A cardiac-enriched microRNA, miR-378, blocks cardiac hypertrophy by targeting Ras signaling

J Biol Chem. 2013 Apr 19;288(16):11216-32. doi: 10.1074/jbc.M112.442384. Epub 2013 Feb 27.

Abstract

Understanding the regulation of cardiomyocyte growth is crucial for the management of adverse ventricular remodeling and heart failure. MicroRNA-378 (miR-378) is a newly described member of the cardiac-enriched miRNAs, which is expressed only in cardiac myocytes and not in cardiac fibroblasts. We have previously shown that miR-378 regulates cardiac growth during the postnatal period by direct targeting of IGF1R (Knezevic, I., Patel, A., Sundaresan, N. R., Gupta, M. P., Solaro, R. J., Nagalingam, R. S., and Gupta, M. (2012) J. Biol. Chem. 287, 12913-12926). Here, we report that miR-378 is an endogenous negative regulator of cardiac hypertrophy, and its levels are down-regulated during hypertrophic growth of the heart and during heart failure. In primary cultures of cardiomyocytes, overexpression of miR-378 blocked phenylephrine (PE)-stimulated Ras activity and also prevented activation of two major growth-promoting signaling pathways, PI3K-AKT and Raf1-MEK1-ERK1/2, acting downstream of Ras signaling. Overexpression of miR-378 suppressed PE-induced phosphorylation of S6 ribosomal kinase, pERK1/2, pAKT, pGSK-3β, and nuclear accumulation of NFAT. There was also suppression of the fetal gene program that was induced by PE. Experiments carried out to delineate the mechanism behind the suppression of Ras, led us to identify Grb2, an upstream component of Ras signaling, as a bona fide direct target of miR-378-mediated regulation. Deficiency of miR-378 alone was sufficient to induce fetal gene expression, which was prevented by knocking down Grb2 expression and blocking Ras activation, thus suggesting that miR-378 interferes with Ras activation by targeting Grb2. Our study demonstrates that miR-378 is an endogenous negative regulator of Ras signaling and cardiac hypertrophy and its deficiency contributes to the development of cardiac hypertrophy.

Publication types

  • Research Support, N.I.H., Extramural
  • Retracted Publication

MeSH terms

  • Adrenergic alpha-1 Receptor Agonists / adverse effects
  • Adrenergic alpha-1 Receptor Agonists / pharmacology
  • Animals
  • Cardiomegaly / chemically induced
  • Cardiomegaly / genetics
  • Cardiomegaly / metabolism*
  • Cardiomegaly / pathology
  • Cells, Cultured
  • GRB2 Adaptor Protein / biosynthesis
  • GRB2 Adaptor Protein / genetics
  • Gene Expression Regulation / drug effects
  • Gene Expression Regulation / genetics
  • Glycogen Synthase Kinase 3 / genetics
  • Glycogen Synthase Kinase 3 / metabolism
  • Glycogen Synthase Kinase 3 beta
  • MAP Kinase Kinase 1 / genetics
  • MAP Kinase Kinase 1 / metabolism
  • MAP Kinase Kinase Kinases / genetics
  • MAP Kinase Kinase Kinases / metabolism
  • MAP Kinase Signaling System*
  • MicroRNAs / genetics
  • MicroRNAs / metabolism*
  • Mitogen-Activated Protein Kinase 3 / genetics
  • Mitogen-Activated Protein Kinase 3 / metabolism
  • Muscle Proteins / genetics
  • Muscle Proteins / metabolism*
  • Phenylephrine / adverse effects
  • Phenylephrine / pharmacology
  • Phosphatidylinositol 3-Kinases / genetics
  • Phosphatidylinositol 3-Kinases / metabolism
  • Proto-Oncogene Proteins c-akt
  • Proto-Oncogene Proteins c-raf
  • Rats
  • Rats, Sprague-Dawley
  • ras Proteins / genetics
  • ras Proteins / metabolism*

Substances

  • Adrenergic alpha-1 Receptor Agonists
  • GRB2 Adaptor Protein
  • Grb2 protein, rat
  • MIRN378 microRNA, rat
  • MicroRNAs
  • Muscle Proteins
  • Phenylephrine
  • Phosphatidylinositol 3-Kinases
  • Glycogen Synthase Kinase 3 beta
  • Proto-Oncogene Proteins c-akt
  • Proto-Oncogene Proteins c-raf
  • Raf1 protein, rat
  • Mitogen-Activated Protein Kinase 3
  • MAP Kinase Kinase Kinases
  • Glycogen Synthase Kinase 3
  • MAP Kinase Kinase 1
  • ras Proteins