AdipoR1/APPL1 potentiates the protective effects of globular adiponectin on angiotensin II-induced cardiac hypertrophy and fibrosis in neonatal rat atrial myocytes and fibroblasts

PLoS One. 2014 Aug 6;9(8):e103793. doi: 10.1371/journal.pone.0103793. eCollection 2014.

Abstract

Atrial hypertrophy and fibrosis are essential pathological features of atrial fibrillation. Recently, adiponectin has become a protein of interest due to its beneficial effects on cardiovascular diseases. However, the molecular mechanism of atrial structural remodeling and signaling pathways evoked by adiponectin remain unclear. In the present study, we investigated the cardioprotective effect of globular adiponectin (gAcrp) on angiotensin II-induced atrial hypertrophy and fibrosis in neonatal Sprague-Dawley rat. To further investigate the molecular mechanisms underlying the preventive effect of gAcrp, transfection of cells with siRNA was used to suppress the mRNA expression of adiponectin receptor 1 (AdipoR1) and its downstream adaptor protein APPL1. Non-silencing-Cy-3 labelled siRNA was used to determine transfection efficiency using fluorescence microscopy. The expression of atrial natriuretic peptide and procollagen type1 α-1, hypertrophy marker and fibrosis one, respectively, was detected by real-time PCR. Furthermore, the expression of adenosine monophosphate-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K) and Akt was detected by western blotting. In addition, nuclear p65 translocation activity was analyzed by EMSA supershift assay. Our results showed that AdipoR1 and the adaptor protein APPL1 mediated the protective effects of gAcrp. In addition, the function of adiponectin and phosphorylation of AMPK were prominently diminished by inhibition of PI3K. Furthermore, nuclear factor-κB (NF-κB) transcription was diminished by the specific inhibition of AMPK. Taken together, AMPK pivotally interacts with NF-κB and PI3K, mediating the cardioprotective effect of adiponectin, and may serve as a therapeutic target for preventing atrial hypertrophy and fibrosis. Our present study suggests that gAcrp could ameliorate AngII-induced cardiac hypertrophy and fibrosis in rat atrial cells, which is mediated by the activation of AMPK signaling pathways. APPL1 and AdipoR1 are the key factors involved in the downstream of gAcrp approach.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • AMP-Activated Protein Kinases / metabolism
  • Adaptor Proteins, Signal Transducing / metabolism*
  • Adiponectin / metabolism*
  • Angiotensin II / adverse effects*
  • Angiotensin II / pharmacology
  • Animals
  • Cardiomegaly / chemically induced
  • Cardiomegaly / metabolism*
  • Cardiomegaly / pathology
  • Cardiomegaly / prevention & control
  • Endomyocardial Fibrosis / chemically induced
  • Endomyocardial Fibrosis / metabolism*
  • Endomyocardial Fibrosis / pathology
  • Endomyocardial Fibrosis / prevention & control
  • Fibroblasts / metabolism*
  • Fibroblasts / pathology
  • Heart Atria / metabolism
  • Heart Atria / pathology
  • Myocytes, Cardiac / metabolism*
  • Myocytes, Cardiac / pathology
  • NF-kappa B / metabolism
  • Nerve Tissue Proteins / metabolism*
  • Phosphatidylinositol 3-Kinases / metabolism
  • Rats
  • Rats, Sprague-Dawley
  • Receptors, Adiponectin / metabolism*
  • Signal Transduction / drug effects
  • Vasoconstrictor Agents / adverse effects*
  • Vasoconstrictor Agents / pharmacology

Substances

  • Adaptor Proteins, Signal Transducing
  • Adiponectin
  • Appl1 protein, rat
  • NF-kappa B
  • Nerve Tissue Proteins
  • Receptors, Adiponectin
  • Vasoconstrictor Agents
  • adiponectin receptor 1, rat
  • Angiotensin II
  • Phosphatidylinositol 3-Kinases
  • AMP-Activated Protein Kinases

Grants and funding

This work was supported by grants from National Natural Science Foundation of China (Young Project, No. 81000083). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.