Nature Communications (Aug 2023)

Integrin β3 directly inhibits the Gα13-p115RhoGEF interaction to regulate G protein signaling and platelet exocytosis

  • Yaping Zhang,
  • Xiaojuan Zhao,
  • Bo Shen,
  • Yanyan Bai,
  • Claire Chang,
  • Aleksandra Stojanovic,
  • Can Wang,
  • Andrew Mack,
  • Gary Deng,
  • Randal A. Skidgel,
  • Ni Cheng,
  • Xiaoping Du

DOI
https://doi.org/10.1038/s41467-023-40531-3
Journal volume & issue
Vol. 14, no. 1
pp. 1 – 12

Abstract

Read online

Abstract The integrins and G protein-coupled receptors are both fundamental in cell biology. The cross talk between these two, however, is unclear. Here we show that β3 integrins negatively regulate G protein-coupled signaling by directly inhibiting the Gα13-p115RhoGEF interaction. Furthermore, whereas β3 deficiency or integrin antagonists inhibit integrin-dependent platelet aggregation and exocytosis (granule secretion), they enhance G protein-coupled RhoA activation and integrin-independent secretion. In contrast, a β3-derived Gα13-binding peptide or Gα13 knockout inhibits G protein-coupled RhoA activation and both integrin-independent and dependent platelet secretion without affecting primary platelet aggregation. In a mouse model of myocardial ischemia/reperfusion injury in vivo, the β3-derived Gα13-binding peptide inhibits platelet secretion of granule constituents, which exacerbates inflammation and ischemia/reperfusion injury. These data establish crucial integrin-G protein crosstalk, providing a rationale for therapeutic approaches that inhibit exocytosis in platelets and possibly other cells without adverse effects associated with loss of cell adhesion.