S-phase Synchronization Facilitates the Early Progression of Induced-Cardiomyocyte Reprogramming through Enhanced Cell-Cycle Exit

Int J Mol Sci. 2018 May 4;19(5):1364. doi: 10.3390/ijms19051364.

Abstract

Direct reprogramming of fibroblasts into induced cardiomyocytes (iCMs) holds a great promise for regenerative medicine and has been studied in several major directions. However, cell-cycle regulation, a fundamental biological process, has not been investigated during iCM-reprogramming. Here, our time-lapse imaging on iCMs, reprogrammed by Gata4, Mef2c, and Tbx5 (GMT) monocistronic retroviruses, revealed that iCM-reprogramming was majorly initiated at late-G1- or S-phase and nearly half of GMT-reprogrammed iCMs divided soon after reprogramming. iCMs exited cell cycle along the process of reprogramming with decreased percentage of 5-ethynyl-20-deoxyuridine (EdU)⁺/α-myosin heavy chain (αMHC)-GFP⁺ cells. S-phase synchronization post-GMT-infection could enhance cell-cycle exit of reprogrammed iCMs and yield more GFPhigh iCMs, which achieved an advanced reprogramming with more expression of cardiac genes than GFPlow cells. However, S-phase synchronization did not enhance the reprogramming with a polycistronic-viral vector, in which cell-cycle exit had been accelerated. In conclusion, post-infection synchronization of S-phase facilitated the early progression of GMT-reprogramming through a mechanism of enhanced cell-cycle exit.

Keywords: cell division; cell-cycle exit; cell-cycle synchronization; epigenetic reprogramming; induced cardiomyocyte.

MeSH terms

  • Animals
  • Cell Cycle / genetics
  • Cell Cycle Checkpoints / genetics*
  • Cell Differentiation / genetics*
  • Cellular Reprogramming / genetics*
  • Fibroblasts / cytology
  • Fibroblasts / metabolism
  • Mice
  • Myocytes, Cardiac / cytology*
  • Myocytes, Cardiac / metabolism
  • Regenerative Medicine / trends