Embryonic stem cell therapy of heart failure in genetic cardiomyopathy

Stem Cells. 2008 Oct;26(10):2644-53. doi: 10.1634/stemcells.2008-0187. Epub 2008 Jul 31.

Abstract

Pathogenic causes underlying nonischemic cardiomyopathies are increasingly being resolved, yet repair therapies for these commonly heritable forms of heart failure are lacking. A case in point is human dilated cardiomyopathy 10 (CMD10; Online Mendelian Inheritance in Man #608569), a progressive organ dysfunction syndrome refractory to conventional therapies and linked to mutations in cardiac ATP-sensitive K(+) (K(ATP)) channel subunits. Embryonic stem cell therapy demonstrates benefit in ischemic heart disease, but the reparative capacity of this allogeneic regenerative cell source has not been tested in inherited cardiomyopathy. Here, in a Kir6.2-knockout model lacking functional K(ATP) channels, we recapitulated under the imposed stress of pressure overload the gene-environment substrate of CMD10. Salient features of the human malignant heart failure phenotype were reproduced, including compromised contractility, ventricular dilatation, and poor survival. Embryonic stem cells were delivered through the epicardial route into the left ventricular wall of cardiomyopathic stressed Kir6.2-null mutants. At 1 month of therapy, transplantation of 200,000 cells per heart achieved teratoma-free reversal of systolic dysfunction and electrical synchronization and halted maladaptive remodeling, thereby preventing end-stage organ failure. Tracked using the lacZ reporter transgene, stem cells engrafted into host heart. Beyond formation of cardiac tissue positive for Kir6.2, transplantation induced cell cycle activation and halved fibrotic zones, normalizing sarcomeric and gap junction organization within remuscularized hearts. Improved systemic function induced by stem cell therapy translated into increased stamina, absence of anasarca, and benefit to overall survivorship. Embryonic stem cells thus achieve functional repair in nonischemic genetic cardiomyopathy, expanding indications to the therapy of heritable heart failure. Disclosure of potential conflicts of interest is found at the end of this article.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cardiomegaly / physiopathology
  • Cardiomegaly / therapy
  • Cardiomyopathies / complications*
  • Cardiomyopathies / genetics*
  • Cardiomyopathies / physiopathology
  • Cell- and Tissue-Based Therapy*
  • Embryonic Stem Cells / cytology*
  • Embryonic Stem Cells / metabolism
  • Genetic Predisposition to Disease
  • Heart Conduction System / physiopathology
  • Heart Failure / complications*
  • Heart Failure / physiopathology
  • Heart Failure / therapy*
  • Heart Function Tests
  • Humans
  • KATP Channels / deficiency
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Myocardial Contraction / physiology
  • Myocardium / pathology
  • Myocardium / ultrastructure
  • Stem Cell Transplantation
  • Stress, Physiological
  • Treatment Outcome
  • Ventricular Remodeling / physiology
  • Wound Healing

Substances

  • KATP Channels

Associated data

  • OMIM/#608569