Induction of the matricellular protein CCN1 through RhoA and MRTF-A contributes to ischemic cardioprotection

J Mol Cell Cardiol. 2014 Oct:75:152-61. doi: 10.1016/j.yjmcc.2014.07.017. Epub 2014 Aug 8.

Abstract

Activation of RhoA, a low molecular-weight G-protein, plays an important role in protecting the heart against ischemic stress. Studies using non-cardiac cells demonstrate that the expression and subsequent secretion of the matricellular protein CCN1 is induced by GPCR agonists that activate RhoA. In this study we determined whether and how CCN1 is induced by GPCR agonists in cardiomyocytes and examined the role of CCN1 in ischemic cardioprotection in cardiomyocytes and the isolated perfused heart. In neonatal rat ventricular myocytes (NRVMs), sphingosine 1-phosphate (S1P), lysophosphatidic acid (LPA) and endothelin-1 induced robust increases in CCN1 expression while phenylephrine, isoproterenol and carbachol had little or no effect. The ability of agonists to activate the small G-protein RhoA correlated with their ability to induce CCN1. CCN1 induction by S1P was blocked when RhoA function was inhibited with C3 exoenzyme or a pharmacological RhoA inhibitor. Conversely overexpression of RhoA was sufficient to induce CCN1 expression. To delineate the signals downstream of RhoA we tested the role of MRTF-A (MKL1), a co-activator of SRF, in S1P-mediated CCN1 expression. S1P increased the nuclear accumulation of MRTF-A and this was inhibited by the functional inactivation of RhoA. In addition, pharmacological inhibitors of MRTF-A or knockdown of MRTF-A significantly diminished S1P-mediated CCN1 expression, indicating a requirement for RhoA/MRTF-A signaling. We also present data indicating that CCN1 is secreted following agonist treatment and RhoA activation, and binds to cells where it can serve an autocrine function. To determine the functional significance of CCN1 expression and signaling, simulated ischemia/reperfusion (sI/R)-induced apoptosis was assessed in NRVMs. The ability of S1P to protect against sI/R was significantly reduced by the inhibition of RhoA, ROCK or MRTF-A or by CCN1 knockdown. We also demonstrate that ischemia/reperfusion induces CCN1 expression in the isolated perfused heart and that this functions as a cardioprotective mechanism, evidenced by the significant increase in infarct development in response to I/R in the cardiac specific CCN1 KO relative to control mice. Our findings implicate CCN1 as a mediator of cardioprotection induced by GPCR agonists that activate RhoA/MRTF-A signaling.

Keywords: CCN1; Cardioprotection; GPCRs; MRTF-A; RhoA.

Publication types

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

MeSH terms

  • Animals
  • Animals, Newborn
  • Cardiotonic Agents / metabolism*
  • Cell Membrane / drug effects
  • Cell Membrane / metabolism
  • Cell Nucleus / drug effects
  • Cell Nucleus / metabolism
  • Cysteine-Rich Protein 61 / metabolism*
  • Heart Ventricles / cytology
  • In Vitro Techniques
  • Lysophospholipids / pharmacology
  • Mice, Knockout
  • Models, Biological
  • Myocardial Ischemia / metabolism*
  • Myocardial Ischemia / pathology
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / metabolism
  • Protein Binding / drug effects
  • RNA, Small Interfering / metabolism
  • Rats
  • Rats, Sprague-Dawley
  • Receptors, G-Protein-Coupled / agonists
  • Receptors, G-Protein-Coupled / metabolism
  • Reperfusion Injury / metabolism
  • Reperfusion Injury / pathology
  • Sphingosine / analogs & derivatives
  • Sphingosine / pharmacology
  • Transcription Factors / metabolism*
  • rhoA GTP-Binding Protein / metabolism*

Substances

  • Cardiotonic Agents
  • Cysteine-Rich Protein 61
  • Lysophospholipids
  • RNA, Small Interfering
  • Receptors, G-Protein-Coupled
  • Transcription Factors
  • myocardin-related transcription factor-A, rat
  • sphingosine 1-phosphate
  • rhoA GTP-Binding Protein
  • Sphingosine