Cell-based delivery of dATP via gap junctions enhances cardiac contractility

SD Lundy, SA Murphy, SK Dupras, J Dai… - Journal of molecular and …, 2014 - Elsevier
Journal of molecular and cellular cardiology, 2014Elsevier
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is a
promising strategy to treat myocardial infarction and reverse heart failure, but to date the
contractile benefit in most studies remains modest. We have previously shown that the
nucleotide 2-deoxyadenosine triphosphate (dATP) can substitute for ATP as the energy
substrate for cardiac myosin, and increasing cellular dATP content by globally
overexpressing ribonucleotide reductase (R1R2) can dramatically enhance cardiac …
Abstract
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is a promising strategy to treat myocardial infarction and reverse heart failure, but to date the contractile benefit in most studies remains modest. We have previously shown that the nucleotide 2-deoxyadenosine triphosphate (dATP) can substitute for ATP as the energy substrate for cardiac myosin, and increasing cellular dATP content by globally overexpressing ribonucleotide reductase (R1R2) can dramatically enhance cardiac contractility. Because dATP is a small molecule, we hypothesized that it would diffuse readily between cells via gap junctions and enhance the contractility of neighboring coupled wild type cells. To test this hypothesis, we performed studies with the goals of (1) validating gap junction-mediated dATP transfer in vitro and (2) investigating the use of R1R2-overexpressing hPSC-CMs in vivo as a novel strategy to increase cardiac function. We first performed intracellular dye transfer studies using dATP conjugated to fluorescein and demonstrated rapid gap junction-mediated transfer between cardiomyocytes. We then cocultured wild type cardiomyocytes with either cardiomyocytes or fibroblasts overexpressing R1R2 and saw more than a twofold increase in the extent and rate of contraction of wild type cardiomyocytes. Finally, we transplanted hPSC-CMs overexpressing R1R2 into healthy uninjured rat hearts and noted an increase in fractional shortening from 41 ± 4% to 53 ± 5% just five days after cell transplantation. These findings demonstrate that dATP is an inotropic factor that spreads between cells via gap junctions. Our data suggest that transplantation of dATP-producing hPSC-CMs could significantly increase the effectiveness of cardiac cell therapy.
Elsevier