Mitochondrial NIX Promotes Tumor Survival in the Hypoxic Niche of Glioblastoma

Cancer Res. 2019 Oct 15;79(20):5218-5232. doi: 10.1158/0008-5472.CAN-19-0198. Epub 2019 Sep 5.

Abstract

Cancer cells rely on mitochondrial functions to regulate key survival and death signals. How cancer cells regulate mitochondrial autophagy (mitophagy) in the tumor microenvironment as well as utilize mitophagy as a survival signal is still not well understood. Here, we elucidate a key survival mechanism of mitochondrial NIX-mediated mitophagy within the hypoxic region of glioblastoma, the most malignant brain tumor. NIX was overexpressed in the pseudopalisading cells that envelop the hypoxic-necrotic regions, and mitochondrial NIX expression was robust in patient-derived glioblastoma tumor tissues and glioblastoma stem cells. NIX was required for hypoxia and oxidative stress-induced mitophagy through NFE2L2/NRF2 transactivation. Silencing NIX impaired mitochondrial reactive oxygen species clearance, cancer stem cell maintenance, and HIF/mTOR/RHEB signaling pathways under hypoxia, resulting in suppression of glioblastoma survival in vitro and in vivo. Clinical significance of these findings was validated by the compelling association between NIX expression and poor outcome for patients with glioblastoma. Taken together, our findings indicate that the NIX-mediated mitophagic pathway may represent a key therapeutic target for solid tumors, including glioblastoma. SIGNIFICANCE: NIX-mediated mitophagy regulates tumor survival in the hypoxic niche of glioblastoma microenvironment, providing a potential therapeutic target for glioblastoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5218/F1.large.jpg.

Publication types

  • Research Support, N.I.H., Intramural

MeSH terms

  • Animals
  • Brain Neoplasms / metabolism*
  • Brain Neoplasms / pathology
  • Cell Hypoxia / physiology*
  • Glioblastoma / metabolism*
  • Glioblastoma / pathology
  • Glioma / metabolism
  • Glioma / pathology
  • Heterografts
  • Humans
  • Hypoxia-Inducible Factor 1 / physiology
  • Membrane Proteins / antagonists & inhibitors
  • Membrane Proteins / genetics
  • Membrane Proteins / physiology*
  • Mice
  • Mice, Inbred BALB C
  • Mice, SCID
  • Mitochondria / metabolism*
  • Mitophagy / physiology*
  • NF-E2-Related Factor 2 / metabolism
  • Neoplasm Proteins / antagonists & inhibitors
  • Neoplasm Proteins / genetics
  • Neoplasm Proteins / physiology*
  • Neoplastic Stem Cells / metabolism
  • Oxidative Stress
  • Proto-Oncogene Proteins / antagonists & inhibitors
  • Proto-Oncogene Proteins / genetics
  • Proto-Oncogene Proteins / physiology*
  • RNA Interference
  • RNA, Small Interfering / genetics
  • RNA, Small Interfering / pharmacology
  • Ras Homolog Enriched in Brain Protein / physiology
  • Reactive Oxygen Species / metabolism
  • Signal Transduction / physiology
  • TOR Serine-Threonine Kinases / physiology
  • Tumor Microenvironment
  • Tumor Suppressor Proteins / antagonists & inhibitors
  • Tumor Suppressor Proteins / genetics
  • Tumor Suppressor Proteins / physiology*

Substances

  • BNIP3L protein, human
  • Hypoxia-Inducible Factor 1
  • Membrane Proteins
  • NF-E2-Related Factor 2
  • NFE2L2 protein, human
  • Neoplasm Proteins
  • Proto-Oncogene Proteins
  • RHEB protein, human
  • RNA, Small Interfering
  • Ras Homolog Enriched in Brain Protein
  • Reactive Oxygen Species
  • Tumor Suppressor Proteins
  • MTOR protein, human
  • TOR Serine-Threonine Kinases