Login / Signup

OMA1 reprograms metabolism under hypoxia to promote colorectal cancer development.

Zhida WuMeiling ZuoLing ZengKaisa CuiBing LiuChaojun YanLi ChenJun DongFugen ShangguanWanglai HuHe HeBin LuZhiyin Song
Published in: EMBO reports (2020)
Many cancer cells maintain enhanced aerobic glycolysis due to irreversible defective mitochondrial oxidative phosphorylation (OXPHOS). This phenomenon, known as the Warburg effect, is recently challenged because most cancer cells maintain OXPHOS. However, how cancer cells coordinate glycolysis and OXPHOS remains largely unknown. Here, we demonstrate that OMA1, a stress-activated mitochondrial protease, promotes colorectal cancer development by driving metabolic reprogramming. OMA1 knockout suppresses colorectal cancer development in AOM/DSS and xenograft mice models of colorectal cancer. OMA1-OPA1 axis is activated by hypoxia, increasing mitochondrial ROS to stabilize HIF-1α, thereby promoting glycolysis in colorectal cancer cells. On the other hand, under hypoxia, OMA1 depletion promotes accumulation of NDUFB5, NDUFB6, NDUFA4, and COX4L1, supporting that OMA1 suppresses OXPHOS in colorectal cancer. Therefore, our findings support a role for OMA1 in coordination of glycolysis and OXPHOS to promote colorectal cancer development and highlight OMA1 as a potential target for colorectal cancer therapy.
Keyphrases
  • oxidative stress
  • endothelial cells
  • cancer therapy
  • signaling pathway
  • type diabetes
  • drug delivery
  • metabolic syndrome
  • adipose tissue
  • reactive oxygen species
  • risk assessment
  • high fat diet induced