Original article
ROCK1 plays an essential role in the transition from cardiac hypertrophy to failure in mice

https://doi.org/10.1016/j.yjmcc.2010.08.008Get rights and content

Abstract

Pathological cardiac hypertrophy caused by diverse etiologies eventually leads to cardiac dilation and functional decompensation. We have recently reported that genetic deletion of Rho-associated coiled-coil containing protein kinase 1 (ROCK1) inhibited several pathological events including cardiomyocyte apoptosis in compensated hypertrophic hearts. The present study investigated whether ROCK1 deficiency can prevent the transition from hypertrophy to heart failure. Transgenic mice with cardiac-restricted overexpression of Gαq develop compensated cardiac hypertrophy at young ages, but progress into lethal cardiomyopathy accompanied by increased apoptosis after pregnancy or at old ages. The studies were first carried out using age- and pregnancy-matched wild-type, Gαq, ROCK1−/−, and Gαq/ROCK1−/− mice. The potent beneficial effect of ROCK1 deletion is demonstrated by abolishment of peripartum mortality, and significant attenuation of left ventricular (LV) dilation, wall thinning, and contractile dysfunction in the peripartum Gαq transgenic mice. Increase in cardiomyocyte apoptosis was suppressed by ROCK1 deletion, associated with increased extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) activation and inhibition of mitochondrial translocation of Bax. In addition, ROCK1 deficiency also improved survival, inhibited cardiomyocyte apoptosis, and preserved LV dimension and function in old Gαq mice at 12 months. Furthermore, transgenic overexpression of ROCK1 increased cardiomyocyte apoptosis and accelerated hypertrophic decompensation in Gαq hearts in the absence of pregnancy stress. The present study provides for the first time in vivo evidence for the long-term beneficial effects of ROCK1 deficiency in hypertrophic decompensation and suggests that ROCK1 may be an attractive therapeutic target to limit heart failure progression.

Research highlights

► ROCK1 deletion abolished peripartum mortality, prevented the development of heart failure, and inhibited cardiomycoyte apoptosis while preserving cardiomyocyte hypertrophy in the peripartum Gαq mice. ► ROCK1 deficiency improved survival, inhibited cardiomyocyte apoptosis, and preserved LV dimension and function in old Gαq mice at 12 months. ► Transgenic overexpression of ROCK1 increased cardiomyocyte apoptosis and accelerated hypertrophic decompensation in Gαq hearts in the absence of pregnancy stress. ► The present study demonstrated the long-term beneficial effects of ROCK1 deficiency in hypertrophic decompensation.

Introduction

Heart failure is a leading cause for human morbidity and mortality, and the incidence of heart failure has been constantly increasing during the past decades. Cardiac hypertrophy is initially a compensatory response to diverse etiologies, but it eventually leads to heart failure or sudden death due to decompensation [1], [2], [3]. Identifying the signaling mechanisms underlying the development of cardiac hypertrophy and the transition to heart failure will be helpful for the design of effective therapeutics. Rho-associated coiled-coil containing protein kinase (ROCK) is a downstream mediator of RhoA, and plays a critical role in mediating the effects of RhoA on stress fiber formation, smooth muscle contraction, cell adhesion, membrane ruffling, cell motility and apoptosis [4], [5], [6]. Studies using pharmacological inhibitors, Y27632 and fasudil, suggest an in vivo role for ROCK in the pathogenesis of cardiac hypertrophy and remodeling [7], [8], [9]. However, these inhibitors do not distinguish between ROCK1 and ROCK2, the two isoforms of ROCK family, and could also have non-selective effects [10]. Recent genetic studies by our laboratory and others support the concept that ROCK1 and ROCK2 have distinct non-redundant functions in cardiac hypertrophy and remodeling [11], [12], [13], [14].

We showed that ROCK1 deletion did not impair compensatory hypertrophic response, but significantly reduced cardiomyocyte apoptosis and fibrosis in response to pressure overload induced by transverse aortic constriction [11], [12]. In addition, ROCK1 deletion did not affect the development of cardiac hypertrophy in Gαq transgenic mice, but prevented chamber dilation and contractile dysfunction at young ages (12 weeks) [13]. The Gq class of heterotrimeric G proteins is an important transducer of humoral (i.e., α1-adrenergic agonists, angiotensin II, endothelin and prostaglandin F2α) and mechanical stimuli that are important in cardiac hypertrophy. Transgenic expression of Gαq in the myocardium elicits cardiac hypertrophy and contractile dysfunction, but without significant increase in cardiomyocyte apoptosis at young ages [15], [16]. These results indicate that ROCK1 does not play a significant role in compensatory hypertrophic responses, and raise the possibility that ROCK1 plays a critical role in the maladaptive response which contributes to the transition from compensatory cardiac hypertrophy to heart failure.

To explore this concept and determine long-term impact of ROCK1 deficiency in the setting of cardiomyopathy, the present study examined the effects of ROCK1 deletion on decompensation of the hypertrophic Gαq hearts under two different stress conditions: multiple pregnancy and at 12-month-old age. Previous reports have validated this decompensation model as hypertrophic Gαq hearts progress into heart failure after additional stresses such as pregnancy, aging or pressure overload [15], [17], [18], [19]. Our results show that ROCK1 deletion strikingly improved animal survival and prevented the development of heart failure under both conditions by preserving chamber dimension and contractile function, suppressing increase in cardiomyocyte apoptosis and cardiac fibrosis. In addition, transgenic overexpression of ROCK1 alone did not cause significant changes in heart structure/function, but increased cardiomyocyte apoptosis and accelerated hypertrophic decompensation in Gαq hearts in the absence of pregnancy stress. These results provide the first in vivo evidence for an essential role for ROCK1 in cardiac decompensation.

Section snippets

Methods

All animal experiments were conducted in accordance with the National Institutes Health “Guide for the Care and Use of Laboratory Animals” (NIH Publication No. 85-23, revised 1996) and were approved by the Institutional Animal Care and Use Committee at Indiana University School of Medicine.

ROCK1 deletion abolished peripartum mortality in the peripartum Gαq mice

Previous study has reported about 30% to 50% of female Gαq mice develop lethal failure in the peripartum period [15], [18], [19]. To determine whether ROCK1 deletion decreases the mortality of the peripartum Gαq mice, 8-week-old female wild-type (WT), ROCK1−/−, Gαq and Gαq/ROCK1−/− mice were mated with WT males. Gαq mice exhibited 30% lethality after first pregnancy and 100% lethality after the fourth pregnancy (Fig. 1(A)) as predicted. Strikingly, Gαq-induced peripartum lethality was

Discussion

The present study examined the effects of ROCK1 deficiency on hypertrophy decompensation in the context of Gαq-induced cardiomyopathy under two conditions associated with lethal dilated cardiomyopathy such as pregnancy (single or multiple pregnancies) and aging (12 months). Our results provide multiple new insights into the role of ROCK1 in the development of heart failure. First, ROCK1 deficiency abolished Gαq-induced animal death after single or multiple pregnancies as well as at 12 months of

Acknowledgments

This work was supported by National Institutes of Health grants [HL072897 and HL085098 to L.W.] and by the Riley Children's Foundation and the Lilly Endowment. The authors thank Jonathan Lee and Mica Gosnell for technical assistance. We thank Dr. Loren J. Field and Dr. Michael Rubart for many insightful comments on this study. We are grateful to Dr. Gerald W. Dorn at Washington University for the αMHC-Gαq line.

References (30)

  • J. Heineke et al.

    Regulation of cardiac hypertrophy by intracellular signalling pathways

    Nat Rev

    (2006)
  • J.H. Brown et al.

    The Rac and Rho hall of fame: a decade of hypertrophic signaling hits

    Circ Res

    (2006)
  • J.A. Hill et al.

    Cardiac plasticity

    N Engl J Med

    (2008)
  • J. Shi et al.

    Rho kinase in the regulation of cell death and survival

    Arch Immunol Ther Exp (Warsz)

    (2007)
  • G. Loirand et al.

    Rho kinases in cardiovascular physiology and pathophysiology

    Circ Res

    (2006)
  • K. Noma et al.

    Physiological role of ROCKs in the cardiovascular system

    Am J Physiol Cell Physiol

    (2006)
  • S. Satoh et al.

    Chronic inhibition of Rho kinase blunts the process of left ventricular hypertrophy leading to cardiac contractile dysfunction in hypertension-induced heart failure

    J Mol Cell Cardiol

    (2003)
  • M. Higashi et al.

    Long-term inhibition of Rho-kinase suppresses angiotensin II-induced cardiovascular hypertrophy in rats in vivo: effect on endothelial NAD(P)H oxidase system

    Circ Res

    (2003)
  • T. Hattori et al.

    Long-term inhibition of Rho-kinase suppresses left ventricular remodeling after myocardial infarction in mice

    Circulation

    (2004)
  • M. Uehata et al.

    Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension

    Nature

    (1997)
  • J. Chang et al.

    Activation of Rho-associated coiled-coil protein kinase 1 (ROCK-1) by caspase-3 cleavage plays an essential role in cardiac myocyte apoptosis

    Proc Natl Acad Sci USA

    (2006)
  • Y.M. Zhang et al.

    Targeted deletion of ROCK1 protects the heart against pressure overload by inhibiting reactive fibrosis

    FASEB J

    (2006)
  • J. Shi et al.

    Disruption of ROCK1 gene attenuates cardiac dilation and improves contractile function in pathological cardiac hypertrophy

    J Mol Cell Cardiol

    (2008)
  • Y. Rikitake et al.

    Decreased perivascular fibrosis but not cardiac hypertrophy in ROCK1+/− haploinsufficient mice

    Circulation

    (2005)
  • J.W. Adams et al.

    Enhanced Galphaq signaling: a common pathway mediates cardiac hypertrophy and apoptotic heart failure

    Proc Natl Acad Sci USA

    (1998)
  • Cited by (56)

    • Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting

      2020, Advanced Drug Delivery Reviews
      Citation Excerpt :

      For example, rosuvastatin activates pro-survival kinases of the reperfusion injury salvage pathway (RISK), including AKT, ERK and PKC, in hypercholesterolemic pigs subjected to myocardial infarction, thereby limiting infarct size and improving cardiac function [341]. In addition to blocking AKT signaling [307], ROCK activity promotes apoptotic signaling by caspase-3 activation [342], inhibition of pro-survival ERK/MAPK signaling and increased expression of pro-apoptotic Bax [343,344]. Lastly, statin-mediated inhibition of the Rho/ROCK pathway improves cardiac contractile function by increasing expression of sarcoplasmic reticulum Ca2+-ATPase (SERCA) [345,346].

    • Inhibition of Rho-associated kinases suppresses cardiac myofibroblast function in engineered connective and heart muscle tissues

      2019, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      Both have been suggested to play a role in cardiac disease and in remodelling of the myocardium [6]. Studies using ROCK1 and ROCK2-deficient mice indicate that ROCKs regulate cardiomyocyte apoptosis and cardiac fibrosis [7–12]. In addition, ROCK2 was shown to contribute to cardiac hypertrophy [8,11].

    • Rho-kinase inhibition reverses impaired Ca<sup>2+</sup> handling and associated left ventricular dysfunction in pressure overload-induced cardiac hypertrophy

      2017, Cell Calcium
      Citation Excerpt :

      The increase in lung weight implies the development of pulmonary edema at the end of the 10-week period and indicates that the left ventricle of the heart cannot perform its function adequately. Regressed hypertrophy and decreased pulmonary edema in rats in the TAC + Fas group indicate that ROCK activity plays an important role in the onset of pathological cardiac hypertrophy [17,20,21]. Pathological cardiac hypertrophy is associated with significant functional changes, including depressed contractile activity and delayed kinetics at the tissue or myocyte level [4,17].

    View all citing articles on Scopus
    View full text