AAV-mediated knock-down of HRC exacerbates transverse aorta constriction-induced heart failure

PLoS One. 2012;7(8):e43282. doi: 10.1371/journal.pone.0043282. Epub 2012 Aug 28.

Abstract

Background: Histidine-rich calcium binding protein (HRC) is located in the lumen of sarcoplasmic reticulum (SR) that binds to both triadin (TRN) and SERCA affecting Ca(2+) cycling in the SR. Chronic overexpression of HRC that may disrupt intracellular Ca(2+) homeostasis is implicated in pathogenesis of cardiac hypertrophy. Ablation of HRC showed relatively normal phenotypes under basal condition, but exhibited a significantly increased susceptibility to isoproterenol-induced cardiac hypertrophy. In the present study, we characterized the functions of HRC related to Ca(2+) cycling and pathogenesis of cardiac hypertrophy using the in vitro siRNA- and the in vivo adeno-associated virus (AAV)-mediated HRC knock-down (KD) systems, respectively.

Methodology/principal findings: AAV-mediated HRC-KD system was used with or without C57BL/6 mouse model of transverse aortic constriction-induced failing heart (TAC-FH) to examine whether HRC-KD could enhance cardiac function in failing heart (FH). Initially we expected that HRC-KD could elicit cardiac functional recovery in failing heart (FH), since predesigned siRNA-mediated HRC-KD enhanced Ca(2+) cycling and increased activities of RyR2 and SERCA2 without change in SR Ca(2+) load in neonatal rat ventricular cells (NRVCs) and HL-1 cells. However, AAV9-mediated HRC-KD in TAC-FH was associated with decreased fractional shortening and increased cardiac fibrosis compared with control. We found that phospho-RyR2, phospho-CaMKII, phospho-p38 MAPK, and phospho-PLB were significantly upregulated by HRC-KD in TAC-FH. A significantly increased level of cleaved caspase-3, a cardiac cell death marker was also found, consistent with the result of TUNEL assay.

Conclusions/significance: Increased Ca(2+) leak and cytosolic Ca(2+) concentration due to a partial KD of HRC could enhance activity of CaMKII and phosphorylation of p38 MAPK, causing the mitochondrial death pathway observed in TAC-FH. Our results present evidence that down-regulation of HRC could deteriorate cardiac function in TAC-FH through perturbed SR-mediated Ca(2+) cycling.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Aorta / pathology*
  • Calcium / metabolism*
  • Calcium-Binding Proteins / genetics
  • Calcium-Binding Proteins / metabolism*
  • Cardiomegaly / pathology
  • Carrier Proteins / metabolism*
  • Constriction
  • Cytosol / metabolism
  • Dependovirus / metabolism*
  • Disease Models, Animal
  • Echocardiography / methods
  • Gene Expression Regulation*
  • Heart / physiology
  • Heart Failure / genetics*
  • Heart Failure / physiopathology
  • Histidine / chemistry*
  • Homeostasis
  • Isoproterenol / pharmacology
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Muscle Proteins / metabolism*
  • Phenotype
  • Phosphorylation
  • RNA, Small Interfering / metabolism
  • Sarcoplasmic Reticulum / metabolism*
  • Sarcoplasmic Reticulum Calcium-Transporting ATPases / metabolism*

Substances

  • Calcium-Binding Proteins
  • Carrier Proteins
  • Muscle Proteins
  • RNA, Small Interfering
  • triadin
  • Histidine
  • Sarcoplasmic Reticulum Calcium-Transporting ATPases
  • Isoproterenol
  • Calcium

Grants and funding

This project was supported by: National Research Foundation (NRF) for the Ministry of Education, Science and Technology (MEST) grant (2011-0002144). Gwangju Institute of Science and Technology.(GIST) Systems Biology Infrastructure Establishment Grant (2011). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.