cGAS Mediates Inflammation by Polarizing Macrophages to M1 Phenotype via the mTORC1 Pathway

J Immunol. 2023 Apr 15;210(8):1098-1107. doi: 10.4049/jimmunol.2200351.

Abstract

Cyclic GMP-AMP synthase (cGAS), as a cytosolic DNA sensor, plays a crucial role in antiviral immunity, and its overactivation induces excess inflammation and tissue damage. Macrophage polarization is critically involved in inflammation; however, the role of cGAS in macrophage polarization during inflammation remains unclear. In this study, we demonstrated that cGAS was upregulated in the LPS-induced inflammatory response via the TLR4 pathway, and cGAS signaling was activated by mitochondria DNA in macrophages isolated from C57BL/6J mice. We further demonstrated that cGAS mediated inflammation by acting as a macrophage polarization switch, which promoted peritoneal macrophages and the bone marrow-derived macrophages to the inflammatory phenotype (M1) via the mitochondrial DNA-mTORC1 pathway. In vivo studies verified that deletion of Cgas alleviated sepsis-induced acute lung injury by promoting macrophages to shift from the M1 phenotype to the M2 phenotype. In conclusion, our study demonstrated that cGAS mediated inflammation by regulating macrophage polarization through the mTORC1 pathway, and it further provided a potential therapeutic strategy for inflammatory diseases, especially sepsis-induced acute lung injury.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acute Lung Injury*
  • Animals
  • DNA, Mitochondrial / metabolism
  • Inflammation
  • Macrophages* / metabolism
  • Mechanistic Target of Rapamycin Complex 1* / metabolism
  • Mice
  • Mice, Inbred C57BL
  • Nucleotidyltransferases* / genetics
  • Nucleotidyltransferases* / metabolism
  • Phenotype
  • Sepsis*

Substances

  • DNA, Mitochondrial
  • Nucleotidyltransferases
  • cGAS protein, mouse
  • Mechanistic Target of Rapamycin Complex 1