Nrf2 protects against diabetic dysfunction of endothelial progenitor cells via regulating cell senescence

Int J Mol Med. 2018 Sep;42(3):1327-1340. doi: 10.3892/ijmm.2018.3727. Epub 2018 Jun 11.

Abstract

Diabetes is associated with an increased risk of cardiovascular disease. A decrease in the number and functionality of endothelial progenitor cells (EPCs) leads to reduced endothelial repair and the development of cardiovascular disease. The aim of the present study was to explore the effect and underlying mechanisms of nuclear factor erythroid 2‑related factor 2 (Nrf2) on EPC dysfunction caused by diabetic mellitus. The biological functions of EPCs in streptozotocin‑induced diabetic mice were evaluated, including migration, proliferation, angiogenesis and the secretion of vascular endothelial growth factor (VEGF), stromal‑derived growth factor (SDF) and nitric oxide (NO). Oxidative stress levels in diabetic EPCs were also assessed by detecting intracellular reactive oxygen species (ROS), superoxide dismutase (SOD) and malondialdehyde (MDA). EPC senescence was evaluated by measuring p16 and b‑gal expression and observing the senescence‑associated secretory phenotype. In addition, the function of EPCs and level of oxidative stress were assessed following Nrf2 silencing or activation. Nrf2 silencing resulted in a decrease of EPC biological functions, accelerated cell senescence and increased oxidative stress, as indicated by ROS and MDA upregulation accompanied with decreased SOD activity. Furthermore, Nrf2 silencing inhibited migration, proliferation and secretion in EPCs, while it increased oxidative stress and cell senescence. Nrf2 activation protected diabetic EPCs against the effects of oxidative stress and cell senescence, ameliorating the biological dysfunction of EPCs derived from mice with diabetes. In conclusion, Nrf2 overexpression protected against oxidative stress‑induced functional damage in EPCs derived from diabetic mice by regulating cell senescence.

MeSH terms

  • Animals
  • Cell Movement / physiology
  • Cells, Cultured
  • Cellular Senescence / physiology
  • Diabetes Mellitus, Experimental / genetics
  • Diabetes Mellitus, Experimental / metabolism*
  • Endothelial Progenitor Cells / cytology*
  • Endothelial Progenitor Cells / metabolism*
  • Male
  • Malondialdehyde / metabolism
  • Mice
  • Mice, Inbred C57BL
  • NF-E2-Related Factor 2 / metabolism*
  • Nitric Oxide / metabolism
  • Oxidative Stress / physiology
  • Reactive Oxygen Species / metabolism
  • Superoxide Dismutase / metabolism
  • Vascular Endothelial Growth Factor A / metabolism

Substances

  • NF-E2-Related Factor 2
  • Reactive Oxygen Species
  • Vascular Endothelial Growth Factor A
  • Nitric Oxide
  • Malondialdehyde
  • Superoxide Dismutase