Cell Death and Disease (Jul 2021)

HO-1 nuclear accumulation and interaction with NPM1 protect against stress-induced endothelial senescence independent of its enzymatic activity

  • Wenwei Luo,
  • Jingyan Li,
  • Ziqing Li,
  • Tong Lin,
  • Lili Zhang,
  • Wanqi Yang,
  • Yanqi Mai,
  • Ruiming Liu,
  • Meiting Chen,
  • Chunmei Dai,
  • Hanwei Yang,
  • Jing Lu,
  • Hong Li,
  • Guimei Guan,
  • Min Huang,
  • Peiqing Liu,
  • Zhuoming Li

DOI
https://doi.org/10.1038/s41419-021-04035-6
Journal volume & issue
Vol. 12, no. 8
pp. 1 – 13

Abstract

Read online

Abstract Heme oxygenase-1 (HO-1) has attracted accumulating attention for its antioxidant enzymatic activity. However, the exact regulatory role of its non-enzymatic activity in the cardiovascular system remains unaddressed. Here, we show that HO-1 was accumulated in the nuclei of stress-induced senescent endothelial cells, and conferred protection against endothelial senescence independent of its enzymatic activity. Overexpression of ΔHO-1, a truncated HO-1 without transmembrane segment (TMS), inhibited H2O2-induced endothelial senescence. Overexpression of ΔHO-1H25A, the catalytically inactive form of ΔHO-1, also exhibited anti-senescent effect. In addition, infection of recombinant adenovirus encoding ΔHO-1 with three nuclear localization sequences (NLS), alleviated endothelial senescence induced by knockdown of endogenous HO-1 by CRISPR/Cas9. Moreover, repression of HO-1 nuclear translocation by silencing of signal peptide peptidase (SPP), which is responsible for enzymatic cleavage of the TMS of HO-1, exacerbated endothelial senescence. Mechanistically, nuclear HO-1 interacted with NPM1 N-terminal portion, prevented NPM1 translocation from nucleolus to nucleoplasm, thus disrupted NPM1/p53/MDM2 interactions and inhibited p53 activation by NPM1, finally resisted endothelial senescence. This study provides a novel understanding of HO-1 as a promising therapeutic strategy for vascular senescence-related cardiovascular diseases.