metastatic disease. The metabolic changes that occur during ARHI induction and their role in modulating death and dormancy are unknown. Methods: We employed Nuclear Magnetic Resonance (NMR)-based metabolomic strategies to characterize changes in key metabolic pathways in both cell culture and xenograft models of ARHI expression and autophagy. These pathways were further interrogated by cell-based immunofluorescence imaging, tracer uptake studies, targeted metabolic inhibition, and in vivo PET/CT imaging. Results: Induction of ARHI in cell culture models resulted in an autophagy-dependent increase in lactate production along with increased glucose uptake and enhanced sensitivity to glycolytic inhibitors. Increased uptake of glutamine was also dependent on autophagy and dramatically sensitized cultured ARHI-expressing ovarian cancer cell lines to glutaminase inhibition. Induction of ARHI resulted in a reduction in mitochondrial respiration, decreased mitochondrial membrane potential, and decreased Tom20 staining suggesting an ARHI-dependent loss of mitochondrial function. ARHI induction in mouse xenograft models resulted in an increase in free amino acids, a transient increase in [18F]-FDG uptake, and significantly altered choline metabolism. Conclusions: ARHI expression has previously been shown to trigger autophagy-associated necroptosis in cell culture. In this study, we have demonstrated that ARHI expression results in decreased cellular ATP/ADP, increased oxidative stress, and decreased mitochondrial function. While this bioenergetic shock is consistent with programmed necrosis, our data indicates that the accompanying up-regulation of glycolysis and glutaminolysis is autophagy-dependent and serves to support cell viability rather than facilitate necroptotic cell death. While the mechanistic basis for metabolic up-regulation following ARHI induction is unknown, our preliminary data suggest that decreased mitochondrial function and increased metabolic demand may play a role. These alterations in fundamental metabolic pathways during autophagy-associated necroptosis may provide the basis for new therapeutic strategies for the treatment of dormant ovarian tumors.

Original languageEnglish (US)
Article number824
JournalBMC cancer
Volume16
Issue number1
DOIs
StatePublished - Oct 26 2016

Keywords

  • ARHI
  • Autophagy
  • Glutaminolysis
  • Metabolism
  • NMR
  • Necroptosis
  • Ovarian cancer

ASJC Scopus subject areas

  • Genetics
  • Oncology
  • Cancer Research

MD Anderson CCSG core facilities

  • Research Animal Support Facility
  • Small Animal Imaging Facility

Fingerprint

Dive into the research topics of 'Induction of autophagy by ARHI (DIRAS3) alters fundamental metabolic pathways in ovarian cancer models'. Together they form a unique fingerprint.

Cite this