Aberrant hyper-expression of the RNA binding protein GIGYF2 in endothelial cells modulates vascular aging and function

Redox Biol. 2023 Sep:65:102824. doi: 10.1016/j.redox.2023.102824. Epub 2023 Jul 24.

Abstract

Vascular endothelial cells (ECs) senescence plays a crucial role in vascular aging that promotes the initiation and progression of cardiovascular disease. The mutation of Grb10-interacting GYF protein 2 (GIGYF2) is strongly associated with the pathogenesis of aging-related diseases, whereas its role in regulating ECs senescence and dysfunction still remains elusive. In this study, we found aberrant hyperexpression of GIGYF2 in senescent human ECs and aortas of old mice. Silencing GIGYF2 in senescent ECs suppressed eNOS-uncoupling, senescence, and endothelial dysfunction. Conversely, in nonsenescent cells, overexpressing GIGYF2 promoted eNOS-uncoupling, cellular senescence, endothelial dysfunction, and activation of the mTORC1-SK61 pathway, which were ablated by rapamycin or antioxidant N-Acetyl-l-cysteine (NAC). Transcriptome analysis revealed that staufen double-stranded RNA binding protein 1 (STAU1) is remarkably downregulated in the GIGYF2-depleted ECs. STAU1 depletion significantly attenuated GIGYF2-induced cellular senescence, dysfunction, and inflammation in young ECs. Furthermore, we disclosed that GIGYF2 acting as an RNA binding protein (RBP) enhances STAU1 mRNA stability, and that the intron region of the late endosomal/lysosomal adaptor MAPK and mTOR activator 4 (LAMTOR4) could bind to STAU1 protein to upregulate LAMTOR4 expression. Immunofluorescence staining showed that GIGYF2 overexpression promoted the translocation of mTORC1 to lysosome. In the mice model, GIGYF2flox/flox Cdh-Cre+ mice protected aged mice from aging-associated vascular endothelium-dependent relaxation and arterial stiffness. Our work discloses that GIGYF2 serving as an RBP enhances the mRNA stability of STAU1 that upregulates LAMTOR4 expression through binding with its intron region, which activates the mTORC1-S6K1 signaling via recruitment of mTORC1 to the lysosomal membrane, ultimately leading to ECs senescence, dysfunction, and vascular aging. Disrupting the GIGYF2-STAU1-mTORC1 signaling cascade may represent a promising therapeutic approach against vascular aging and aging-related cardiovascular diseases.

Keywords: Endothelial cell senescence; Endothelial dysfunction; Signaling; Vascular aging; eNOS uncoupling.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Aging* / genetics
  • Aging* / metabolism
  • Animals
  • Carrier Proteins / genetics
  • Carrier Proteins / metabolism
  • Cellular Senescence / physiology
  • Cytoskeletal Proteins / metabolism
  • Endothelial Cells* / metabolism
  • Guanine Nucleotide Exchange Factors / metabolism
  • Humans
  • Mechanistic Target of Rapamycin Complex 1
  • Mice
  • RNA-Binding Proteins / genetics
  • RNA-Binding Proteins / metabolism

Substances

  • Carrier Proteins
  • Cytoskeletal Proteins
  • GIGYF2 protein, human
  • Guanine Nucleotide Exchange Factors
  • LAMTOR4 protein, human
  • Mechanistic Target of Rapamycin Complex 1
  • RNA-Binding Proteins
  • STAU1 protein, human
  • GIGYF2 protein, mouse