Enhanced Therapeutic and Long-Term Dynamic Vascularization Effects of Human Pluripotent Stem Cell-Derived Endothelial Cells Encapsulated in a Nanomatrix Gel

Circulation. 2017 Nov 14;136(20):1939-1954. doi: 10.1161/CIRCULATIONAHA.116.026329. Epub 2017 Sep 29.

Abstract

Background: Human pluripotent stem cell (hPSC)-derived endothelial cells (ECs) have limited clinical utility because of undefined components in the differentiation system and poor cell survival in vivo. Here, we aimed to develop a fully defined and clinically compatible system to differentiate hPSCs into ECs. Furthermore, we aimed to enhance cell survival, vessel formation, and therapeutic potential by encapsulating hPSC-ECs with a peptide amphiphile (PA) nanomatrix gel.

Methods: We induced differentiation of hPSCs into the mesodermal lineage by culturing on collagen-coated plates with a glycogen synthase kinase 3β inhibitor. Next, vascular endothelial growth factor, endothelial growth factor, and basic fibroblast growth factor were added for endothelial lineage differentiation, followed by sorting for CDH5 (VE-cadherin). We constructed an extracellular matrix-mimicking PA nanomatrix gel (PA-RGDS) by incorporating the cell adhesive ligand Arg-Gly-Asp-Ser (RGDS) and a matrix metalloproteinase-2-degradable sequence. We then evaluated whether the encapsulation of hPSC-CDH5+ cells in PA-RGDS could enhance long-term cell survival and vascular regenerative effects in a hind-limb ischemia model with laser Doppler perfusion imaging, bioluminescence imaging, real-time reverse transcription-polymerase chain reaction, and histological analysis.

Results: The resultant hPSC-derived CDH5+ cells (hPSC-ECs) showed highly enriched and genuine EC characteristics and proangiogenic activities. When injected into ischemic hind limbs, hPSC-ECs showed better perfusion recovery and higher vessel-forming capacity compared with media-, PA-RGDS-, or human umbilical vein EC-injected groups. However, the group receiving the PA-RGDS-encapsulated hPSC-ECs showed better perfusion recovery, more robust and longer cell survival (> 10 months), and higher and prolonged angiogenic and vascular incorporation capabilities than the bare hPSC-EC-injected group. Surprisingly, the engrafted hPSC-ECs demonstrated previously unknown sustained and dynamic vessel-forming behavior: initial perivascular concentration, a guiding role for new vessel formation, and progressive incorporation into the vessels over 10 months.

Conclusions: We generated highly enriched hPSC-ECs via a clinically compatible system. Furthermore, this study demonstrated that a biocompatible PA-RGDS nanomatrix gel substantially improved long-term survival of hPSC-ECs in an ischemic environment and improved neovascularization effects of hPSC-ECs via prolonged and unique angiogenic and vessel-forming properties. This PA-RGDS-mediated transplantation of hPSC-ECs can serve as a novel platform for cell-based therapy and investigation of long-term behavior of hPSC-ECs.

Keywords: endothelial cells; pluripotent stem cells; regeneration; stem cells; vascular diseases.

MeSH terms

  • Animals
  • Cell Differentiation / physiology
  • Cell- and Tissue-Based Therapy / methods
  • Cells, Cultured
  • Endothelial Cells / physiology
  • Endothelial Cells / transplantation
  • Hindlimb / blood supply
  • Human Umbilical Vein Endothelial Cells / physiology
  • Human Umbilical Vein Endothelial Cells / transplantation*
  • Humans
  • Ischemia / physiopathology
  • Ischemia / therapy*
  • Male
  • Matrix Metalloproteinase 2 / administration & dosage*
  • Mice
  • Mice, Nude
  • Nanostructures / administration & dosage*
  • Oligopeptides / administration & dosage*
  • Pluripotent Stem Cells / physiology
  • Pluripotent Stem Cells / transplantation*
  • Random Allocation
  • Treatment Outcome

Substances

  • Oligopeptides
  • arginyl-glycyl-aspartyl-serine
  • MMP2 protein, human
  • Matrix Metalloproteinase 2