Affiliations 

  • 1 Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
  • 2 Department of Basic Medical Sciences, University of Arizona, Phoenix, AZ 85004, USA
  • 3 Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
  • 4 Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
  • 5 Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
  • 6 Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 909 S Wolcott Avenue, Chicago, IL 60612, USA
  • 7 Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan
  • 8 Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea. Electronic address: shinehr@skku.edu
  • 9 Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 909 S Wolcott Avenue, Chicago, IL 60612, USA. Electronic address: sangging@uic.edu
Stem Cell Reports, 2019 03 05;12(3):597-610.
PMID: 30799273 DOI: 10.1016/j.stemcr.2019.01.017

Abstract

The relationship between diabetes and endothelial dysfunction remains unclear, particularly the association with pathological activation of calpain, an intracellular cysteine protease. Here, we used human induced pluripotent stem cells-derived endothelial cells (iPSC-ECs) to investigate the effects of diabetes on vascular health. Our results indicate that iPSC-ECs exposed to hyperglycemia had impaired autophagy, increased mitochondria fragmentation, and was associated with increased calpain activity. In addition, hyperglycemic iPSC-ECs had increased susceptibility to cell death when subjected to a secondary insult-simulated ischemia-reperfusion injury (sIRI). Importantly, calpain inhibition restored autophagy and reduced mitochondrial fragmentation, concurrent with maintenance of ATP production, normalized reactive oxygen species levels and reduced susceptibility to sIRI. Using a human iPSC model of diabetic endotheliopathy, we demonstrated that restoration of autophagy and prevention of mitochondrial fragmentation via calpain inhibition improves vascular integrity. Our human iPSC-EC model thus represents a valuable platform to explore biological mechanisms and new treatments for diabetes-induced endothelial dysfunction.

* Title and MeSH Headings from MEDLINE®/PubMed®, a database of the U.S. National Library of Medicine.