The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
Contents lists available at ScienceDirect
The International Journal of Biochemistry & Cell Biology journal homepage: www.elsevier.com/locate/biocel
Review
Molecular regulation of cardiac hypertrophy Sean P. Barry a,1 , Sean M. Davidson b,1 , Paul A. Townsend c,∗ a
Medical Molecular Biology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N IEH, United Kingdom The Hatter Cardiovascular Institute, Department of Medicine, Royal Free and University College Medical School, 67 Chenies Mews, University College Hospital, London WC1E 6HX, United Kingdom c Human Genetics Division, MP808, Duthie Building, Southampton General Hospital, Tremona Road, University of Southampton, Southampton SO16 6YD, United Kingdom b
a r t i c l e
i n f o
Article history: Received 30 November 2007 Received in revised form 13 February 2008 Accepted 15 February 2008 Available online 26 February 2008 Keywords: Cardiac Hypertrophy Molecular Signalling Transcription
a b s t r a c t Heart failure is one of the leading causes of mortality in the western world and encompasses a wide spectrum of cardiac pathologies. When the heart experiences extended periods of elevated workload, it undergoes hypertrophic enlargement in response to the increased demand. Cardiovascular disease, such as that caused by myocardial infarction, obesity or drug abuse promotes cardiac myocyte hypertrophy and subsequent heart failure. A number of signalling modulators in the vasculature milieu are known to regulate heart mass including those that influence gene expression, apoptosis, cytokine release and growth factor signalling. Recent evidence using genetic and cellular models of cardiac hypertrophy suggests that pathological hypertrophy can be prevented or reversed and has promoted an enormous drive in drug discovery research aiming to identify novel and specific regulators of hypertrophy. In this review we describe the molecular characteristics of cardiac hypertrophy such as the aberrant re-expression of the fetal gene program. We discuss the various molecular pathways responsible for the co-ordinated control of the hypertrophic program including: natriuretic peptides, the adrenergic system, adhesion and cytoskeletal proteins, IL-6 cytokine family, MEK-ERK1/2 signalling, histone acetylation, calcium-mediated modulation and the exciting recent discovery of the role of microRNAs in controlling cardiac hypertrophy. Characterisation of the signalling pathways leading to cardiac hypertrophy has led to a wealth of knowledge about this condition both physiological and pathological. The challenge will be translating this knowledge into potential pharmacological therapies for the treatment of cardiac pathologies. © 2008 Elsevier Ltd. All rights reserved.
Abbreviations: ACE, angiotensin-converting enzyme; ANF, atrial natriuretic factor; AngII, angiotensin II; ANP, atrial natriuretic peptide; ASK-1, apoptosis signalling kinase-1; AT-1, angiotensin type-1; AR, adrenergic receptors; BNP, B-type natriuretic peptide; CaMKII, calcineurin and Ca2+ /calmodulin-dependent kinase II; CBP, CREB-binding protein; cGMP, guanosine 3 ,5 -cyclic monophosphate; CHF, coronary heart failure; CNP, C-type natriuretic peptide; CT-1, cardiotrophin-1; CVD, cardiovascular disease; DCM, dilated cardiomyopathy; DUSP6, dual-specific phosphatase-6; ECM, extracellular matrix; ERK, extracellular signal receptor-regulated kinase; GPCR, G-protein-coupled receptor; GTP, guanosine triphosphate; HAND, heart and neural crest derivatives; HCM, familial hypertrophic cardiomyopathy; JAK, Janus-activated kinase; LIF-1, leukaemia-inhibitory factor-1; MAPK, mitogen-activated protein kinase; MEF2, myocyte-specific enhancer factor 2; MEK, MAPK/ERK kinase; ␣-/-MHC, myosin heavy chain alpha or beta; miRNA, microRNA; NFAT, nuclear factor of activated T cells; NF-B, nuclear factor-kappa B; NP, natriuretic peptides; NPR, natriuretic peptide receptor; PDE5A, phosphodiesterase 5A; PE, phenylephrine; PGK-1, protein kinase-1; PI(3)K, phosphatidyl inositol-3 kinase; PKA, protein kinase A; PKC, protein kinase C; SERCA, sarco-/endo-plasmic reticulum Ca2+ -ATPase; SirT, SirT-related protein; SR, sarcoplasmic reticulum; STAT, signal transducer and activator of transcription; TAC, thoracic aortic constriction; THRAP1, thyroid receptor-associated protein 1; VSM, vascular smooth muscle; YFP, yellow fluorescent protein. ∗ Corresponding author. Tel.: +44 23 8079 8692; fax: +44 23 8079 4264. E-mail addresses:
[email protected] (S.P. Barry),
[email protected] (S.M. Davidson),
[email protected] (P.A. Townsend). 1 SPB and SMD contributed equally to this article (surnames listed alphabetically). 1357-2725/$ – see front matter © 2008 Elsevier Ltd. All rights reserved. doi:10.1016/j.biocel.2008.02.020
2024
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
Contents 1. 2. 3.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Inherited cardiomyopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . The fetal gene program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Myosin heavy chain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Natriuretic peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4. G-protein-coupled receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5. Interluekin-6 family of cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6. Adhesion and cytoskeletal proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7. Mitogen-activated protein kinase/extracellular signal receptor-regulated kinase signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8. Histone acetylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8.1. Histone acetyltransferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8.2. Histone deacetylases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8.3. Class III HDACs role in cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9. Calcium-mediated pathways towards hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10. MicroRNAs—new players in cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1. Introduction Cardiovascular disease (CVD) is one of the leading causes of death worldwide, accounting for 16.7 million deaths per annum (the World Health Organization— http://www.who.int/dietphysicalactivity/publications/ facts/cvd/en/). CVD encompasses a wide spectrum of cardiac pathologies including 5.3 million people living with heart failure in the USA alone (the American Heart Association—http://www.americanheart.org/presenter. jhtml?identifier=4478). In the various forms of heart failure, excessive cardiac workload leads to an enlargement of the heart in an endeavour to manage the increased hemodynamic demand. This process, known as hypertrophy, is classified as “physiological” hypertrophy when it occurs in healthy individuals following exercise and is not associated with cardiac damage, or “pathological” hypertrophy (Fig. 1). In the case of pathological hypertrophy, although the increased heart size is initially a compensatory mechanism, sustained hypertrophy can ultimately lead to a decline in left ventricular function and thus represents an independent risk factor for heart failure (Levy, Garrison, Savage, Kannel, & Castelli, 1990). The main causes of pathological hypertrophy are hypertension, genetic polymorphisms, and loss of myocytes following ischaemic damage. Altered cardiac metabolism can also be an important component leading to hypertrophy (Rajabi, Kassiotis, Razeghi, & Taegtmeyer, 2007). Indeed, this mechanism is so prevalent in the setting of diabetes that it has led to the description of a specific syndrome of “diabetic cardiomyopathy” (Boudina & Abel, 2007). Hypertrophic growth develops in two ways: concentric hypertrophy is caused by chronic pressure overload and leads to reduced left ventricular volume and increased wall thickness whereas eccentric hypertrophy is caused by volume overload and causes dilation and thinning of the heart wall (Wakatsuki, Schlessinger, & Elson, 2004). Mechanistically, eccentric expansion occurs by addition of sarcomeres in series causing cell elongation; concentric enlargement on the other hand is caused by addition of sarcomeres
2024 2024 2026 2026 2026 2028 2030 2031 2031 2032 2032 2032 2033 2034 2034 2035 2035 2035
in parallel resulting in increased cell thickness. Accompanying the increase in myocyte size, there is an increase in the number of cardiac fibroblasts causing fibrosis and increased myocardial stiffness (Wakatsuki et al., 2004). This in turn leads to overload and promotes further hypertrophy and cell death, resulting in a detrimental cycle of cardiac enlargement and myocyte loss. Although hypertrophy is ultimately a detrimental process, it is nonetheless highly organized and tailored to the specific needs of the heart. Thus, pressure and volume overload lead to concentric and eccentric hypertrophy via co-ordinated activation of specific intracellular signalling pathways, ultimately altering myocyte shape in a way that is best suited to deal with the specific burden. In this review we will discuss the various molecular pathways that are responsible for the co-ordinated control of the hypertrophic program. 2. Inherited cardiomyopathy The aetiology of cardiomyopathies can be idiopathic, immunological, toxicological or genetic but in general, a defect in normal contractile function is compensated for by hypertrophic remodelling. The two major inherited forms are hypertrophic cardiomyopathy (HCM) characterized by increased wall thickness, reduced ventricular chamber volumes and reduced diastolic function and dilated cardiomyopathy (DCM) characterized by increased chamber volume, defects in systolic function and ventricular tachyarrhythmias. A diverse range of mutations underlie familial hypertrophy, but the great majority are located in genes encoding sarcomeric proteins, and can drastically alter Ca2+ handling and contractility, leading to arrhythmias and sudden cardiac death. These have been thoroughly reviewed previously (Nicol, Frey, & Olson, 2000), but some pertinent examples are given below. The first identified mutation causing a cardiomyopathy was a missense mutation in the -myosin heavy chain (-MHC) gene associated with HCM (Geisterfer-Lowrance et al., 1990). The thick filament of the sarcomere con-
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
2025
Fig. 1. Pathological and physiological hypertrophic response to stimuli. When the heart experiences extended periods of elevated workload, it undergoes enlargement in an effort to cope with the increased demand. This process, known as hypertrophy, is classified as “physiological” when it occurs in healthy individuals following exercise or pregnancy and is not associated with cardiac damage. Whereas the increased heart size in “pathological” hypertrophy presents initially as a compensatory mechanism, sustained hypertrophy may ultimately lead to a decline in left ventricular function and as such represents an independent risk factor for heart failure. The main causes of pathological hypertrophy are hypertension, genetic polymorphisms, and loss of myocytes following ischaemic damage. Hypertrophic growth develops in two ways: concentric hypertrophy caused by chronic pressure overload leading to reduced left ventricular volume and increased wall thickness; eccentric hypertrophy due to volume overload and causing dilation and thinning of the heart wall. Eccentric expansion occurs by addition of the contractile sarcomere in series causing cell elongation whereas concentric enlargement is caused by addition of sarcomeres in parallel resulting in increased cell thickness.
sists primarily of myosin, a protein composed of two heavy chains and four light chains which hydrolyses ATP to move actin myofilaments in response to Ca2+ —thereby mediating cardiac contraction. Over 400 mutations in patients with cardiomyopathy have now been identified in 9 separate sarcomeric genes, including troponin, ␣tropomyosin, myosin-binding protein C and actin (Lowey, 2002; Richard et al., 2003) but the majority are localized in the -MHC (MYH7) gene (Richard et al., 2003). Early work on the prominent R403Q mutation in MYH7 showed that it reduced ATPase activity, resulting in an 80% decrease in maximum velocity compared to wildtype myosin (Cuda, Fananapazir, Epstein, & Sellers, 1997), although recent studies have shown the same mutation can actually enhance mechanical performance at the single molecule level (Palmiter et al., 2000). Other -MHC mutations can affect aspects other than contractility, for example the A223T mutation affects thermostability and protein folding, while the S642L mutation may alter the conformation of the actin-binding site (Karkkainen & Peuhkurinen, 2007). In all cases, the end result is compensation for altered workload though hypertrophy. The thin filament of the sarcomere is composed of a double helix of actin bound to tropomyosin and troponin. Troponin is comprised of three subunits: troponin T (TnT), which binds to tropomyosin; troponin C (TnC), the Ca2+ binding subunit and the inhibitory troponin I (TnI)
(Metzger & Westfall, 2004). Mutations have been identified in all three troponin subunits, with mutations in TnT being the most prevalent (Chang & Potter, 2005). Though the mutations responsible for DCM tend to be more diverse and are less well understood than those for HCM, in general it seems that HCM mutations cause increased Ca2+ sensitivity while DCM mutations decrease it. For example the A2V mutation in TnI reduces myocyte contraction and results in DCM (Murphy et al., 2004), while the R21C mutation increases Ca2+ sensitivity and force development and results in HCM (Gomes, Harada, & Potter, 2005). Similarly, TnT mutations from HCM patients tend to increase Ca2+ sensitivity of myofilaments, enhancing contractility, impairing relaxation and causing diastolic abnormalities, whereas many TnT mutations from DCM patients have the opposite effect, i.e. decreased Ca2+ sensitivity. Importantly, in a knock-in mouse model harbouring the TnT K210 mutation which causes DCM (Kamisago et al., 2000; Karkkainen & Peuhkurinen, 2007), increasing Ca2+ sensitivity with the inotropic agent pimobendan ameliorated the cardiac hypertrophy and heart failure, suggesting that restoring Ca2+ sensitivity may be very beneficial in certain DCM patients (Du et al., 2007). These and other examples illustrate that mutations in practically any sarcomeric proteins may eventuate in cardiomyopathy if contractility is affected, thereby initiating the hypertrophic program.
2026
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
3. The fetal gene program Following the onset of increased mechanical stress or pressure overload one of the initial molecular changes is reactivation of the so-called fetal gene program—a set of genes that are normally expressed only in the developing heart and are repressed in the adult myocardium. Activation of the fetal gene program allows co-ordinated synthesis of the proteins needed to bring about increased cardiac myocyte size and adjustment to the altered energy demands of these larger cells. Indeed, correspondence analysis between gene expression profiles in normal adult heart, failing heart and fetal heart revealed that the pattern of highly differentially expressed genes is very similar in fetal and failing hearts (Thum et al., 2007). Reinduction of the fetal gene program is associated with pathology—eccentric and concentric hypertrophy induces fetal gene expression, whereas physiological hypertrophy does not. While there are a host of re-expressed fetal genes during hypertrophy, the expression levels of myosin heavy chains, the natriuretic peptides and ␣-actinin, are used most commonly as molecular markers of hypertrophy. 3.1. Myosin heavy chain It has been appreciated for over 20 years that one of the hallmarks of cardiac hypertrophy in patients and in rodent models is an increase in -MHC expression and a decrease in ␣-MHC expression (Izumo et al., 1987; Mahdavi, Lompre, Chambers, & Nadal-Ginard, 1984). Since each isoform has a distinct enzymatic activity, it means that their relative ratios greatly influence cardiac function, thus an increase in -MHC decreases the myosin ATPase enzyme velocity which in turn slows the myocyte contraction rate, a key adaptation to altered workload (Lowes et al., 1997). Reversion of hypertrophy is associated with a return of MHC isoform levels to normal, for example, in patients with dilated cardiomyopathy undergoing -blocker therapy, recovery was associated with increased ␣-MHC and decreased -MHC expression (Lowes et al., 2002). While left ventricular MHC isoform switching has been studied extensively in humans and animal models, one complication is that whereas in adult humans 90% of the total MHC pool is of the -MHC isoform, in adult rodents ␣-MHC is the predominant form, probably because of the much greater heart rate of rodents (LeWinter, 2005). Therefore, while -MHC expression is increased in human heart failure it is not as pronounced as the increase seen in rodents. This has complicated the interpretation of results from animal models. Despite the undeniable genetic link between MHC proteins and hypertrophy, recent work has suggested that increased -MHC expression in an individual cell is not necessarily associated with an increase in hypertrophy (Pandya, Kim, & Smithies, 2006). 3.2. Natriuretic peptides The natriuretic peptides (NP) are a family of hormones that affect the cardiovascular and endocrine systems through their actions in diuresis, natriuresis, vasorelaxation and aldosterone and renin inhibition (Nishikimi, Maeda, &
Matsuoka, 2006). The NPs are potent endogenous inhibitors of hypertrophy. There are three members: atrial natriuretic peptide (ANP) also known atrial natriuretic factor (ANF), brain or B-type natriuretic peptide (BNP) and C-type (CNP). In the heart, ANP expression is confined to the atria, BNP is expressed in both the atria and ventricles and the extent of CNP expression in the heart is unclear. Following their secretion, the N-terminals of NPs are cleaved to release the biologically active C-terminal derived peptide and exert their effects by binding to the natriuretic peptide receptors (NPRs), also known as guanylyl cyclase receptors. Binding to the receptor induces guanylyl cyclase activity and subsequent conversion of guanosine triphosphate (GTP) to the second messenger guanosine 3 ,5 -cyclic monophosphate (cGMP) (Gardner, Chen, Glenn, & Grigsby, 2007). NP signalling is terminated either though binding to NPR-C which internalises and degrades NPs thus removing them from the circulation or through cleavage by neutral endoperoxidase (Gardner, 2003; Richards, 2007). A feedback loop also exists where ANP inhibits the expression of its own receptor by cGMP-mediated transcriptional repression (Hum et al., 2004). ANP and BNP are found at high levels during embryonic development and in early neonates but are absent in healthy adults (Gardner, 2003). Hypertrophic stimuli dramatically increase the expression of ANP and BNP through the transcription factor GATA-4 and their main function in the myocardium is to inhibit the hypertrophic response (Gardner et al., 2007; Richards, 2007). Deletion of ANP in mice causes hypertension and hypertrophy under resting conditions, while volume and pressure overload both activate the hypertrophic response in the mutant mice to a greater extent than in wild-type mice (Mori et al., 2004; Wang et al., 2003). When wild-type and ANP−/− mice were fed a low-salt diet, differences in blood pressure disappeared, however ANP−/− mice still had more extensive hypertrophy, demonstrating that the anti-hypertrophic effects of ANP are independent of increased blood pressure (Feng et al., 2003). ANP-deficient mice also had enhanced expression of extracellular matrix (ECM) proteins following hypertrophy, suggesting that ANP may reduce hypertrophic remodelling through inhibition of ECM deposition (Wang et al., 2003). Deletion of BNP, unlike ANP, did not result in hypertrophy or hypertension but caused an increase in interstitial fibrosis in the ventricles which was exacerbated in response to ventricular overload (Tamura et al., 2000). Further evidence for the anti-hypertrophic activity of ANP and BNP comes from studies in NPR-A-deficient mice. NPR-A is the sole receptor for ANP and BNP and deletion of the receptor abolishes their functionality in the cardiovascular system (Lopez, Garbers, & Kuhn, 1997). NPR-A−/− mice suffer from hypertension, interstitial fibrosis, ventricular hypertrophy and heart failure-like symptoms (Kishimoto, Rossi, & Garbers, 2001; Knowles et al., 2001; Oliver et al., 1997). As with the ANP-deficient mice, controlling hypertension through drugs fails to ameliorate the hypertrophic phenotype, again demonstrating that hypertrophy is functionally distinct from elevated blood pressure in the context of reduced natriuretic signalling (Knowles et al., 2001). Specific deletion of NPR-A in the heart using an ␣-MHC-Cre driver definitively showed that natriuretic peptides have
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
a direct effect on the myocardium (Holtwick et al., 2003). These mice exhibited a mild increase in hypertrophy associated with upregulation of ANP, -MHC and ␣-skeletal actin, and hypertrophy following transverse aortic constriction was exacerbated when compared to wild-type controls (Holtwick et al., 2003). Again, these effects were not attributable to differences in BP since the mice were found to be slightly hypotensive. Interestingly, in humans, certain ANP- and BNP-receptor polymorphisms have been associated with left ventricular mass in essential hypertension (Rubattu et al., 2006). In contrast to ANP and BNP, less is known about the role of CNP in hypertrophy. CNP is predominately produced in vascular and endothelial cells where it is important in the regulation of vascular tone, blood flow and pressure (Ahluwalia & Hobbs, 2005). CNP has also been shown to have anti-hypertrophic effects in vitro and at the same concentration appears to be equally as potent as ANP and BNP (Rosenkranz, Woods, Dusting, & Ritchie, 2003; Tokudome et al., 2004). Moreover, CNP administration in vivo inhibits both fibrosis and hypertrophy following coronary artery ligation (Soeki et al., 2005). This result has been confirmed in transgenic mice overexpressing CNP and in transgenic rats expressing a dominant-negative NPR-B, the sole receptor for CNP (Langenickel et al., 2006; Wang et al., 2007). CNP has been shown to be produced by the heart, where levels increase during CHF and indeed CNP may have some prognostic value in terms of disease severity (Del Ry, Passino, Maltinti, Emdin, & Giannessi, 2005; Kalra et al., 2003). The signalling pathways responsible for NP-mediated inhibition of hypertrophy are not entirely established. It is clear that the mitogen-activated protein kinase (MAPK) pathway is involved; ANP was shown to inhibit p38 MAPK and ERK activity in smooth muscle cells in response to angiotensin II and NPR-A-deficient mice have increased basal phosphorylation of p38, ERK and Akt in the heart (Kilic et al., 2005; Sharma et al., 2002). All three NP hormones induce cGMP production which appears to be the common mediator of NPs in hypertrophy. Stimulating adult cardiac myocytes with a cGMP analogue has the same anti-hypertrophic effect as treating with the NPs themselves (Calderone, Thaik, Takahashi, Chang, & Colucci, 1998; Rosenkranz et al., 2003). Similarly, nitric oxide, which induces cGMP via the soluble GC receptor, can block hypertrophy (Calderone et al., 1998). Precisely how increased cGMP production prevents cardiac hypertrophy has not been entirely elucidated but it likely involves cGMPdependent protein kinase-1 (PGK-1) a serine/threonine kinase activated by cGMP. PGK-1 inhibits l-type Ca2+ channels thus reducing Ca2+ transient amplitude and blocking calcineurin-mediated activation of NFAT, a transcription factor that is obligatory for hypertrophy (Fiedler et al., 2002) (Fig. 2). Indeed, pharmacological inhibition of PGK-1 abrogates the effects of BNP and CNP in cardiac myocytes (Calderone et al., 1998). cGMP is normally broken down by phosphodiesterase-5A (PDE5A) and Takimoto et al. recently showed that the PDE5A inhibitor sildenafil (commonly known as Viagra) reduces hypertrophy and improves heart function in response to transverse aortic constriction (TAC) in mice, demonstrating that increased bioavailablility of cGMP has beneficial effects in hypertrophy (Takimoto et
2027
al., 2005). Importantly, sildenafil was able to reverse preestablished hypertrophy, and this was accompanied by activation of PKG-1. Another facet to NP signalling is their effect on intracellular Ca2+ concentrations (Fig. 2). NPR-A-deficient mice have increased activity of the cardiac Na+ /H+ exchanger NHE-1, which increases intracellular pH and [Ca2+ ] and leads in turn to induction of calcineurin and Ca2+ /calmodulin-dependent kinase II (CaMKII) (Kilic et al., 2005). Moreover, pharmacological inhibition of NHE1 reverses the hypertrophic phenotype of NPR-A−/− mice. NPs are also known to inhibit the renin-angiotensin system (RAS), for example pharmacological inhibition of the angiotensin type 1 (AT1) receptor with losartan reverses the left ventricular hypertrophy of NPR-A−/− mice. Upon crossing NPR-A−/− mice onto an AT1-receptor-deficient background, fibrosis is no longer present. However, in these mice, hypertrophy and survival in response to coronary artery ligation remains the same, indicating that NP-mediated RAS inhibition may play a prominent role in causing fibrosis during remodelling (Nakanishi et al., 2005). These studies therefore all clearly demonstrate that ANP, BNP and CNP reduce blood pressure, hypertrophy and fibrosis through multiple pathways. Why then does hypertrophy still occur in patients with high-endogenous levels of antihypertrophic NPs? Possibly they serve to slow down the progression of hypertrophy for a time but are insufficient to halt progression in the setting of prolonged pressure or volume overload. Interestingly, a study from Kuhn’s group suggests that the anti-hypertrophic effect of NPs may be impaired during heart failure. Patients with congestive heart failure were found to have elevated levels of the NP metabolizing NPR-C and produced meagre amounts of cGMP in response to ANP, suggesting hypo responsiveness of NRP-A (Kuhn et al., 2004). When hemodynamic stress was eased with a left ventricular assist device, NPR-A activity was restored and NPR-C levels returned to normal. A recent study has also suggested that NPR-B may have a heretofore-unappreciated role in heart failure. In failing hearts from mice which underwent TAC, CNP induced twice as much guanylyl cyclase activity as ANP, suggesting that NPR-B activity is unaltered in heart failure while NPR-A activity is attenuated (Dickey et al., 2007). The anti-hypertrophic, vasodilatory and diuretic properties of NPs have generated interest in the potential clinical use of NPs. Indeed, human BNP (nesiritide) has been approved by the US FDA in the treatment of heart failure, although a recent meta-analysis suggested that there may be some unappreciated safety issues (Aaronson & SacknerBernstein, 2006; Colucci et al., 2000; Mentzer et al., 2007). Since BNP is highly upregulated during hypertrophy, there has been intense interest in its use as a biomarker for the diagnosis of heart failure. In a multi-centre study of 1586 patients, measuring serum levels of BNP proved very effective—the predictive accuracy was 83.4% using a cut off of 100 pg/ml and using a cut off point of 50 pg/ml, the negative predictive value was 96% (McCullough et al., 2002; Moe, 2006). Plasma BNP levels are also predictive of cardiovascular mortality and heart failure hospitalisation and using BNP levels as a guide in clinical decision making may
2028
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
Fig. 2. Molecular pathways controlling transcription factor activation during hypertrophy. Binding of IL-6 family cytokines (IL-6, CT-1 and LIF) to the heterodimeric gp130 receptor allows recruitment of STAT3 and Gab1. STAT3 translocates to the nucleus while Gab1 binds to SHP2, promoting downstream activity of the Raf/MEK/ERK, PI(3)K/Akt and MEK5/ERK5 pathways and subsequent activation of the NFAT and GATA-4 transcription factors. Negative control of hypertrophy is brought about by binding of ANP and BNP to the NPR-A receptor. This receptor contains guanylyl cyclase activity which promotes accumulation of cGMP as does nitric oxide (NO). cGMP-mediated activation of PKG-1 abrogates calcineurin-mediated NFAT nuclear localisation and also affects Ca2+ levels by inhibiting the cardiac Na+ /H+ exchanger NHE-1.
lead to improved patient outcome (de Denus, Pharand, & Williamson, 2004; Koglin et al., 2001; Moe, 2006). 4. G-protein-coupled receptors G-protein-coupled receptors (GPCRs) are crucial in normal cardiovascular function and in mediating the “fight or flight” response to endogenous catecholamines such as adrenaline, noradrenaline and angiotensin (see Table 1). The adrenergic receptors (ARs) are GPCRs that are highly abundant in the major arteries of the body; including the aorta, pulmonary and coronary arteries and are instrumental in orchestrating the hypertrophic response through control of contractility, vascular tone and metabolism. Like all GPCRs, adrenergic receptors couple to heterodimeric G-proteins which consist of G␣ and G␥ subunits (see Fig. 3). -Adrenergic receptors couple to
G␣s (or simply Gs), inducing adenylyl cyclase activity, accumulation of cAMP and consequently activation of protein kinase A (PKA). PKA phosphorylates several proteins involved in cardiac contraction, including l-type calcium channels, ryanodine receptors, phospholamban and troponin (Marian, 2006), increasing their activity and hence cardiac contraction. ␣-ARs couple to Gq and activate phospholipase C (PLC), resulting in hydrolysis of phosphoinositol 4,5-bisphosphate. The products of this reaction are inositol 1,4,5-triphosphate (IP3 ) and diacyl glycerol (DAG). DAG in turn activates protein kinase C (PKC), an important step in the development of concentric hypertrophy—indeed, inhibition of PKC abrogates GPCR-mediated hypertrophy in mice (Arimoto et al., 2006; Takeishi et al., 2000). Patients with heart failure have elevated circulating catecholamines and increased adrenergic drive which
Table 1 The major distinguishing characteristics of the pathways activated by ␣- and -adrenoreceptor types, and common agonists and antagonists used to distinguish between them Receptor type
Agonists
Antagonists
Coupled G-protein type
Downstream signalling pathways
␣-Adrenergic
Serotonin Angiotensin II Phenylephrine
Phentolamine Prazosin
Gq (G␣q/␣11 )
InsP3 DAG PKC, MAPK
-Adrenergic
Adrenaline Noradrenaline Isoproterenol
Atenalol Propranalol
Gs
Adenyl cyclase PKA
The many different sub-types of ␣- and -receptors are not indicated.
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
2029
Fig. 3. Adrenergic signalling in hypertrophy. Activation of the ␣1 -adrenergic receptor allows coupling of the G-protein Gq and activation of phospholipase C (PLC). PLC promotes hydrolysis of phosphoinositol 4,5-bisphosphate and the products of this reaction are inositol 1,4,5-triphosphate (IP3 ) and diacyl glycerol (DAG). DAG in turn activates protein kinase C (PKC) and MAPK which are involved in hypertrophy and vasoconstriction. -Adrenergic receptors on the other hand couple to Gs, induce adenylyl cyclase and accumulation of the second messenger cAMP. The resulting PKA activity affects several aspects of the hypertrophy program, including phosphorylation and inactivation of the SERCA inhibitor phospholamban. This has the effect of increasing Ca2+ influx into the cytoplasm and increasing Ca2+ re-uptake into the sarcoplasmic reticulum, causing myocyte contraction and vasodilation.
inversely correlate with survival (Lohse, Engelhardt, & Eschenhagen, 2003). This chronic adrenergic drive is one of the hallmarks of hypertrophy. The development of blockers by Sir James Black revolutionized the treatment of heart failure, reducing the overall risk of death by over 30% (Black, Crowther, Shanks, Smith, & Dornhorst, 1964). Treatment with -blockers halts left ventricular remodelling, improves left ventricular ejection fraction and has a positive effect on mortality outcomes (Sabbah, 2004). In patients with DCM, hemodynamic recovery subsequent to -blocker treatment is associated with a restoration of normal ␣-MHC/-MHC balance, reduced ANP expression and increased sarco-/endo-plasmic reticulum Ca2+ -ATPase (SERCA) expression (Lowes et al., 1997). In heart failure, the increased adrenergic drive is initially beneficial through increased contractility and heart rate, however chronic adrenergic stimulation is thought to be ultimately detrimental (Bristow, 2000). This is borne out in studies of transgenic mice overexpressing 1 -ARs, which have increased cardiac contractility when young, but pronounced cardiac hypertrophy and fibrosis when older, which eventually leads to heart failure (Engelhardt, Hein, Wiesmann, & Lohse, 1999). This suggests that short-term 1 -ARs may indeed improve
cardiac function but prolonged hyper-adrenergic drive eventually leads to pathology. Thus, in humans a gain of function 1 -receptor polymorphism leads to greater risk of cardiomyopathies (Mason, Moore, Green, & Liggett, 1999). Despite the increased levels of circulating catecholamines accompanying hypertrophy, heart failure is associated with attenuated -AR signalling as a consequence of receptor desensitisation (Sabbah, 2004). This is brought about by receptor phosphorylation by PKA, PKC or G-protein-coupled receptor kinases (GRKs), leading to receptor internalisation, uncoupling from G-proteins or downregulation of AR mRNA expression (Lohse et al., 2003). While -blockers are undoubtedly the most efficacious class of drugs in the treatment of heart failure, there is still some debate as to the exact nature of their action. They may either combat the negative effects of prolonged -adrenergic stimulation or cause re-sensitization of the -AR system (Lohse et al., 2003). Overexpression of GRK2 in the vasculature reduces sensitivity to -AR vasorelaxation and induces hypertension and hypertrophy in the myocardium (Eckhart, Ozaki, Tevaearai, Rockman, & Koch, 2002), while inhibition of GRK2 can restore normal GPCR signalling and lead to improved outcomes in a mouse heart failure model (Rockman et al., 1998).
2030
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
A plethora of animal studies confirm that adrenergic signalling is a potent hypertrophic stimulus. Adrenergic agonists such as PE, endothelin-1 and angiotensin II have long been recognized as potent inducers of hypertrophy and transgenic overexpression of 1 -ARs, Gs or Gq leads to hypertrophic remodelling and systolic dysfunction (D’Angelo et al., 1997; Engelhardt et al., 1999; Iwase et al., 1996). Likewise, overexpression of angiotensin II (AngII) is sufficient to induce hypertrophy and heart failure in transgenic mice (Paradis, Dali-Youcef, Paradis, Thibault, & Nemer, 2000). Indeed angiotensin-converting enzyme (ACE) inhibitors are comparable to -blockers in treating hypertrophy in the clinic. Intriguingly though, AngII receptor-deficient mice still undergo hypertrophy in response to mechanical stretch, suggesting that other factors are involved (Harada et al., 1998). Several studies have used mice overexpressing a Gq inhibitor peptide (GqI) in cardiomyocytes to demonstrate that TAC-mediated pressure overload requires Gq to induce hypertrophy although the mice suffered fibrosis and apoptosis to the same extent as wild-type mice (Akhter et al., 1998; Wettschureck et al., 2001). Interestingly, these mice remain susceptible to hypertrophy in response to chronic PE, angiotensin II and serotonin (Keys, Greene, Koch, & Eckhart, 2002). In contrast, mice in which expression of GqI was restricted to vascular smooth muscle (VSM) under the control of the SM22␣ promoter were resistant to these forms of hypertrophy (Keys et al., 2002). This may indicate that chronic Gq-mediated signalling induces hypertrophy through its hypertensive effects rather than by directly affecting the myocardium (Keys et al., 2002). This study suggests that it is peripheral resistance and catecholamine production in hypertensive individuals that is largely responsible for cardiac hypertrophy. Interestingly, mechanisms other than hypertrophy can contribute to improved cardiac function following transverse aortic constriction. Mice overexpressing GqI show better recovery from TAC than wild-type controls even though they cannot increase left ventricular size to compensate (Esposito et al., 2002). 5. Interluekin-6 family of cytokines Along with adrenergic agonists, cytokines play a major role in inducing hypertrophy. The main hypertrophic cytokines are all members of the IL-6 family and include IL-6 itself, leukaemia-inhibitory factor (LIF) and cardiotrophin-1 (CT-1). All IL-6 cytokines utilise a common receptor unit glycoprotein 130 (gp130) in combination with ligand-specific receptors and mediate their effects by the JAK/STAT, MAPK and PI(3)K pathways (Fig. 2). In the early nineties, a search for novel mediators of hypertrophy using an expression cloning approach lead to the identification of CT-1 (Pennica et al., 1995). Soon after, CT-1 was shown to promote cardiac myocyte survival and induce an eccentric form of hypertrophy both in vitro and in vivo (Jin et al., 1996; Sheng, Pennica, Wood, & Chien, 1996; Wollert et al., 1996). CT-1 signals through the heterodimeric LIF-R/gp130 receptor and it seems that separate downstream pathways are responsible for the hypertrophic and anti-apoptotic effects of this cytokine. In cardiac myocytes, CT-1 induces the activity of several signalling mediators
including ERK1/2, p38 MAPK, STAT3, PI(3)K/Akt and NFB (Craig, Wagner, McCardle, Craig, & Glembotski, 2001). Blocking the MAPKs, Akt or NF-B abrogated the protective effect of CT-1 against hypoxia, while inhibiting STAT3 blocked CT-1-mediated hypertrophy, clearly delineating a separate signalling pathway in each process (Brar, Stephanou, Pennica, & Latchman, 2001; Craig et al., 2001; Sheng et al., 1997). While the downstream pathways that control CT-1mediated hypertrophy were being deciphered, several studies found that CT-1 levels are increased in animal models of hypertrophy (Ishikawa et al., 1996; Pan et al., 1998) and in human patients (Lopez et al., 2005; Talwar, Squire, Downie, Davies, & Ng, 2000; Talwar, Squire, Downie, O’Brien, et al., 2000; Talwar et al., 2002; Tsutamoto et al., 2001), and in most cases high-CT-1 levels correlate with disease severity. Currently, BNP is one of the most widely used markers of heart failure, however a recent study suggests that combining CT-1 and BNP measurements may be a greater predictor of mortality in patients suffering from congestive heart failure than either marker alone (Tsutamoto et al., 2007). Interestingly, in patients with DCM, ischaemic cardiomyopathy (ICM) or hypertensive heart failure, augmented CT-1 expression is accompanied by downregulation of gp130, suggesting a compensatory mechanism may control excessive IL-6 family cytokine-mediated hypertrophy (Zolk, Ng, O’Brien, Weyand, & Eschenhagen, 2002). CT-1 may reduce gp130 levels by promoting its ubiquitination and degradation (Gonzalez et al., 2007). CT-1 can be induced by adrenergic stimulation via a cAMP response element in the CT-1 promoter (Funamoto et al., 2000) but whether CT-1 contributes to hypertrophic remodelling induced by adrenergic drive is unknown. An interesting experiment therefore would be to examine the extent of adrenergic-induced hypertrophy in mice with a cardiac-specific knockout of CT-1. As well as crosstalk with the adrenergic system, CT-1 may have a role in hypertrophy induced by other GPCRs. Angiotensin II (AngII) treatment induces CT-1 and LIF release from cardiac fibroblasts, while pharmacological inhibition of AngII blocks CT-1 and LIF-mediated hypertrophy (Fukuzawa et al., 2000; Sano et al., 2000). Like CT-1, LIF induces hypertrophy in cultured cardiac cells and leads to rapid tyrosine phosphorylation of gp130, JAK1, STAT1, STAT3 and ERK (Kodama et al., 1997; Kunisada et al., 1996; Villegas, Villarreal, & Dillmann, 2000a). LIF and CT-1 are the only factors known to induce serial assembly of sarcomeres resulting in myocyte elongation, and several downstream pathways are postulated to be involved in this effect. Expression of the dominant-negative MEK5 blocks LIF-induced elongation of cardiac myocytes in culture, and hearts of MEK5 transgenic mice have a similar appearance to hearts of mice treated with LIF or CT-1 (Nicol et al., 2001). As well as promoting addition of sarcomeres in series, LIF/MEK5/ERK5 may also interfere with addition of sarcomeres in parallel and thus may serve as a focal point in the decision of myocytes to undergo eccentric or concentric hypertrophy (Nicol et al., 2001). It is important to keep in mind that chemical inhibitors commonly used to determine the involvement of ERK1/2 in signalling pathways also inhibit MEK5/ERK5 (Nishimoto & Nishida, 2006).
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
LIF is also involved in the altered Ca2+ signalling that accompanies hypertrophy through downregulation of SERCA (Villegas, Villarreal, & Dillmann, 2000b) and phosphorylation of the l-type Ca2+ channel Ca(v)1.2 in cardiac myocytes leading to increases in Ca2+ currents (Takahashi et al., 2004). Ca(v)1.2 phosphorylation is dependent on ERK and results in downstream activation of CaMKII, CaMKIV and calcineurin. The resulting increase in Ca2+ signalling may play a prominent role in LIF-mediated hypertrophy since inhibitors of CaMKII and CaMKIV block LIF-dependent fetal gene activation and cell-size increase (Kato et al., 2000; Takahashi et al., 2004). In addition to ERK1, 2 and 5, LIF promotes STAT3 phosphorylation and inhibition of STAT3 with a dominant-negative construct in cardiac myocytes blocks LIF-mediated hypertrophy (Kunisada et al., 1998). Like CT-1, LIF is induced in response to hemodynamic overload and is upregulated in patients with CHF (Eiken et al., 2001; Tsutamoto et al., 2001). Mice with a cardiac-specific deletion of gp130 are overtly normal but whereas wild-type mice undergo compensatory hypertrophy following TAC, gp130−/− mice experience dilated cardiomyopathy and increased myocyte apoptosis (Hirota et al., 1999). This maladaption to mechanical stress causes all the gp130−/− to die within 7 days of TAC and suggests that the gp130 system controls a critical juncture in the transition from compensatory hypertrophy to heart failure (Hirota et al., 1999). The JAK/STAT pathway (particularly STAT3) appears to be centrally responsible for the biological effects of gp130 stimulation, and overexpression of the JAK inhibitor SOCS3 abolishes gp130 signalling in cardiac myocytes (Yasukawa et al., 2001). Transgenic overexpression of STAT3 in the heart is sufficient to induce hypertrophy (Kunisada et al., 2000); crossing IL-6 transgenic mice with IL-6 receptor transgenic mice showed that chronic activation of IL6/gp130 and STAT3 is sufficient to induce hypertrophy (Hirota, Yoshida, Kishimoto, & Taga, 1995). It seems most plausible that each of the JAK/STAT, PI(3)K and MAPK pathways are necessary for a fully fledged gp130-dependent hypertrophic response but that each pathway controls primarily a separate facet. There are also likely several points of crosstalk and it will be important to delineate precise signalling pathways in order to better tailor pharmaceutical interventions. Since there is evidence that gp130 signalling can have a protective effect in addition to its role in hypertrophy (Berry et al., 2004; Zou et al., 2003), the challenge will lie in dissecting out the protective facets of gp130 signalling from those that are responsible for maladaption to hypertrophic stress.
6. Adhesion and cytoskeletal proteins In addition to the cytoskeletal proteins being primary end-targets of the hypertrophic response, they co-operate with adhesion molecules to sense the initial stress and transmit the hypertrophic signal inside the cells. Overload of the myocardium causes mechanical stress to the myocytes. This cellular stretch signal is transduced by integrins at focal adhesion sites on the cell surface and can result in direct rearrangement of
2031
the actin cytoskeleton. Overload also causes kinases such as the Src tyrosine kinase (Kuppuswamy et al., 1997) and the integrin-linked kinase, ILK-1, to associate with the cytoskeleton and become activated. Overexpression of ILK-1 in mouse myocardium results in a compensated ventricular hypertrophic phenotype whereas mice with cardiomyocyte-restricted expression of a kinase-inactive ILK are apparently resistant to the development of cardiac hypertrophy (Lu et al., 2006). In response to Src activation or ILK-1 overexpression (Kuppuswamy et al., 1997; Lu et al., 2006), the induction of hypertrophy is mediated by certain members of the Ras family of small GTPases. Overexpression of some of these small GTPases is sufficient to induce hypertrophy. For example, overexpression of the prototypical Ras protein in transgenic mice causes development of cardiac hypertrophy and diastolic dysfunction (Hunter, Tanaka, Rockman, Ross, & Chien, 1995). Similarly, cardiac overexpression of V12Rac1 produces profound cardiac hypertrophy and ventricular dilation (Sussman et al., 2000). On the other hand, cardiac overexpression of RhoA leads to atrial enlargement and conduction defects but not ventricular hypertrophy (Sah et al., 1999). Thus, a network of small GTPases act downstream of the cytoskeleton to control the activation of pro-hypertrophic signalling pathways including the JAK/STAT, MAPK, and calcineurin pathways (reviewed in Ruwhof & van der, 2000) described below. 7. Mitogen-activated protein kinase/extracellular signal receptor-regulated kinase signalling A large body of evidence supports a role for MEK and its downstream kinases ERK1/2 (extracellular signalregulated kinases) in the development of hypertrophy. For instance, ERK1/2 are activated in response to practically every known hypertrophic agonist (Bueno & Molkentin, 2002) and transgenic expression of constitutively active MEK1 induces concentric hypertrophy in vivo (Bueno & Molkentin, 2002; Bueno et al., 2000; Clerk, Fuller, Michael, & Sugden, 1998). Similarly, inhibition of the MEK1-ERK1/2 pathway by drugs or adenoviral expression of dominantnegative constructs reduces the hypertrophic response of cardiac myocytes in culture (Bueno et al., 2000; Bueno & Molkentin, 2002; Clerk et al., 1998). Surprisingly however, it was recently shown that the hypertrophic response to either pressure overload stimulation, neuroendocrine agonist infusion or exercise is unaffected in ERK1−/− ERK2+/− mice (Purcell et al., 2007). In order to eliminate the possibility that the remaining ERK2 expression in these mice was sufficient to account for this response, the experiments were repeated in mice in which ERK1/2 was completely inhibited by overexpression of dual-specificity phosphatase-6 (DUSP6) in the heart. Again, there was no effect on the development of hypertrophy (Purcell et al., 2007). It seems that a possible explanation for this observation is that DUSP6 overexpression was driven by an inducible ␣-MHC promoter and was therefore restricted to cardiomyocytes. As explained previously, emerging evidence suggests that GPCR pathways in other cell types such as vascular smooth muscle can also be central to the development of hypertrophy. Furthermore, by analogy with
2032
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
experiments with mice expressing GqI in cardiac myocytes, some hypertrophic stimuli may act via induction of hypertension and/or secondary release of -adrenergic stimuli, which would then act via signalling pathways not requiring MEK-ERK1/2 (see Fig. 3). Interestingly, after long-term transverse aortic constriction, DUSP6 transgenic and ERK1−/− ERK2+/− , mice suffer from cardiac decompression and signs of heart failure associated with enhanced myocyte cell death, indicating that this pathway does play an important role in myocyte survival. Mutations in upstream activators of the ERK pathway are associated with cardiomyopathies in humans. For example, of the patients with Noonan or LEOPARD syndrome with mutations in two mutation hotspots in the serine/threonine kinase RAF1 gene, 95% suffered from hypertrophic cardiomyopathy (Pandit et al., 2007). These mutations impair binding of the inhibitory 14-3-3 protein to RAF1, resulting in insufficient auto-inhibition and thus increased ERK activity. Interestingly, despite the evidence cited above that cardiac ERK activity is not required for the development of hypertrophy, expression of cardiac-specific dominantnegative Raf-1 in mice attenuates hypertrophy after pressure overload (Hunter et al., 1995). As has been pointed out however, Raf-1 directly complexes with many other signalling proteins, including Ras, apoptosis signal-regulating kinase-1 (ASK-1), Rok-␣, cdc25, casein kinase-2, retinoblastoma protein, and MEKK1, any of which could also affect the cardiac hypertrophic response independently of ERK1/2 (Purcell et al., 2007). For example, ASK-1 is also involved in myocyte survival by way of regulating apoptosis, and mice with cardiomyocyte-specific disruption of Raf-1 demonstrate cardiomyocyte-specific apoptosis, left ventricular systolic dysfunction and heart dilatation, all of which are ASK-1 dependent (Yamaguchi et al., 2004). 8. Histone acetylation 8.1. Histone acetyltransferases Histone acetyltransferases (HATs) such as p300 and CREB-binding protein (CBP) are transcriptional coactivators that cause the relaxation of chromatin structure and promote gene activation. Overexpression of CBP/p300 is sufficient to induce hypertrophy and left ventricular remodelling in transgenic mice (Gusterson, Jazrawi, Adcock, & Latchman, 2003; Yanazume, Hasegawa, et al., 2003). Interestingly, cardiac remodelling after myocardial infarction, which involves hypertrophy of the remaining cardiomyocytes, also requires p300 HAT activity (Miyamoto et al., 2006). Acetylation of histones alters gene expression in cardiac cells in response to pharmacologically induced hypertrophy and simulated ischaemia and reperfusion (Davidson et al., 2005; Davis, Pillai, Gupta, & Gupta, 2005; McKinsey & Olson, 2004b)—see Table 2. Treatment of primary neonatal cardiomyocytes with PE causes them to undergo hypertrophy, as demonstrated by an increase in ANP production, protein-to-DNA ratio, mean cell area and length, and sarcomeric organization as well as an increase in the activity of p300 (Davidson et al., 2005; Yanazume, Hasegawa, et al., 2003). A major mechanism of
PE-mediated cardiac hypertrophy therefore appears to be enhancement of p300/CBP HAT activity. Proteins with HAT activity such as p300 and CBP tend to be large, and act as scaffolding proteins that can bridge between basal transcription factors and various transactivators. Thus, it seems likely that CBP/p300 is an essential co-factor for a distinct subset of genes necessary to initiate the hypertrophic response. Acetylation of the GATA-4 transcription factor by CBP/p300 results in eccentric left ventricular hypertrophy (Molkentin & Olson, 1997; Yanazume, Morimoto, Wada, Kawamura, & Hasegawa, 2003). In fact, GATA-4-mediated hypertrophy is an interesting example of how different post-translational modifications on the same protein can have distinct effects on cardiac remodelling, since in contrast to the above, activation of the MEK-ERK pathway by hypertrophic stimuli leads to phosphorylation of GATA-4 at serine 105 and development of concentric hypertrophy. Activation of p300/CBP in response to PE occurs via the downstream protein kinase pathways downstream of Gq that were described above, specifically ERK, p38, MSK1 and PKA (Davidson et al., 2005; Markou, HadzopoulouCladaras, & Lazou, 2004). 8.2. Histone deacetylases Hypertrophic growth involves control of cardiomyocyte gene expression at multiple molecular levels. In addition to their interaction with HATs, master transcriptional regulators of gene expression in cardiac myocytes such as myoD, MEF2, and GATA-4/-5/-6 (Chanalaris et al., 2005; Kolodziejczyk et al., 1999; Liang & Molkentin, 2002) recruit histone deacetylases (HDACs) to remodel chromatin as part of the mechanism by which they control gene expression (Davis et al., 2005; Lu, McKinsey, Nicol, & Olson, 2000; Townsend et al., 2007). In direct opposition to the HATs described above, HDACs mediate the removal of acetyl groups. There are more than a dozen individual HDAC enzymes which can be divided into three main classes—class I HDACs (HDACs 1, 2, 3, and 8), class II HDACs (HDACs 4, 5, 6, 7, 9, and 10) and class III which are discussed in the following section. HDACs have been shown to be important endpoint targets of cell-signalling pathways involved in the induction of altered gene expression in cardiac hypertrophy and ischaemia/reperfusion injury (Frey, Katus, Olson, & Hill, 2004) and are currently undergoing intense investigation as possible therapeutic regulators of hypertrophy-associated cardiac disease. In adult cardiac myocytes activation of the MEF2 transcription factor in response to stress signalling activates a pro-hypertrophic gene expression profile. Stress signalling activates MEF2 by causing the nuclear export of class II HDACs, possibly regulated through protein kinase D (Vega et al., 2004), which would normally associate with MEF2 and suppress its activity in normal cells. Thus, class II HDACs play a key role in suppression of hypertrophy (Kolodziejczyk et al., 1999; Lu et al., 2000; Miska et al., 1999). Consistent with this notion, mice lacking HDAC9 or HDAC 5, class II HDACs, are supersensitive to stress signals and both mouse models showed enhanced hypertrophy in response to pathological stimuli (Chang et al., 2004; Zhang
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
2033
Table 2 p300/CBP histone acetyltransferase acts as a scaffold factor, bridging transcription factors and cofactors, by bringing a number of proteins into a ternary transcriptional complexes which lead to cardiac hypertrophy (a selection of hypertrophic stimuli, acting through p300/CBP and their corresponding transcription cofactors are shown) Hypertrophy agent
Transcription (co)factor
Hypertrophic target protein
Reference
PE, MAPK activation
MyoD MEF2D SRF AP-1 GATA-4, -5, -6 NFAT Nished NF-B MEF2 GATA-4 MEF2 SRF dHAND Nkx2.5 GATA-5, -6 (NFAT)
ANP MHC Desmin
Yanazume, Hasegawa, et al. (2003), Davidson et al. (2005)
MLC-2v
Mathew et al. (2004)
Cardiac actin ANP ET-1
Molinari et al. (2004) Molkentin and Olson (1997), Yanazume, Morimoto, et al. (2003)
AngII Isoproterenol infusion Emb1 overexpression Pressure overload
(SERCA)
et al., 2002). In contrast, class I HDACs are considered to play a pro-hypertrophic role although the relevant gene targets are less well described (McKinsey & Olson, 2004a; Townsend et al., 2007). Taken together, class I and II HDACs play opposing roles in control of hypertrophy and there is a clear benefit to the development of class I HDAC-specific inhibitors as therapeutic agents which would not interfere with the class II-dependent inhibition of pro-hypertrophic pathways (Fig. 4). It is anticipated that the clinical use of HDAC inhibitors would allow the control of key hypertrophic genes, and would provide a novel molecular and therapeutic approach. The development of HDAC inhibitors as experimental therapeutic agents for the treatment of cancer has progressed well, although most of these agents are not isoform specific. By contrast, the bicyclic tetrapeptide HDAC inhibitors FK228 and Spiruchostatin A appear to possess selectivity towards class I HDACs (unpublished data; YurekGeorge et al., 2007), an activity profile which may make
Fig. 4. The role of histone acetylation in regulating cardiac hypertrophy. Histones play a key role in packaging DNA in chromatin and the acetylation of lysine in histone protein subunits modulates the “open” or “closed” status of the DNA. Chromatin “opening”, or relaxation, permits the access of transcriptional machinery to potential hypertrophy-regulating genes, such as the anti-hypertrophic GSK3 gene or pro-hypertrophic ANP gene. The regulation of lysine acetylation is governed by histone deacetylases (HDACs) and histone acetylases (HATs). Class I and II HDACs play key opposing roles in modulating cardiac hypertrophy. Class II HDACs are thought to repress whereas class I HDACs induce pro-hypertrophic genes. A novel molecular therapy might aim to target and inhibit class I HDACs thereby regulating cardiac hypertrophy.
them particularly attractive as starting points to develop novel therapies for cardiovascular disease. Indeed, it has already been shown that Spiruchostatin A is a highly potent inhibitor (∼pM) of HDACs in cardiac myocytes and effectively interferes with the pro-hypertrophic effects of phenylephrine and urocortin (Davis et al., 2005).
8.3. Class III HDACs role in cardiac hypertrophy Class III HDACs, sirtuins, have an impact on a range of cellular processes, which include gene silencing, cellular ageing and genotoxic/DNA damage repair (Longo & Kennedy, 2006). Class III HDACs are unique in that they require nicotinamide adenine dinucleotide (NAD) for catalytic activity, as such they are thought to function as rheostats of metabolism and accordingly stress. Recently, class III HDAC activity has been associated with inhibition of cardiac hypertrophy and enhanced cardiac myocyte survival. Sir2␣ is expressed in the nuclei of cardiac myocytes and its overexpression has been shown to reduce apoptosis in response to serum starvation and also to significantly increase the size of cardiac myocytes (Alcendor, Kirshenbaum, Imai, Vatner, & Sadoshima, 2004). Conversely, inhibition of Sir2␣ with nicotinamide, sirtinol, or by expression of dominant-negative Sir2␣ induced myocyte apoptosis. An in vivo cardioprotective role for Sir2 expression was seen in dogs where levels were increased significantly in hearts with heart failure induced by rapid pacing superimposed on stable, severe hypertrophy. A direct role for Sir2␣ in cardiac myocyte hypertrophy is suggested by experiments demonstrating that it modifies chromatin structure by deacetylating histone protein H2A.z (Chen I.Y. et al., 2006). Recent animal studies using Sirt1, the mammalian homologue of yeast Sir2␣, showed that its overexpression induces ␣-MHC expression. Cardiac Sirt1 levels also increase during cardiac hypertrophy following pressure overload (Alcendor et al., 2004; Alcendor et al., 2007; Pillai et al., 2008). Strikingly, however, moderate overex-
2034
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
pression of Sirt1 attenuated age-dependent increases in cardiac hypertrophy, apoptosis, cardiac dysfunction, and expression of senescence markers, and therefore seems to cause a paradoxical cardioprotective response. However, high-level expression of Sirt1 promotes apoptosis, hypertrophy and decreased cardiac function, thereby stimulating the development of cardiomyopathy. These observations suggest that Sirt1 can impede aging and confer stress resistance to the heart in vivo, yet these beneficial effects are dose/expression dependent. Moreover, a transcriptional influence of Sirt1 in a model of ventricular remodelling in type 2 diabetes, a disorder which predisposes to fatal coronary heart disease, has shown a role for forkhead transcription factors (Vahtola et al., 2008). In Goto-Kakizaki rats Sirt1 modulation of the proapoptotic forkhead class O transcription factor 3a (FOXO3a) regulates the cardiac myocyte lifespan and hypertrophy. Clearly the sirtuins, have a remarkable role in the heart which is yet to be fully understood. These genetic studies demonstrate that sirtuins, if carefully regulated, can repress cardiac hypertrophy, reducing the hearts sensitivity to stress, and promote longevity. 9. Calcium-mediated pathways towards hypertrophy Calcium increase is another potential trigger of the translocation of pro-hypertrophic transcription factors to the nucleus. Nuclear factor of activated T cells (NFAT) was originally discovered as a transcription factor involved in the development of T cells. However it is also involved in the development of the heart, as well as skeletal muscle and the nervous system. As per many transcription factors involved in cardiac development, it is also activated during hypertrophy. In this case it is normally retained in the cytosol, but when dephosphorylated by calcineurin it translocates to the nucleus (Fig. 2). Calcineurin is a serine-threonine protein phosphatase that is activated in the presence of calcium, and can also be affected by the redox state of the cell (reviewed in Wilkins & Molkentin, 2004). Since calcineurin is a target of the immunosuppressive drugs ciclosporin A and FK506, these drugs have been used to evaluate the role of the calcineurin/NFAT pathway in rodent models of cardiac hypertrophy (Wilkins & Molkentin, 2004). However, since these drugs also affect NFAT, more specific inhibition of calcineurin by cardiac-specific overexpression of endogenous inhibitors Cain/Cabin-1 or A-kinase anchoring protein 79 has been used to more specifically demonstrate the involvement of calcineurin in hypertrophy (De Windt et al., 2001). Furthermore, genetic deletion of specific isoforms of calcineurin or NFAT attenuates development of hypertrophy. NFAT appears to serve as an integrator of many different signalling pathways, some of which (GSK3, JNK, p38, PKA, MEKK1, CK1) cause its nuclear export, and others of which (AP1, MEF-2, GATA-4) interact with and modulate its transcriptional activity as co-factors (reviewed in Crabtree & Olson, 2002). A link between calcium and the regulation of hypertrophic transcriptional pathways by acetylation has recently been discovered. In this case, it appears to be the nuclear export of HDAC4 (Backs, Song, Bezprozvannaya,
Chang, & Olson, 2006) that is increased after phosphorylation by calcium/calmodulin kinase II. Thus, an increase in cellular calcium results in the balance tipping in favour of histone acetylation, and therefore the loosening of inhibitory nucleosomal structure. Impaired calcium homeostasis is a prominent feature in the transition from compensatory hypertrophy to heart failure and causes contractile dysfunction and development of arrhythmias. It is now well appreciated that downregulation of the SERCA is a major cause of dysregulated Ca2+ signalling and impaired potentiation of contractile force in heart failure (Frank, Bolck, Erdmann, ¨ & Schwinger, 2003; Kogler et al., 2006; Meyer et al., 1995). Several studies have demonstrated that SERCA expression and hence Ca2+ SR re-uptake is reduced in overloadinduced hypertrophy and heart failure (Frank et al., 2003; Mercadier et al., 1990; Schultz et al., 2004). Treating neonatal cardiac myocytes with an ␣1 -adrenergic agonist induces a hypertrophic response in parallel with a reduction in SERCA2 levels (Prasad et al., 2007). This is accompanied by a reduction in SERCA activity and downregulation of Ca2+ signalling. SERCA-deficient mice are not viable but the effect of deleting a single allele shows that SERCA is necessary for adaptation to pressure overload. SERCA2+/− mice have reduced levels of SR Ca2+ uptake and release and have a much greater propensity towards developing heart failure (Schultz et al., 2004). They also exhibit greater hypertrophy and more extensive fibrosis following 10 weeks of TAC-induced pressure overload (Schultz et al., 2004). Likewise, inhibition of phospholamban blocks hypertrophy and improves cardiac function in rodents and has been suggested as a therapeutic strategy to combat hypertrophy in patients (Hoshijima et al., 2002; Minamisawa et al., 1999; Sato et al., 2001). However, loss of a functional phospholamban allele in humans actually results in dilated cardiomyopathy, suggesting that increasing SR Ca2+ uptake may not always be a positive goal in treating hypertrophy (Haghighi et al., 2003). Using right ventricular myocar¨ dial muscle preparations, Kogler et al. showed that SERCA2 is downregulated in unloaded conditions but upregulated by prolonged preload, which is an adaptive response to ¨ increased demand (Kogler et al., 2006). While the desirability of targeting SERCA for therapy is still under debate it is clear that altered levels of SERCA affects the heart’s ability to adapt to chronic pressure overload and studies consistently show that SERCA has a prominent role to play in the progression to heart failure. 10. MicroRNAs—new players in cardiac hypertrophy MicroRNAs (miRNA) are non-coding RNAs 18–25 nucleotides in length whose function is to functionally silence-specific mRNA transcripts. There are over 400 known miRNAs in the human genome, a number which is increasing weekly (Kim, 2005). In the past 12 months, a series of papers have placed miRNAs at the centre stage in regulating the hypertrophic program. Several studies have employed a microarray approach to detect miRNAs differentially expressed during hypertrophy. Van Rooij and colleagues identified 28 differentially expressed miRNAs common to TAC and calcineurin A-mediated hypertrophy
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
and found that many of these were also overexpressed in failing human hearts (van Rooij et al., 2006). Moreover, several of these miRNAs have the ability to induce hypertrophy when expressed in cardiac myocytes in culture. A transgenic approach revealed that myocardial overexpression of miR-195 in mice causes an increase in left ventricular wall size, upregulation of ANP, BNP and -MHC and reduced cardiac output (van Rooij et al., 2006). Three separate groups analysed miRNA expression at various times after aortic banding in mice and found dozens of differentially expressed miRNAs (Cheng et al., 2007; Sayed, Hong, Chen, Lypowy, & Abdellatif, 2007; Tatsuguchi et al., 2007). Thum et al. analysed the miRNA expression profiles from heart failure patients and found a high degree of similarity to fetally expressed miRNAs (Thum et al., 2007). Care` et al. focused their studies on miR-1 and miR-133 which had previously been shown to have roles in skeletal myoblast differentiation and proliferation (Chen J.F. et al., 2006). They utilized three separate models of hypertrophy: TAC, transgenic expression of constitutively active Akt and exercise. In all three models both miR-1 and miR-133 were downregulated, a finding which was recapitulated in hearts from patients with DCM (Care` et al., 2007). Functional studies revealed that in culture, either miRNA could block the induction of hypertrophy, while in vivo, inhibition of miR-133 using an oligonucleotide “antagomir” had the opposite effect. The authors highlight possible targets of miR-133 such as RhoA, Cdc42 and NELFA/Whsc2 which have previously been shown to have roles in hypertrophic remodelling. Expression of miR-1 in culture inhibits serum-induced hypertrophy and is associated with inhibition of RasGAP, Cdk9 and fibronectin expression (Sayed et al., 2007). Elegant work from the Olson lab demonstrated that in mice, co-ordinate regulation of ␣- and -MHC is controlled by miR-208 located in intron 27 of the ␣-MHC gene (van Rooij et al., 2007). Specific deletion of miR208 resulted in compromised cardiac function and sarcomeric abnormalities. When these mice were subjected to TAC there was little sign of increased myocyte size or fibrosis. Moreover the miR-208-deficient mice failed to upregulate -MHC but instead increased ␣-MHC expression to compensate. In contrast, ANP and BNP were upregulated as in wild-type mice, demonstrating that miR-208 is needed for -MHC upregulation and cardiac remodelling but not for induction of other stress response factors. Intriguingly, miR-208’s role in hypertrophy may involve silencing of thyroid receptorassociated protein 1 (THRAP1), a previously unappreciated target of miR-208 (van Rooij et al., 2007). This silencing of THRAP1 modulates the activity of thyroid hormone and in turn expression of -MHC. While understanding the role of each miRNA in hypertrophy is a few years away, these early studies have clearly shown that miRNAs can control myocyte hypertrophy in vitro and in vivo. Although some reports have corroborated the role of a specific miRNA – for example two groups independently reported that miR-1 blocked hypertrophy – other reports conflict. For example, both pro- and antihypertrophic effects have been claimed for miR-21 (Cheng et al., 2007; Tatsuguchi et al., 2007). As is often the case in an emerging field of study, there is initially much conflict-
2035
ing data and controversy. Nonetheless, exciting times are ahead in understanding the role of microRNAs in cardiac pathology. Taken together, these recent reports show that miRNA play a powerful role in co-ordinating the response to cardiac stress and are sure to open the door to previously unappreciated medical therapies. 11. Conclusion By extension of the general observation that cardiac “hypertrophy recapitulates ontogeny”, one might anticipate that other cardiac developmental signalling pathways would be integral to hypertrophy. Indeed, emerging evidence suggests that the Wnt/-catenin pathway is involved in hypertrophy, although, its role appears to more complicated than simply that of an on/off activator/repressor. Again, this appears to reflect its multi-faceted roles in cardiac development, during the early stages of which activation is required for commitment to the cardiac lineage, while downregulation is required for later stages of differentiation (reviewed in Zelarayan, Gehrke, & Bergmann, 2007). Dissection and characterisation of the signalling pathways leading to cardiac hypertrophy has led to a wealth of knowledge about this condition both physiological and pathological (Heineke & Molkentin, 2006). Although these pathways have still to be defined further and there are undoubtedly future pathways and players to be discovered; the challenge will be to translate this scientific knowledge and understanding into potential pharmacological therapies for the treatment of pathological cardiac hypertrophy. Acknowledgements The authors would like to thank all of the laboratories cited within this review for their insightful and exceptional research investigating cardiac hypertrophy—we would also like to apologise to those researchers whose data we could not discuss due to space constraints. Research mentioned from the authors own laboratories was funded by the Biotechnology and Biological Sciences Research Council (PAT), the British Heart Foundation (PAT), the Gerald Kerkut Charitable Trust (PAT) and the Medical Research Council (SMD). SPB is funded by a British Heart Foundation PhD studentship. References Aaronson KD, Sackner-Bernstein J. Risk of death associated with nesiritide in patients with acutely decompensated heart failure. J Am Med Assoc 2006;296:1465–6. Ahluwalia A, Hobbs AJ. Endothelium-derived C-type natriuretic peptide: More than just a hyperpolarizing factor. Trends Pharmacol Sci 2005;26:162–7. Akhter SA, Luttrell LM, Rockman HA, Iaccarino G, Lefkowitz RJ, Koch WJ. Targeting the receptor-Gq interface to inhibit in vivo pressure overload myocardial hypertrophy. Science 1998;280:574– 7. Alcendor RR, Gao SM, Zhai PY, Zablocki D, Holle E, Yu XZ, et al. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res 2007;100:1512–21. Alcendor RR, Kirshenbaum LA, Imai S, Vatner SF, Sadoshima J. Silent information regulator 2 alpha, a longevity factor and class III histone deacetylase, is an essential endogenous apoptosis inhibitor in cardiac myocytes. Circ Res 2004;95:971–80.
2036
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
Arimoto T, Takeishi Y, Takahashi H, Shishido T, Niizeki T, Koyama Y, et al. Cardiac-specific overexpression of diacylglycerol kinase zeta prevents Gq protein-coupled receptor agonist-induced cardiac hypertrophy in transgenic mice. Circulation 2006;113:60–6. Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN. CaM kinase II selectively signals to histone deacetylase 4 during cardiomyocyte hypertrophy. J Clin Investig 2006;116:1853–64. Berry MF, Pirolli TJ, Jayasankar V, Morine KJ, Moise MA, Fisher O, et al. Targeted overexpression of leukemia inhibitory factor to preserve myocardium in a rat model of postinfarction heart failure. J Thorac Cardiovasc Surg 2004;128:866–75. Black JW, Crowther AF, Shanks RG, Smith LH, Dornhorst AC. A new adrenergic betareceptor antagonist. Lancet 1964;1:1080–1. Boudina S, Abel ED. Diabetic cardiomyopathy revisited. Circulation 2007;115:3213–23. Brar BK, Stephanou A, Pennica D, Latchman DS. CT-1 mediated cardioprotection against ischaemic re-oxygenation injury is mediated by PI3 kinase, Akt and MEK1/2 pathways. Cytokine 2001;16:93–6. Bristow MR. beta-adrenergic receptor blockade in chronic heart failure. Circulation 2000;101:558–69. Bueno OF, De Windt LJ, Tymitz KM, Witt SA, Kimball TR, Klevitsky R, et al. The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J 2000;19:6341–50. Bueno OF, Molkentin JD. Involvement of extracellular signal-regulated kinases 1/2 in cardiac hypertrophy and cell death. Circ Res 2002;91:776–81. Calderone A, Thaik CM, Takahashi N, Chang DL, Colucci WS. Nitric oxide, atrial natriuretic peptide, and cyclic GMP inhibit the growthpromoting effects of norepinephrine in cardiac myocytes and fibroblasts. J Clin Investig 1998;101:812–8. Care` A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, et al. MicroRNA133 controls cardiac hypertrophy. Nat Med 2007;13:613–8. Chanalaris A, Lawrence KM, Townsend PA, Davidson S, Jamshidi Y, Stephanou A, et al. Hypertrophic effects of urocortin homologous peptides are mediated via activation of the Akt pathway. Biochem Biophys Res Commun 2005;328:442–8. Chang AN, Potter JD. Sarcomeric protein mutations in dilated cardiomyopathy. Heart Fail Rev 2005;10:225–35. Chang S, McKinsey TA, Zhang CL, Richardson JA, Hill JA, Olson EN. Histone deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress signals and play redundant roles in heart development. Mol Cell Biol 2004;24:8467–76. Chen IY, Lypowy J, Pain J, Sayed D, Grinberg S, Alcendor RR, et al. Histone H2A.z is essential for cardiac myocyte hypertrophy but opposed by silent information regulator 2 alpha. J Biol Chem 2006a;281:19369– 77. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet 2006b;38:228–33. Cheng Y, Ji R, Yue J, Yang J, Liu X, Chen H, et al. MicroRNAs are aberrantly expressed in hypertrophic heart: Do they play a role in cardiac hypertrophy? Am J Pathol 2007;170:1831–40. Clerk A, Fuller SJ, Michael A, Sugden PH. Stimulation of “stress-regulated” mitogen-activated protein kinases (stress-activated protein kinases/cJun N-terminal kinases and p38-mitogen-activated protein kinases) in perfused rat hearts by oxidative and other stresses. J Biol Chem 1998;273:7228–34. Colucci WS, Elkayam U, Horton DP, Abraham WT, Bourge RC, Johnson AD, et al. Intravenous nesiritide, a natriuretic peptide, in the treatment of decompensated congestive heart failure. Nesiritide Study Group. N Engl J Med 2000;343:246–53. Crabtree GR, Olson EN. NFAT signaling: Choreographing the social lives of cells. Cell 2002;109 Suppl.:S67–79. Craig R, Wagner M, McCardle T, Craig AG, Glembotski CC. The cytoprotective effects of the glycoprotein 130 receptor-coupled cytokine, cardiotrophin-1, require activation of NF-kappa B. J Biol Chem 2001;276:37621–9. Cuda G, Fananapazir L, Epstein ND, Sellers JR. The in vitro motility activity of beta-cardiac myosin depends on the nature of the beta-myosin heavy chain gene mutation in hypertrophic cardiomyopathy. J Muscle Res Cell Motil 1997;18:275–83. D’Angelo DD, Sakata Y, Lorenz JN, Boivin GP, Walsh RA, Liggett SB, et al. Transgenic Galphaq overexpression induces cardiac contractile failure in mice. Proc Natl Acad Sci USA 1997;94:8121–6. Davidson SM, Townsend PA, Carroll C, Yurek-George A, Balasubramanyam K, Kundu TK, et al. The transcriptional coactivator p300 plays a critical role in the hypertrophic and protective pathways induced by phenylephrine in cardiac cells but is specific to the hypertrophic effect of urocortin. ChemBioChem 2005;6:162–70.
Davis FJ, Pillai JB, Gupta M, Gupta MP. Concurrent opposite effects of trichostatin A, an inhibitor of histone deacetylases, on expression of alpha-MHC and cardiac tubulins: Implication for gain in cardiac muscle contractility. Am J Physiol Heart Circ Physiol 2005;288:H1477–90. de Denus S, Pharand C, Williamson DR. Brain natriuretic peptide in the management of heart failure: The versatile neurohormone. Chest 2004;125:652–68. Del Ry S, Passino C, Maltinti M, Emdin M, Giannessi D. C-type natriuretic peptide plasma levels increase in patients with chronic heart failure as a function of clinical severity. Eur J Heart Fail 2005;7:1145–8. De Windt LJ, Lim HW, Bueno OF, Liang Q, Delling U, Braz JC, et al. Targeted inhibition of calcineurin attenuates cardiac hypertrophy in vivo. Proc Natl Acad Sci USA 2001;98:3322–7. Dickey DM, Flora DR, Bryan PM, Xu X, Chen Y, Potter LR. Differential regulation of membrane guanylyl cyclases in congestive heart failure: Natriuretic peptide receptor (NPR)-B, not NPR-A, is the predominant natriuretic peptide receptor in the failing heart. Endocrinology 2007;148:3518–22. Du CK, Morimoto S, Nishii K, Minakami R, Ohta M, Tadano N, et al. Knock-in mouse model of dilated cardiomyopathy caused by troponin mutation. Circ Res 2007;101:185–94. Eckhart AD, Ozaki T, Tevaearai H, Rockman HA, Koch WJ. Vascular-targeted overexpression of G protein-coupled receptor kinase-2 in transgenic mice attenuates beta-adrenergic receptor signaling and increases resting blood pressure. Mol Pharmacol 2002;61:749–58. Eiken HG, Oie E, Damas JK, Yndestad A, Bjerkeli V, Aass H, et al. Myocardial gene expression of leukaemia inhibitory factor, interleukin-6 and glycoprotein 130 in end-stage human heart failure. Eur J Clin Investig 2001;31:389–97. Engelhardt S, Hein L, Wiesmann F, Lohse MJ. Progressive hypertrophy and heart failure in beta1-adrenergic receptor transgenic mice. Proc Natl Acad Sci USA 1999;96:7059–64. Esposito G, Rapacciuolo A, Naga Prasad SV, Takaoka H, Thomas SA, Koch WJ, et al. Genetic alterations that inhibit in vivo pressure-overload hypertrophy prevent cardiac dysfunction despite increased wall stress. Circulation 2002;105:85–92. Feng JA, Perry G, Mori T, Hayashi T, Oparil S, Chen YF. Pressure-independent enhancement of cardiac hypertrophy in atrial natriuretic peptidedeficient mice. Clin Exp Pharmacol Physiol 2003;30:343–9. Fiedler B, Lohmann SM, Smolenski A, Linnemuller S, Pieske B, Schroder F, et al. Inhibition of calcineurin-NFAT hypertrophy signaling by cGMPdependent protein kinase type I in cardiac myocytes. Proc Natl Acad Sci USA 2002;99:11363–8. Frank KF, Bolck B, Erdmann E, Schwinger RH. Sarcoplasmic reticulum Ca2+ ATPase modulates cardiac contraction and relaxation. Cardiovasc Res 2003;57:20–7. Frey N, Katus HA, Olson EN, Hill JA. Hypertrophy of the heart: A new therapeutic target? Circulation 2004;109:1580–9. Fukuzawa J, Booz GW, Hunt RA, Shimizu N, Karoor V, Baker KM, et al. Cardiotrophin-1 increases angiotensinogen mRNA in rat cardiac myocytes through STAT3: An autocrine loop for hypertrophy. Hypertension 2000;35:1191–6. Funamoto M, Fujio Y, Kunisada K, Negoro S, Tone E, Osugi T, et al. Signal transducer and activator of transcription 3 is required for glycoprotein 130-mediated induction of vascular endothelial growth factor in cardiac myocytes. J Biol Chem 2000;275:10561–6. Gardner DG. Natriuretic peptides: Markers or modulators of cardiac hypertrophy? Trends Endocrinol Metab 2003;14:411–6. Gardner DG, Chen S, Glenn DJ, Grigsby CL. Molecular biology of the natriuretic peptide system: Implications for physiology and hypertension. Hypertension 2007;49:419–26. Geisterfer-Lowrance AA, Kass S, Tanigawa G, Vosberg HP, McKenna W, Seidman CE, et al. A molecular basis for familial hypertrophic cardiomyopathy: A beta cardiac myosin heavy chain gene missense mutation. Cell 1990;62:999–1006. Gomes AV, Harada K, Potter JD. A mutation in the N-terminus of troponin I that is associated with hypertrophic cardiomyopathy affects the Ca(2+)-sensitivity, phosphorylation kinetics and proteolytic susceptibility of troponin. J Mol Cell Cardiol 2005;39:754–65. Gonzalez A, Ravassa S, Loperena I, Lopez B, Beaumont J, Querejeta R, et al. Association of depressed cardiac gp130-mediated antiapoptotic pathways with stimulated cardiomyocyte apoptosis in hypertensive patients with heart failure. J Hypertens 2007;25:2148–57. Gusterson RJ, Jazrawi E, Adcock IM, Latchman DS. The transcriptional coactivators CREB-binding protein (CBP) and p300 play a critical role in cardiac hypertrophy that is dependent on their histone acetyltransferase activity. J Biol Chem 2003;278:6838–47. Haghighi K, Kolokathis F, Pater L, Lynch RA, Asahi M, Gramolini AO, et al. Human phospholamban null results in lethal dilated cardiomyopa-
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039 thy revealing a critical difference between mouse and human. J Clin Investig 2003;111:869–76. Harada K, Komuro I, Shiojima I, Hayashi D, Kudoh S, Mizuno T, et al. Pressure overload induces cardiac hypertrophy in angiotensin II type 1A receptor knockout mice. Circulation 1998;97:1952–9. Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol 2006;7:589–600. Hirota H, Chen J, Betz UA, Rajewsky K, Gu Y, Ross Jr J, et al. Loss of a gp130 cardiac muscle cell survival pathway is a critical event in the onset of heart failure during biomechanical stress. Cell 1999;97:189–98. Hirota H, Yoshida K, Kishimoto T, Taga T. Continuous activation of gp130, a signal-transducing receptor component for interleukin 6-related cytokines, causes myocardial hypertrophy in mice. Proc Natl Acad Sci USA 1995;92:4862–6. Holtwick R, van Eickels M, Skryabin BV, Baba HA, Bubikat A, Begrow F, et al. Pressure-independent cardiac hypertrophy in mice with cardiomyocyte-restricted inactivation of the atrial natriuretic peptide receptor guanylyl cyclase-A. J Clin Investig 2003;111:1399– 407. Hoshijima M, Ikeda Y, Iwanaga Y, Minamisawa S, Date MO, Gu Y, et al. Chronic suppression of heart-failure progression by a pseudophosphorylated mutant of phospholamban via in vivo cardiac rAAV gene delivery. Nat Med 2002;8:864–71. Hum D, Besnard S, Sanchez R, Devost D, Gossard F, Hamet P, et al. Characterization of a cGMP-response element in the guanylyl cyclase/natriuretic peptide receptor A gene promoter. Hypertension 2004;43:1270–8. Hunter JJ, Tanaka N, Rockman HA, Ross Jr J, Chien KR. Ventricular expression of a MLC-2v-ras fusion gene induces cardiac hypertrophy and selective diastolic dysfunction in transgenic mice. J Biol Chem 1995;270:23173–8. Ishikawa M, Saito Y, Miyamoto Y, Kuwahara K, Ogawa E, Nakagawa O, et al. cDNA cloning of rat cardiotrophin-1 (CT-1): Augmented expression of CT-1 gene in ventricle of genetically hypertensive rats. Biochem Biophys Res Commun 1996;219:377–81. Iwase M, Bishop SP, Uechi M, Vatner DE, Shannon RP, Kudej RK, et al. Adverse effects of chronic endogenous sympathetic drive induced by cardiac GS alpha overexpression. Circ Res 1996;78:517–24. Izumo S, Lompre AM, Matsuoka R, Koren G, Schwartz K, Nadal-Ginard B, et al. Myosin heavy chain messenger RNA and protein isoform transitions during cardiac hypertrophy. Interaction between hemodynamic and thyroid hormone-induced signals. J Clin Investig 1987;79:970–7. Jin H, Yang R, Keller GA, Ryan A, Ko A, Finkle D, et al. In vivo effects of cardiotrophin-1. Cytokine 1996;8:920–6. Kalra PR, Clague JR, Bolger AP, Anker SD, Poole-Wilson PA, Struthers AD, et al. Myocardial production of C-type natriuretic peptide in chronic heart failure. Circulation 2003;107:571–3. Kamisago M, Sharma SD, DePalma SR, Solomon S, Sharma P, McDonough B, et al. Mutations in sarcomere protein genes as a cause of dilated cardiomyopathy. N Engl J Med 2000;343:1688–96. Karkkainen S, Peuhkurinen K. Genetics of dilated cardiomyopathy. Ann Med 2007;39:91–107. Kato T, Sano M, Miyoshi S, Sato T, Hakuno D, Ishida H, et al. Calmodulin kinases II and IV and calcineurin are involved in leukemia inhibitory factor-induced cardiac hypertrophy in rats. Circ Res 2000;87:937– 45. Keys JR, Greene EA, Koch WJ, Eckhart AD. Gq-coupled receptor agonists mediate cardiac hypertrophy via the vasculature. Hypertension 2002;40:660–6. Kilic A, Velic A, De Windt LJ, Fabritz L, Voss M, Mitko D, et al. Enhanced activity of the myocardial Na+ /H+ exchanger NHE-1 contributes to cardiac remodeling in atrial natriuretic peptide receptor-deficient mice. Circulation 2005;112:2307–17. Kim VN. MicroRNA biogenesis: Coordinated cropping and dicing. Nat Rev Mol Cell Biol 2005;6:376–85. Kishimoto I, Rossi K, Garbers DL. A genetic model provides evidence that the receptor for atrial natriuretic peptide (guanylyl cyclase-A) inhibits cardiac ventricular myocyte hypertrophy. Proc Natl Acad Sci USA 2001;98:2703–6. Knowles JW, Esposito G, Mao L, Hagaman JR, Fox JE, Smithies O, et al. Pressure-independent enhancement of cardiac hypertrophy in natriuretic peptide receptor A-deficient mice. J Clin Investig 2001;107:975–84. Kodama H, Fukuda K, Pan J, Makino S, Baba A, Hori S, et al. Leukemia inhibitory factor, a potent cardiac hypertrophic cytokine, activates the JAK/STAT pathway in rat cardiomyocytes. Circ Res 1997;81:656–63. ¨ Kogler H, Schott P, Toischer K, Milting H, Van PN, Kohlhaas M, et al. Relevance of brain natriuretic peptide in preload-dependent regulation of cardiac sarcoplasmic reticulum Ca2+ ATPase expression. Circulation 2006;113:2724–32.
2037
Koglin J, Pehlivanli S, Schwaiblmair M, Vogeser M, Cremer P, vonScheidt W. Role of brain natriuretic peptide in risk stratification of patients with congestive heart failure. J Am Coll Cardiol 2001;38:1934–41. Kolodziejczyk SM, Wang L, Balazsi K, DeRepentigny Y, Kothary R, Megeney LA. MEF2 is upregulated during cardiac hypertrophy and is required for normal post-natal growth of the myocardium. Curr Biol 1999;9:1203–6. Kuhn M, Voss M, Mitko D, Stypmann J, Schmid C, Kawaguchi N, et al. Left ventricular assist device support reverses altered cardiac expression and function of natriuretic peptides and receptors in end-stage heart failure. Cardiovasc Res 2004;64:308–14. Kunisada K, Hirota H, Fujio Y, Matsui H, Tani Y, Yamauchi-Takihara K, et al. Activation of JAK-STAT and MAP kinases by leukemia inhibitory factor through gp130 in cardiac myocytes. Circulation 1996;94:2626– 32. Kunisada K, Negoro S, Tone E, Funamoto M, Osugi T, Yamada S, et al. Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicininduced cardiomyopathy. Proc Natl Acad Sci USA 2000;97:315– 9. Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K, Kishimoto T. Activation of gp130 transduces hypertrophic signals via STAT3 in cardiac myocytes. Circulation 1998;98:346–52. Kuppuswamy D, Kerr C, Narishige T, Kasi VS, Menick DR, Cooper G. Association of tyrosine-phosphorylated c-Src with the cytoskeleton of hypertrophying myocardium. J Biol Chem 1997;272:4500–8. Langenickel TH, Buttgereit J, Pagel-Langenickel I, Lindner M, Monti J, Beuerlein K, et al. Cardiac hypertrophy in transgenic rats expressing a dominant-negative mutant of the natriuretic peptide receptor B. Proc Natl Acad Sci USA 2006;103:4735–40. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med 1990;322:1561–6. LeWinter MM. Functional consequences of sarcomeric protein abnormalities in failing myocardium. Heart Fail Rev 2005;10:249–57. Liang Q, Molkentin JD. Divergent signaling pathways converge on GATA4 to regulate cardiac hypertrophic gene expression. J Mol Cell Cardiol 2002;34:611–6. Lohse MJ, Engelhardt S, Eschenhagen T. What is the role of beta-adrenergic signaling in heart failure? Circ Res 2003;93:896–906. Longo VD, Kennedy BK. Sirtuins in aging and age-related disease. Cell 2006;126:257–68. Lopez B, Gonzalez A, Lasarte JJ, Sarobe P, Borras F, Diaz A, et al. Is plasma cardiotrophin-1 a marker of hypertensive heart disease? J Hypertens 2005;23:625–32. Lopez MJ, Garbers DL, Kuhn M. The guanylyl cyclase-deficient mouse defines differential pathways of natriuretic peptide signaling. J Biol Chem 1997;272:23064–8. Lowes BD, Gilbert EM, Abraham WT, Minobe WA, Larrabee P, Ferguson D, et al. Myocardial gene expression in dilated cardiomyopathy treated with beta-blocking agents. N Engl J Med 2002;346:1357–65. Lowes BD, Minobe W, Abraham WT, Rizeq MN, Bohlmeyer TJ, Quaife RA, et al. Changes in gene expression in the intact human heart. Downregulation of alpha-myosin heavy chain in hypertrophied, failing ventricular myocardium. J Clin Investig 1997;100:2315–24. Lowey S. Functional consequences of mutations in the myosin heavy chain at sites implicated in familial hypertrophic cardiomyopathy. Trends Cardiovasc Med 2002;12:348–54. Lu H, Fedak PW, Dai X, Du C, Zhou YQ, Henkelman M, et al. Integrin-linked kinase expression is elevated in human cardiac hypertrophy and induces hypertrophy in transgenic mice. Circulation 2006;114:2271–9. Lu J, McKinsey TA, Nicol RL, Olson EN. Signal-dependent activation of the MEF2 transcription factor by dissociation from histone deacetylases. Proc Natl Acad Sci USA 2000;97:4070–5. Mahdavi V, Lompre AM, Chambers AP, Nadal-Ginard B. Cardiac myosin heavy chain isozymic transitions during development and under pathological conditions are regulated at the level of mRNA availability. Eur Heart J 1984;5(Suppl. F):181–91. Marian AJ. Beta-adrenergic receptors signaling and heart failure in mice, rabbits and humans. J Mol Cell Cardiol 2006;41:11–3. Markou T, Hadzopoulou-Cladaras M, Lazou A. Phenylephrine induces activation of CREB in adult rat cardiac myocytes through MSK1 and PKA signaling pathways. J Mol Cell Cardiol 2004;37:1001–11. Mason DA, Moore JD, Green SA, Liggett SB. A gain-of-function polymorphism in a G-protein coupling domain of the human beta1-adrenergic receptor. J Biol Chem 1999;274:12670–4. Mathew S, Mascareno E, Siddiqui MA. A ternary complex of transcrip´ and NFATc4, and co-activator p300 bound to an tion factors, Nished
2038
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039
intronic sequence, intronic regulatory element, is pivotal for the upregulation of myosin light chain-2v gene in cardiac hypertrophy. J Biol Chem 2004;279(39):41018–27. McCullough PA, Nowak RM, McCord J, Hollander JE, Herrmann HC, Steg PG, et al. B-type natriuretic peptide and clinical judgment in emergency diagnosis of heart failure: Analysis from Breathing Not Properly (BNP) Multinational Study. Circulation 2002;106:416–22. McKinsey TA, Olson EN. Cardiac histone acetylation—Therapeutic opportunities abound. Trends Genet 2004a;20:206–13. McKinsey TA, Olson EN. Dual roles of histone deacetylases in the control of cardiac growth. Novartis Found Symp 2004b;259:132– 41. Mentzer Jr RM, Oz MC, Sladen RN, Graeve AH, Hebeler Jr RF, Luber Jr JM, et al. Effects of perioperative nesiritide in patients with left ventricular dysfunction undergoing cardiac surgery: The NAPA Trial. J Am Coll Cardiol 2007;49:716–26. Mercadier JJ, Lompre AM, Duc P, Boheler KR, Fraysse JB, Wisnewsky C, et al. Altered sarcoplasmic reticulum Ca2(+)-ATPase gene expression in the human ventricle during end-stage heart failure. J Clin Investig 1990;85:305–9. Metzger JM, Westfall MV. Covalent and noncovalent modification of thin filament action: The essential role of troponin in cardiac muscle regulation. Circ Res 2004;94:146–58. Meyer M, Schillinger W, Pieske B, Holubarsch C, Heilmann C, Posival H, et al. Alterations of sarcoplasmic reticulum proteins in failing human dilated cardiomyopathy. Circulation 1995;92:778–84. Minamisawa S, Hoshijima M, Chu G, Ward CA, Frank K, Gu Y, et al. Chronic phospholamban-sarcoplasmic reticulum calcium ATPase interaction is the critical calcium cycling defect in dilated cardiomyopathy. Cell 1999;99:313–22. Miska EA, Karlsson C, Langley E, Nielsen SJ, Pines J, Kouzarides T. HDAC4 deacetylase associates with and represses the MEF2 transcription factor. EMBO J 1999;18:5099–107. Miyamoto S, Kawamura T, Morimoto T, Ono K, Wada H, Kawase Y, et al. Histone acetyltransferase activity of p300 is required for the promotion of left ventricular remodeling after myocardial infarction in adult mice in vivo. Circulation 2006;113:679–90. Moe GW. B-type natriuretic peptide in heart failure. Curr Opin Cardiol 2006;21:208–14. ¨ Molinari S, Relaix F, Lemonnier M, Kirschbaum B, Schafer B, Buckingham M. A novel complex regulates cardiac actin gene expression through interaction of Emb, a class VI POU domain protein, MEF2D, and the histone transacetylase p300. Mol Cell Biol 2004;24(7):2944– 57. Molkentin JD, Olson EN. GATA4: A novel transcriptional regulator of cardiac hypertrophy? Circulation 1997;96:3833–5. Mori T, Chen YF, Feng JA, Hayashi T, Oparil S, Perry GJ. Volume overload results in exaggerated cardiac hypertrophy in the atrial natriuretic peptide knockout mouse. Cardiovasc Res 2004;61:771–9. Murphy RT, Mogensen J, Shaw A, Kubo T, Hughes S, McKenna WJ. Novel mutation in cardiac troponin I in recessive idiopathic dilated cardiomyopathy. Lancet 2004;363:371–2. Nakanishi M, Saito Y, Kishimoto I, Harada M, Kuwahara K, Takahashi N, et al. Role of natriuretic peptide receptor guanylyl cyclase-A in myocardial infarction evaluated using genetically engineered mice. Hypertension 2005;46:441–7. Nicol RL, Frey N, Olson EN. From the sarcomere to the nucleus: Role of genetics and signaling in structural heart disease. Annu Rev Genom Hum Genet 2000;1:179–223. Nicol RL, Frey N, Pearson G, Cobb M, Richardson J, Olson EN. Activated MEK5 induces serial assembly of sarcomeres and eccentric cardiac hypertrophy. EMBO J 2001;20:2757–67. Nishikimi T, Maeda N, Matsuoka H. The role of natriuretic peptides in cardioprotection. Cardiovasc Res 2006;69:318–28. Nishimoto S, Nishida E. MAPK signalling: ERK5 versus ERK1/2. EMBO Rep 2006;7:782–6. Oliver PM, Fox JE, Kim R, Rockman HA, Kim HS, Reddick RL, et al. Hypertension, cardiac hypertrophy, and sudden death in mice lacking natriuretic peptide receptor A. Proc Natl Acad Sci USA 1997;94:14730–5. Palmiter KA, Tyska MJ, Haeberle JR, Alpert NR, Fananapazir L, Warshaw DM. R403Q and L908V mutant beta-cardiac myosin from patients with familial hypertrophic cardiomyopathy exhibit enhanced mechanical performance at the single molecule level. J Muscle Res Cell Motil 2000;21:609–20. Pan J, Fukuda K, Kodama H, Sano M, Takahashi T, Makino S, et al. Involvement of gp130-mediated signaling in pressure overload-induced activation of the JAK/STAT pathway in rodent heart. Heart Vessels 1998;13:199–208.
Pandit B, Sarkozy A, Pennacchio LA, Carta C, Oishi K, Martinelli S, et al. Gainof-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. Nat Genet 2007;39:1007–12. Pandya K, Kim HS, Smithies O. Fibrosis, not cell size, delineates betamyosin heavy chain reexpression during cardiac hypertrophy and normal aging in vivo. Proc Natl Acad Sci USA 2006;103:16864–9. Paradis P, Dali-Youcef N, Paradis FW, Thibault G, Nemer M. Overexpression of angiotensin II type I receptor in cardiomyocytes induces cardiac hypertrophy and remodeling. Proc Natl Acad Sci USA 2000;97:931–6. Pennica D, Shaw KJ, Swanson TA, Moore MW, Shelton DL, Zioncheck KA, et al. Cardiotrophin-1. Biological activities and binding to the leukemia inhibitory factor receptor/gp130 signaling complex. J Biol Chem 1995;270:10915–22. Pillai JB, Chen M, Rajamohan SB, Samant S, Pillai VB, Gupta M, et al. Activation of SIRT1, a class III histone deacetylase, contributes to fructose feeding mediated induction of the ␣-myosin heavy chain expression. Am J Physiol Heart Circ Physiol 2008;294(3):H1388–97. Prasad AM, Ma H, Sumbilla C, Lee DI, Klein MG, Inesi G. Phenylephrine hypertrophy, Ca2+ -ATPase (SERCA2), and Ca2+ signaling in neonatal rat cardiac myocytes. Am J Physiol Cell Physiol 2007;292:C2269– 75. Purcell NH, Wilkins BJ, York A, Saba-El-Leil MK, Meloche S, Robbins J, et al. Genetic inhibition of cardiac ERK1/2 promotes stress-induced apoptosis and heart failure but has no effect on hypertrophy in vivo. Proc Natl Acad Sci USA 2007;104:14074–9. Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: A strong hypothesis. Heart Fail Rev 2007;12:331–43. Richard P, Charron P, Carrier L, Ledeuil C, Cheav T, Pichereau C, et al. Hypertrophic cardiomyopathy: Distribution of disease genes, spectrum of mutations, and implications for a molecular diagnosis strategy. Circulation 2003;107:2227–32. Richards AM. Natriuretic peptides: Update on peptide release, bioactivity, and clinical use. Hypertension 2007;50:25–30. Rockman HA, Chien KR, Choi DJ, Iaccarino G, Hunter JJ, Ross Jr J, et al. Expression of a beta-adrenergic receptor kinase 1 inhibitor prevents the development of myocardial failure in gene-targeted mice. Proc Natl Acad Sci USA 1998;95:7000–5. Rosenkranz AC, Woods RL, Dusting GJ, Ritchie RH. Antihypertrophic actions of the natriuretic peptides in adult rat cardiomyocytes: Importance of cyclic GMP. Cardiovasc Res 2003;57:515–22. Rubattu S, Bigatti G, Evangelista A, Lanzani C, Stanzione R, Zagato L, et al. Association of atrial natriuretic peptide and type a natriuretic peptide receptor gene polymorphisms with left ventricular mass in human essential hypertension. J Am Coll Cardiol 2006;48:499–505. Ruwhof C, van der LA. Mechanical stress-induced cardiac hypertrophy: Mechanisms and signal transduction pathways. Cardiovasc Res 2000;47:23–37. Sabbah HN. Biologic rationale for the use of beta-blockers in the treatment of heart failure. Heart Fail Rev 2004;9:91–7. Sah VP, Minamisawa S, Tam SP, Wu TH, Dorn GW, Ross Jr J, et al. Cardiac-specific overexpression of RhoA results in sinus and atrioventricular nodal dysfunction and contractile failure. J Clin Investig 1999;103:1627–34. Sano M, Fukuda K, Kodama H, Pan J, Saito M, Matsuzaki J, et al. Interleukin-6 family of cytokines mediate angiotensin II-induced cardiac hypertrophy in rodent cardiomyocytes. J Biol Chem 2000;275:29717– 23. Sato Y, Kiriazis H, Yatani A, Schmidt AG, Hahn H, Ferguson DG, et al. Rescue of contractile parameters and myocyte hypertrophy in calsequestrin overexpressing myocardium by phospholamban ablation. J Biol Chem 2001;276:9392–9. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res 2007;100:416–24. Schultz JJ, Glascock BJ, Witt SA, Nieman ML, Nattamai KJ, Liu LH, et al. Accelerated onset of heart failure in mice during pressure overload with chronically decreased SERCA2 calcium pump activity. Am J Physiol Heart Circ Physiol 2004;286:H1146–53. Sharma GD, Nguyen HT, Antonov AS, Gerrity RG, von Geldern T, Pandey KN. Expression of atrial natriuretic peptide receptor-A antagonizes the mitogen-activated protein kinases (Erk2 and P38MAPK) in cultured human vascular smooth muscle cells. Mol Cell Biochem 2002;233:165–73. Sheng Z, Knowlton K, Chen J, Hoshijima M, Brown JH, Chien KR. Cardiotrophin 1 (CT-1) inhibition of cardiac myocyte apoptosis via a mitogen-activated protein kinase-dependent pathway. Divergence from downstream CT-1 signals for myocardial cell hypertrophy. J Biol Chem 1997;272:5783–91.
S.P. Barry et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2023–2039 Sheng Z, Pennica D, Wood WI, Chien KR. Cardiotrophin-1 displays early expression in the murine heart tube and promotes cardiac myocyte survival. Development 1996;122:419–28. Soeki T, Kishimoto I, Okumura H, Tokudome T, Horio T, Mori K, et al. C-type natriuretic peptide, a novel antifibrotic and antihypertrophic agent, prevents cardiac remodeling after myocardial infarction. J Am Coll Cardiol 2005;45:608–16. Sussman MA, Welch S, Walker A, Klevitsky R, Hewett TE, Price RL, et al. Altered focal adhesion regulation correlates with cardiomyopathy in mice expressing constitutively active rac1. J Clin Investig 2000;105:875–86. Takahashi E, Fukuda K, Miyoshi S, Murata M, Kato T, Ita M, et al. Leukemia inhibitory factor activates cardiac l-type Ca2+ channels via phosphorylation of serine 1829 in the rabbit Cav1.2 subunit. Circ Res 2004;94:1242–8. Takeishi Y, Ping P, Bolli R, Kirkpatrick DL, Hoit BD, Walsh RA. Transgenic overexpression of constitutively active protein kinase C epsilon causes concentric cardiac hypertrophy. Circ Res 2000;86:1218–23. Takimoto E, Champion HC, Li M, Belardi D, Ren S, Rodriguez ER, et al. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med 2005;11:214–22. Talwar S, Squire IB, Downie PF, Davies JE, Ng LL. Plasma N terminal probrain natriuretic peptide and cardiotrophin 1 are raised in unstable angina. Heart 2000a;84:421–4. Talwar S, Squire IB, Downie PF, O’Brien RJ, Davies JE, Ng LL. Elevated circulating cardiotrophin-1 in heart failure: Relationship with parameters of left ventricular systolic dysfunction. Clin Sci (Lond) 2000b;99:83–8. Talwar S, Squire IB, O’Brien RJ, Downie PF, Davies JE, Ng LL. Plasma cardiotrophin-1 following acute myocardial infarction: Relationship with left ventricular systolic dysfunction. Clin Sci (Lond) 2002;102:9–14. Tamura N, Ogawa Y, Chusho H, Nakamura K, Nakao K, Suda M, et al. Cardiac fibrosis in mice lacking brain natriuretic peptide. Proc Natl Acad Sci USA 2000;97:4239–44. Tatsuguchi M, Seok HY, Callis TE, Thomson JM, Chen JF, Newman M, et al. Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol 2007;42:1137–41. Thum T, Galuppo P, Wolf C, Fiedler J, Kneitz S, van Laake LW, et al. MicroRNAs in the human heart: A clue to fetal gene reprogramming in heart failure. Circulation 2007;116:258–67. Tokudome T, Horio T, Soeki T, Mori K, Kishimoto I, Suga S, et al. Inhibitory effect of C-type natriuretic peptide (CNP) on cultured cardiac myocyte hypertrophy: Interference between CNP and endothelin-1 signaling pathways. Endocrinology 2004;145:2131–40. Townsend PA, Davidson SM, Packham G, Ganesan A. The bicyclic depsipeptide family of histone deacetylase inhibitors. In: Schreiber SL, Kapoor T, Wess G, editors. Chemical Biology: From Small Molecules to Systems Biology and Drug Design, Vol. 1. Wiley-VCH; 2007, ISBN 3527311505 p. 693–722. Tsutamoto T, Asai S, Tanaka T, Sakai H, Nishiyama K, Fujii M, et al. Plasma level of cardiotrophin-1 as a prognostic predictor in patients with chronic heart failure. Eur J Heart Fail 2007;9:1032–7. Tsutamoto T, Wada A, Maeda K, Mabuchi N, Hayashi M, Tsutsui T, et al. Relationship between plasma level of cardiotrophin-1 and left ventricular mass index in patients with dilated cardiomyopathy. J Am Coll Cardiol 2001;38:1485–90. Vahtola E, Louhelainen, Merasto S, Martonen E, Penttinen S, Aahos I, et al. Forkhead class O transcription factor 3a activation and Sirtuin1 overexpression in the hypertrophied myocardium of the diabetic Goto-Kakizaki rat. J Hypertens 2008;26:334–44. van Rooij E, Sutherland LB, Liu N, Williams AH, McAnally J, Gerard RD, et al. A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA 2006;103:18255–60.
2039
van Rooij E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN. Control of stress-dependent cardiac growth and gene expression by a microRNA. Science 2007;316:575–9. Vega RB, Harrison BC, Meadows E, Roberts CR, Papst PJ, Olson EN, et al. Protein kinases C and D mediate agonist-dependent cardiac hypertrophy through nuclear export of histone deacetylase 5. Mol Cell Biol 2004;24:8374–85. Villegas S, Villarreal FJ, Dillmann WH. Leukemia Inhibitory Factor and Interleukin-6 downregulate sarcoplasmic reticulum Ca2+ ATPase (SERCA2) in cardiac myocytes. Basic Res Cardiol 2000b;95:47–54. Villegas S, Villarreal FJ, Dillmann WH. Leukemia inhibitory factor and interleukin-6 downregulate sarcoplasmic reticulum Ca2+ ATPase (SERCA2) in cardiac myocytes. Basic Res Cardiol 2000a;95:47–54. Wakatsuki T, Schlessinger J, Elson EL. The biochemical response of the heart to hypertension and exercise. Trends Biochem Sci 2004;29:609–17. Wang D, Oparil S, Feng JA, Li P, Perry G, Chen LB, et al. Effects of pressure overload on extracellular matrix expression in the heart of the atrial natriuretic peptide-null mouse. Hypertension 2003;42:88–95. Wang Y, de Waard MC, Sterner-Kock A, Stepan H, Schultheiss HP, Duncker DJ, et al. Cardiomyocyte-restricted over-expression of C-type natriuretic peptide prevents cardiac hypertrophy induced by myocardial infarction in mice. Eur J Heart Fail 2007;9:548–57. Wettschureck N, Rutten H, Zywietz A, Gehring D, Wilkie TM, Chen J, et al. Absence of pressure overload induced myocardial hypertrophy after conditional inactivation of Galphaq/Galpha11 in cardiomyocytes. Nat Med 2001;7:1236–40. Wilkins BJ, Molkentin JD. Calcium-calcineurin signaling in the regulation of cardiac hypertrophy. Biochem Biophys Res Commun 2004;322:1178–91. Wollert KC, Taga T, Saito M, Narazaki M, Kishimoto T, Glembotski CC, et al. Cardiotrophin-1 activates a distinct form of cardiac muscle cell hypertrophy. Assembly of sarcomeric units in series VIA gp130/leukemia inhibitory factor receptor-dependent pathways. J Biol Chem 1996;271:9535–45. Yamaguchi O, Watanabe T, Nishida K, Kashiwase K, Higuchi Y, Takeda T, et al. Cardiac-specific disruption of the c-raf-1 gene induces cardiac dysfunction and apoptosis. J Clin Investig 2004;114:937–43. Yanazume T, Hasegawa K, Morimoto T, Kawamura T, Wada H, Matsumori A, et al. Cardiac p300 is involved in myocyte growth with decompensated heart failure. Mol Cell Biol 2003a;23:3593–606. Yanazume T, Morimoto T, Wada H, Kawamura T, Hasegawa K. Biological role of p300 in cardiac myocytes. Mol Cell Biochem 2003b;248: 115–9. Yasukawa H, Hoshijima M, Gu Y, Nakamura T, Pradervand S, Hanada T, et al. Suppressor of cytokine signaling-3 is a biomechanical stress-inducible gene that suppresses gp130-mediated cardiac myocyte hypertrophy and survival pathways. J Clin Investig 2001;108:1459–67. Yurek-George A, Cecil ARL, Mo AHK, Wen SJ, Rogers H, Habens F, et al. The first biologically active synthetic analogues of FK228, the depsipeptide histone deacetylase inhibitor. J Med Chem 2007;50:5720–6. Zelarayan L, Gehrke C, Bergmann MW. Role of beta-catenin in adult cardiac remodeling. Cell Cycle 2007;6:2120–6. Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN. Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell 2002;110:479–88. Zolk O, Ng LL, O’Brien RJ, Weyand M, Eschenhagen T. Augmented expression of cardiotrophin-1 in failing human hearts is accompanied by diminished glycoprotein 130 receptor protein abundance. Circulation 2002;106:1442–6. Zou Y, Takano H, Mizukami M, Akazawa H, Qin Y, Toko H, et al. Leukemia inhibitory factor enhances survival of cardiomyocytes and induces regeneration of myocardium after myocardial infarction. Circulation 2003;108:748–53.