The TRPM7 channel reprograms cellular glycolysis to drive tumorigenesis and angiogenesis

Cell Death Dis. 2023 Mar 6;14(3):183. doi: 10.1038/s41419-023-05701-7.

Abstract

Cancer or endothelial cells preferably catabolize glucose through aerobic glycolysis rather than oxidative phosphorylation. Intracellular ionic signaling has been shown to regulate glucose metabolism, but the underlying ion channel has yet to be identified. RNA-seq, metabolomics and genetic assay revealed that the TRPM7 channel regulated cellular glycolysis. Deletion of TRPM7 suppressed cancer cell glycolysis and reduced the xenograft tumor burden. Deficiency of endothelial TRPM7 inhibited postnatal retinal angiogenesis in mice. Mechanistically, TRPM7 transcriptionally regulated the solute carrier family 2 member 3 (SLC2A3, also known as GLUT3) via Ca2+ influx-induced calcineurin activation. Furthermore, CREB-regulated transcription coactivator 2 (CRTC2) and CREB act downstream of calcineurin to relay Ca2+ signal to SLC2A3 transcription. Expression of the constitutively active CRTC2 or CREB in TRPM7 knockout cell normalized glycolytic metabolism and cell growth. The TRPM7 channel represents a novel regulator of glycolytic reprogramming. Inhibition of the TRPM7-dependent glycolysis could be harnessed for cancer therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Calcineurin
  • Carcinogenesis / genetics
  • Cell Transformation, Neoplastic
  • Endothelial Cells*
  • Glycolysis
  • Humans
  • Mice
  • Protein Serine-Threonine Kinases
  • TRPM Cation Channels* / genetics

Substances

  • Calcineurin
  • TRPM Cation Channels
  • TRPM7 protein, human
  • Protein Serine-Threonine Kinases
  • Trpm7 protein, mouse