m6A and m5C modification of GPX4 facilitates anticancer immunity via STING activation

Cell Death Dis. 2023 Dec 8;14(12):809. doi: 10.1038/s41419-023-06241-w.

Abstract

Cancer immunotherapy is arguably the most rapidly advancing realm of cancer treatment. Glutathione peroxidase 4 (GPX4) has emerged as the vital enzyme to prevent lipid peroxidation and maintain cellular redox homeostasis. However, the mechanism of GPX4 in the regulation of cancer immunotherapy of colon adenocarcinoma (COAD) are incompletely understood. In pan-cancer analysis, we found that GPX4 showed remarkably upregulated expression and exhibited significant association with overall survival in multiple cancer types, especially COAD. Furthermore, upregulated GPX4 expression was positively correlated with increased immune cells infiltration and enhanced expression of immunomodulators. Mechanistically, RBM15B- and IGFBP2-mediated N6-methyladenosine (m6A) modification and NSUN5-mediated 5-methylcytosine (m5C) modification of GPX4 facilitated anticancer immunity via activation of cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (STING) signaling by maintaining redox homeostasis in COAD. The risk model and nomogram model constructed based on the GPX4-derived genes further confirmed the prognostic and treatment-guiding value of GPX4. In all, our study demonstrated that m6A and m5C modification of GPX4 may be a promising target for cancer immunotherapy via activating the cGAS-STING signaling pathway in COAD.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenocarcinoma* / genetics
  • Adenocarcinoma* / immunology
  • Colonic Neoplasms* / genetics
  • Colonic Neoplasms* / immunology
  • Humans
  • Membrane Proteins* / biosynthesis
  • Nucleotidyltransferases / genetics
  • Phospholipid Hydroperoxide Glutathione Peroxidase* / genetics

Substances

  • 6-methyladenine
  • Nucleotidyltransferases
  • Phospholipid Hydroperoxide Glutathione Peroxidase
  • GPX4 protein, human
  • STING1 protein, human
  • Membrane Proteins