Abstract

The cardiomyocyte cell cycle is a poorly understood process. Mammalian cardiomyocytes permanently withdraw from the cell cycle shortly after birth but can re-enter the cell cycle and proliferate when subjected to injury within a brief temporal window in the neonatal period. Thus, investigating the mechanisms of cell cycle regulation in neonatal cardiomyocytes may provide critical insight into the molecular events that prevent adult myocytes from proliferating in response to injury or stress. MEF2D is a key transcriptional mediator of pathological remodeling in the adult heart downstream of various stress-promoting insults. However, the specific gene programs regulated by MEF2D in cardiomyocytes are unknown. By performing genome-wide transcriptome analysis using MEF2D-depleted neonatal cardiomyocytes, we found a significant impairment in the cell cycle, characterized by the up-regulation of numerous positive cell cycle regulators. Expression of Pten, the primary negative regulator of PI3K/Akt, was significantly reduced in MEF2D-deficient cardiomyocytes and found to be a direct target gene of MEF2D. Consistent with these findings mutant cardiomyocytes showed activation of the PI3K/Akt survival pathway. Paradoxically, prolonged deficiency of MEF2D in neonatal cardiomyocytes did not trigger proliferation but instead resulted in programmed cell death, which is likely mediated by the E2F transcription factor. These results demonstrate a critical role for MEF2D in cell cycle regulation of post-mitotic, neonatal cardiomyocytes in vitro.

Highlights

  • Myocyte enhancer factor 2 (MEF2) proteins are key regulators of cardiac muscle differentiation and hypertrophy, but additional roles in this cell type have not been defined

  • MEF2D Regulates a Cell Cycle Gene Program in Neonatal Cardiomyocytes—In order to gain a better understanding of the gene programs regulated by MEF2D in cardiac muscle homeostasis, we used RNA interference to deplete this factor in NRVMs

  • Knockdown of MEF2D in neonatal cardiomyocytes resulted in the dysregulation of 279 genes by Ϯ1.5-fold or more

Read more

Summary

Background

Myocyte enhancer factor 2 (MEF2) proteins are key regulators of cardiac muscle differentiation and hypertrophy, but additional roles in this cell type have not been defined. Prolonged deficiency of MEF2D in neonatal cardiomyocytes did not trigger proliferation but instead resulted in programmed cell death, which is likely mediated by the E2F transcription factor. These results demonstrate a critical role for MEF2D in cell cycle regulation of post-mitotic, neonatal cardiomyocytes in vitro. Deficiency of MEF2D in neonatal cardiomyocytes stimulated re-entry into the cell cycle, which was characterized by the up-regulation of numerous positive cell cycle regulators and activation of the PI3K/Akt pathway Despite activation of these cell cycle-promoting pathways, prolonged depletion of MEF2D did not induce proliferation but instead resulted in widespread programmed cell death. MEF2D Regulates the Cell Cycle in Cardiomyocytes in Vitro results demonstrate an unanticipated critical function for MEF2D in regulating the cell cycle and survival of post-mitotic, neonatal cardiomyocytes

Experimental Procedures
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call