Bioluminescent imaging of genetically selected induced pluripotent stem cell-derived cardiomyocytes after transplantation into infarcted heart of syngeneic recipients

PLoS One. 2014 Sep 16;9(9):e107363. doi: 10.1371/journal.pone.0107363. eCollection 2014.

Abstract

Cell loss after transplantation is a major limitation for cell replacement approaches in regenerative medicine. To assess the survival kinetics of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) we generated transgenic murine iPSC lines which, in addition to CM-specific expression of puromycin N-acetyl-transferase and enhanced green fluorescent protein (EGFP), also constitutively express firefly luciferase (FLuc) for bioluminescence (BL) in vivo imaging. While undifferentiated iPSC lines generated by random integration of the transgene into the genome retained stable FLuc activity over many passages, the BL signal intensity was strongly decreased in purified iPS-CM compared to undifferentiated iPSC. Targeted integration of FLuc-expression cassette into the ROSA26 genomic locus using zinc finger nuclease (ZFN) technology strongly reduced transgene silencing in iPS-CM, leading to a several-fold higher BL compared to iPS-CM expressing FLuc from random genomic loci. To investigate the survival kinetics of iPS-CM in vivo, purified CM obtained from iPSC lines expressing FLuc from a random or the ROSA26 locus were transplanted into cryoinfarcted hearts of syngeneic mice. Engraftment of viable cells was monitored by BL imaging over 4 weeks. Transplanted iPS-CM were poorly retained in the myocardium independently of the cell line used. However, up to 8% of cells survived for 28 days at the site of injection, which was confirmed by immunohistological detection of EGFP-positive iPS-CM in the host tissue. Transplantation of iPS-CM did not affect the scar formation or capillary density in the periinfarct region of host myocardium. This report is the first to determine the survival kinetics of drug-selected iPS-CM in the infarcted heart using BL imaging and demonstrates that transgene silencing in the course of iPSC differentiation can be greatly reduced by employing genome editing technology. FLuc-expressing iPS-CM generated in this study will enable further studies to reduce their loss, increase long-term survival and functional integration upon transplantation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Differentiation
  • Cell Line
  • Gene Expression
  • Gene Silencing
  • Genes, Reporter
  • Genetic Loci
  • Induced Pluripotent Stem Cells / cytology*
  • Induced Pluripotent Stem Cells / metabolism
  • Luminescent Measurements / methods*
  • Male
  • Mice
  • Molecular Imaging*
  • Myocardial Infarction / diagnosis*
  • Myocardial Infarction / mortality
  • Myocardial Infarction / pathology
  • Myocardial Infarction / therapy*
  • Myocytes, Cardiac / cytology*
  • Myocytes, Cardiac / metabolism
  • Myocytes, Cardiac / transplantation*
  • Promoter Regions, Genetic
  • RNA, Untranslated / genetics
  • Transduction, Genetic
  • Transgenes

Substances

  • Gt(ROSA)26Sor non-coding RNA, mouse
  • RNA, Untranslated

Grants and funding

This study was supported by the grants from the Federal Ministry for Education and Research (BMBF, http://www.bmbf.de/) to T.Š. (grant No. 01GN0947) and from the Else-Kröner-Fresenius Stiftung (http://www.ekfs.de/de/start.html) to T.Š. and K.N. (grant No. A93/2008). Further funding was provided by the Maria Pesch Stiftung and Köln-Fortune Program (http://www.medfak.uni-koeln.de/index.php?id=195) to T.Š. and by the Volkswagen Foundation (http://www.volkswagenstiftung.de/) to M.H. (I/83 443). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.