7
Circulation Journal Official Journal of the Japanese Circulation Society http://www.j-circ.or.jp yclic GMP (cGMP) is the intracellular second messen- ger that mediates the action of nitric oxide (NO) and natriuretic peptides (NPs). 1,2 It regulates a broad array of physiologic processes in the cardiovascular system, includ- ing cardiac contractility, vascular tone, platelet function, and cardiac and vascular remodeling. 1 NO activates soluble guany- lyl cyclase (GC) and NP activates membrane-bound GC, to produce cGMP. 2 cGMP exerts its physiological action through cGMP-dependent protein kinase (PKG), cGMP-regulated phos- phodiesterases (PDE2, PDE3) and cGMP-gated cation chan- nels, among which PKG might be the primary mediator. Impor- tantly, the cGMP signal is compartmentalized within a cell so that specific targeted proteins can be regulated by the same “generic” cGMP to exert differential physiological effects. 3 Key players in this regulation are the cyclic nucleotide degrad- ing PDEs that are localized to specific subcellular space with confined activity. 3 This review highlights the cardiac regulation of cGMP signaling and its role in cardiac pathophysiology, focusing on cardiac myocytes (Figure). cGMP Generation, Degradation, and Its Effector Kinase Generation Via 2 Distinct Pathways NO-sGC-cGMP Synthetic Pathway NO is a gaseous sig- naling molecule with a half-life of several seconds. Many key biological signals of NO are mediated by cGMP, while NO also directly modifies protein function via covalent attachment of NO to reduced thiol groups, known as S-nitrosylation. 4 NO is endogenously synthesized from L-arginine by the catalytic action of NO synthases (NOS), and it can be released from nitrovasodilators such as glyceryl trinitrate (nitroglycerin). 5 There are 3 isozymes of NO synthase, including neuronal NOS (nNOS or NOS1), inducible NOS (iNOS or NOS2) and endo- thelial NOS (eNOS or NOS3). 6 eNOS and nNOS are expressed constitutively and activated by Ca 2+ /calmodulin, whereas iNOS is induced by (inflammatory) stimuli and its activity is Ca 2+ independent. eNOS is highly expressed in endothelial cells, whereas nNOS is abundant in neurons and skeletal muscle. However, many tissues express all 3 isoforms, including car- diac myocytes. NO activates soluble GC (sGC), a heterodi- meric enzyme consisting of α- and β- subunits and a prosthetic heme group with a ferrous iron, and produces cGMP. 7 This enzyme activity is critically affected by redox status: oxidation of the heme moiety on the β-subunit renders the enzyme in- sensitive to NO. Oxidative stress could compromise this cGMP synthetic pathway by other mechanisms as well. NO forms a reactive oxidant peroxynitrite (ONOO) 8 in the presence of superoxide (O 2− ), and NOS becomes dysfunctional (uncoupled) under oxidative stress to produce O 2− rather than NO. 9 NPs-pGC-cGMP Synthetic Pathway The NPs are a family of structurally related polypeptide hormones, including atrial NP (ANP), B-type NP (BNP) and C-type NP (CNP). 1,10 ANP and BNP are derived primarily from the atria and ventricles of the heart, respectively, whereas CNP is secreted by endothelial cells. NPs bind to membrane-associated GC receptors (pGC or rGC) and produce cGMP. Among the 7 particulate GCs identified, GC-A (also called NP A receptor or NPR-A) and GC-B (also called NPR-B) are important in the cardiovascular system. GC-A is the dominant form, expressed in various tis- C Received May 17, 2012; revised manuscript received June 7, 2012; accepted June 12, 2012; released online June 29, 2012 Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA Mailing address: Eiki Takimoto, MD, PhD, FAHA, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 850, Baltimore, MD 21205, USA. E-mail: [email protected] ISSN-1346-9843 doi:10.1253/circj.CJ-12-0664 All rights are reserved to the Japanese Circulation Society. For permissions, please e-mail: [email protected] Cyclic GMP-Dependent Signaling in Cardiac Myocytes Eiki Takimoto, MD, PhD Cyclic GMP (cGMP) and its effector kinase PKG regulate diverse cellular functions. In cardiac myocytes, cGMP is produced by soluble and particulate guanylyl cyclases (GCs), the former stimulated by nitric oxide and the latter by natriuretic peptides, and is hydrolyzed to inactive 5-GMP by cGMP-phosphodiesterases (PDEs). cGMP-PKG modulates cardiac contractility, hypertrophy and remodeling, and exerts cardioprotection. Although early research efforts have mostly focused on cGMP synthetic pathways, recent studies have revealed that cGMP degradation controlled by PDEs plays a critical role in the physiological action of cGMP. Among several cGMP-PDEs, cGMP- specific PDE5 has been intensively investigated. Studies in experimental animal models and humans consistently demonstrate benefits from PDE5 inhibitors in various cardiac pathologies. Several clinical trials are ongoing or planned to test the efficacy of PDE5 inhibitors in human heart disease, including a large multicenter clinical trial (RELAX) led by the NIH evaluating sildenafil efficacy in heart failure with preserved ejection fraction. This review underscores the current understanding of cGMP-PKG signal regulation and its pathophysiological role in the heart, focusing on cardiac myocytes. Key Words: Cyclic GMP (cGMP); cGMP-dependent protein kinase; Myocytes; Phosphodiesterases REVIEW Advance Publication by-J-STAGE

ARTÍCULO-Cyclic GMP-Dependent Signaling in Cardiac Myocytes

Embed Size (px)

DESCRIPTION

ARTÍCULO-Cyclic GMP-Dependent Signaling in Cardiac Myocytes

Citation preview

  • Circulation JournalOfficial Journal of the Japanese Circulation Societyhttp://www.j-circ.or.jp

    yclic GMP (cGMP) is the intracellular second messen-ger that mediates the action of nitric oxide (NO) and natriuretic peptides (NPs).1,2 It regulates a broad array

    of physiologic processes in the cardiovascular system, includ-ing cardiac contractility, vascular tone, platelet function, and cardiac and vascular remodeling.1 NO activates soluble guany-lyl cyclase (GC) and NP activates membrane-bound GC, to produce cGMP.2 cGMP exerts its physiological action through cGMP-dependent protein kinase (PKG), cGMP-regulated phos-phodiesterases (PDE2, PDE3) and cGMP-gated cation chan-nels, among which PKG might be the primary mediator. Impor-tantly, the cGMP signal is compartmentalized within a cell so that specific targeted proteins can be regulated by the same generic cGMP to exert differential physiological effects.3 Key players in this regulation are the cyclic nucleotide degrad-ing PDEs that are localized to specific subcellular space with confined activity.3 This review highlights the cardiac regulation of cGMP signaling and its role in cardiac pathophysiology, focusing on cardiac myocytes (Figure).

    cGMP Generation, Degradation, and Its Effector Kinase

    Generation Via 2 Distinct PathwaysNO-sGC-cGMP Synthetic Pathway NO is a gaseous sig-

    naling molecule with a half-life of several seconds. Many key biological signals of NO are mediated by cGMP, while NO also directly modifies protein function via covalent attachment of NO to reduced thiol groups, known as S-nitrosylation.4 NO is endogenously synthesized from L-arginine by the catalytic

    action of NO synthases (NOS), and it can be released from nitrovasodilators such as glyceryl trinitrate (nitroglycerin).5 There are 3 isozymes of NO synthase, including neuronal NOS (nNOS or NOS1), inducible NOS (iNOS or NOS2) and endo-thelial NOS (eNOS or NOS3).6 eNOS and nNOS are expressed constitutively and activated by Ca2+/calmodulin, whereas iNOS is induced by (inflammatory) stimuli and its activity is Ca2+ independent. eNOS is highly expressed in endothelial cells, whereas nNOS is abundant in neurons and skeletal muscle. However, many tissues express all 3 isoforms, including car-diac myocytes. NO activates soluble GC (sGC), a heterodi-meric enzyme consisting of - and - subunits and a prosthetic heme group with a ferrous iron, and produces cGMP.7 This enzyme activity is critically affected by redox status: oxidation of the heme moiety on the -subunit renders the enzyme in-sensitive to NO. Oxidative stress could compromise this cGMP synthetic pathway by other mechanisms as well. NO forms a reactive oxidant peroxynitrite (ONOO)8 in the presence of superoxide (O2), and NOS becomes dysfunctional (uncoupled) under oxidative stress to produce O2 rather than NO.9

    NPs-pGC-cGMP Synthetic Pathway The NPs are a family of structurally related polypeptide hormones, including atrial NP (ANP), B-type NP (BNP) and C-type NP (CNP).1,10 ANP and BNP are derived primarily from the atria and ventricles of the heart, respectively, whereas CNP is secreted by endothelial cells. NPs bind to membrane-associated GC receptors (pGC or rGC) and produce cGMP. Among the 7 particulate GCs identified, GC-A (also called NP A receptor or NPR-A) and GC-B (also called NPR-B) are important in the cardiovascular system. GC-A is the dominant form, expressed in various tis-

    C

    Received May 17, 2012; revised manuscript received June 7, 2012; accepted June 12, 2012; released online June 29, 2012Division of Cardiology, Johns Hopkins University, Baltimore, MD, USAMailing address: Eiki Takimoto, MD, PhD, FAHA, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 850,

    Baltimore, MD 21205, USA. E-mail: [email protected] doi: 10.1253/circj.CJ-12-0664All rights are reserved to the Japanese Circulation Society. For permissions, please e-mail: [email protected]

    CyclicGMP-DependentSignalinginCardiacMyocytesEiki Takimoto, MD, PhD

    Cyclic GMP (cGMP) and its effector kinase PKG regulate diverse cellular functions. In cardiac myocytes, cGMP is produced by soluble and particulate guanylyl cyclases (GCs), the former stimulated by nitric oxide and the latter by natriuretic peptides, and is hydrolyzed to inactive 5-GMP by cGMP-phosphodiesterases (PDEs). cGMP-PKG modulates cardiac contractility, hypertrophy and remodeling, and exerts cardioprotection. Although early research efforts have mostly focused on cGMP synthetic pathways, recent studies have revealed that cGMP degradation controlled by PDEs plays a critical role in the physiological action of cGMP. Among several cGMP-PDEs, cGMP-specific PDE5 has been intensively investigated. Studies in experimental animal models and humans consistently demonstrate benefits from PDE5 inhibitors in various cardiac pathologies. Several clinical trials are ongoing or planned to test the efficacy of PDE5 inhibitors in human heart disease, including a large multicenter clinical trial (RELAX) led by the NIH evaluating sildenafil efficacy in heart failure with preserved ejection fraction. This review underscores the current understanding of cGMP-PKG signal regulation and its pathophysiological role in the heart, focusing on cardiac myocytes.

    Key Words: Cyclic GMP (cGMP); cGMP-dependent protein kinase; Myocytes; Phosphodiesterases

    REVIEW

    Advance Publication by-J-STAGE

  • TAKIMOTO E

    sues, including cardiac myocytes, and the principal receptor for ANP and BNP.11 GC-B, a receptor for CNP, is mainly ex-pressed in the brain and vascular tissues, but some studies have suggested its presence and role in cardiac myocytes.12,13 NPs-pGC-cGMP synthesis is compromised under disease condi-tions such as heart failure, because of the desensitization of GC-A.14

    Degradation by PDEsThe PDEs are enzymes that hydrolyze cyclic nucleotides (cGMP, cAMP) to the inactive linear form (5-GMP, 5-AMP), thereby regulating both the duration and the amplitude of cy-clic nucleotide signaling. PDEs comprise a 21-gene superfam-ily categorized into 11 families (PDE1PDE11).15 PDEs 1, 2, 3, 4, 5, 8 and 9 are expressed in the heart (Table) and the roles for PDEs 1, 2, 3 and 5 in cardiac cGMP regulation have been investigated. PDE2 and PDE5 harbor allosteric cGMP-binding sites, named GAF domains, located in the N-terminus portion. cGMP-binding affinity to the GAF domain is much higher than to PDE catalytic sites, and cGMP binds to the GAF do-main before cyclic nucleotide catalytic action to induce the active state.15

    PDE1 PDE1 is a dual substrate PDE and is potently ac-

    tivated by Ca+/calmodulin.15 There are 3 isoforms (PDE1A, PDE1B and PDE1C), all expressed in cardiac myocytes.16,17 PDE1A and PDE1B hydrolyze cGMP with higher affinity than cAMP, whereas PDE1C, the dominant isoform in human car-diac myocytes, hydrolyzes both with similar affinity.17 Although PDE1 is a major esterase for both cAMP and cGMP in the myocardium or cardiac myocytes in humans,16,18 its physio-logical effect, particularly in vivo, has not been fully clarified.

    PDE2 PDE2 is a dual substrate esterase with similar affin-ity for cAMP and cGMP.15 PDE2 is expressed in cardiac myo-cytes in various species, including humans,16 and localized at the plasma membrane and Z-line of cardiac myocytes.19 Due to the cGMP-sensitive GAF domain, the enzyme functions mainly as a cGMP-stimulated cAMP esterase and thereby a cross-talk regulator between the 2 cyclic nucleotides.20 Through a cAMP modulating function, PDE2 regulates the -adrener-gic cardiac response in a subcellular compartment-selective manner.

    PDE3 PDE3 is also a dual substrate enzyme, known as a cGMP-inhibited cAMP esterase.15 Although both subtypes, PDE3A and PDE3B, exist in cardiac myocytes, PDE3A is the main subtype and responsible for cardiac functional change caused by PDE3 inhibition.21 PDE3 has similar affinity for

    Figure. CardiacmyocytecGMP-PKGpathways.cGMPissynthesizedby2pathways,NO-sGCandNP-pGCpathways,andisundercompartmentalizedregulationbyPDEs.cGMP-PKGregulatescardiacfunction,hypertrophy/remodelingandcellsurvivalresponse(seetextfordetails).cGMP,cyclicGMP;NO,nitricoxide;NP,natriureticpeptide;PDE,phosphodiesterase;PKG,cGMP-dependentproteinkinase.LTCC,L-typeCa2+channel;TRPC,transientreceptorpotentialcanonical;GqPCR,Gqproteincoupledreceptor;AngII,angiotensinII;PE,phenylephrine;ET1,endothelin1;3AR,3adrenergicreceptor;cTnI,cardiactroponinI;PLB,phospholamban;p,phosphorylation.

    Advance Publication by-J-STAGE

  • cGMP-Dependent Signaling in Cardiac Myocyte

    cAMP and cGMP, but because of the higher catalytic rate for cAMP than for cGMP, cGMP acts as a competitive inhibitor against its cAMP catalytic action.2

    PDE5 PDE5A is a cGMP-specific esterase, abundantly ex-pressed in vascular smooth muscle. Although PDE5A was not originally thought to be expressed in the heart, evidence has accumulated that PDE5A protein is expressed in cardiac myo-cytes.1 The most recent study revealed that PDE5 represents ~20% of total cGMP-esterase activity in human cardiac myo-cytes,16 though there still remains some debate.18 PDE5 ex-pression is upregulated in the diseased heart2225 via an oxidant stress-dependent mechanism,26 and is activated by cGMP bind-ing to the GAF domain and by PKG phosphorylation at Ser 92.15 A PDE5 inhibitor such as sildenafil occupies its catalytic site as a false substrate to increase cGMP and activate PKG, leading to enhanced activity of the enzyme, and thus the inhib-itors efficacy.2,15 PDE5 is localized to the Z-band of cardiac myocytes under physiological conditions, but is diffusely dis-tributed in disease conditions.27,28

    cGMP Effector KinasecGMP-Dependent Protein Kinase I (PKGI) cGMP-depen-

    dent protein kinases (PKGs) are serine/threonine kinases that mediate many of the biological effects of cGMP. PKGI and PKGII are encoded by 2 genes,2 and PKGI is the primary isotype in the cardiovascular system. PKGI is a homodimer of identical subunits. Each subunit has 3 functional domains: (1) an N-terminus domain containing a leucine/isoleucine zipper and auto-inhibitory/pseudosubstrate region, (2) a regulatory domain with 2 allosteric cGMP biding sites, and (3) a catalytic domain. The N-terminus of PKGI is encoded by 2 alternative exons that produce 2 splice-variants, PKGI and PKGI. This N-terminal difference provides different cGMP sensitivity (PKGI is more sensitive to cGMP than PKGI by 10-fold) and protein targeting. PKGI has a unique cysteine (Cys 43), the oxidative modification of which leads to the enzyme acti-vation independently of cGMP.29 This activation mechanism operates basally to control resistance vessel tone, but its role in cardiac myocytes is unknown.30

    cGMP Signal Regulation of Cardiac MyocytescGMP Regulation of Cardiac ContractilitycGMP negatively modulates contractility, accelerates relaxation and improves the stiffness of cardiac myocytes. These effects might be mediated by direct PKG phosphorylation of proteins, including cardiac troponin I, L-type Ca2+ channel, phosphol-

    amban and titin.1 The role of PKG in the negative inotropy induced by cGMP was demonstrated in a study of mice hearts lacking PKGI. Wegener et al reported that cGMP analogs (8-Br-cGMP, 8-pCPT-cGMP) have negative inotropic effects in control hearts but not in hearts lacking PKGI,31 independent of cAMP signaling.

    Exogenous NO exerts a negative inotropic effect, which is mediated by cGMP-PKG-induced reduction in myofilament calcium responsiveness. Layland et al32 demonstrated that a NO donor induces negative inotropic effects and accelerates relaxation, without affecting the calcium transient, but with increased troponin I phosphorylation, in rat ventricular myo-cytes. These effects were abrogated by soluble GC inhibitor or PKG inhibitor. Endogenous NO has cGMP-dependent and cGMP-independent effects, depending on the source.6 Studies have revealed that NO from eNOS is coupled to the cGMP-PKG signal pathway, regulating cardiac function,28,33 whereas NO from nNOS directly modifies the ryanodine receptor (RyR) by S-nitrosylation and affects cardiac function.34 NO-cGMP derived from eNOS is functionally coupled to PDE5, modulat-ing cardiac -adrenergic stimulated contractility.28 PDE5 inhi-bition suppresses -adrenergic receptor-stimulated cardiac con-tractility in wild-type mice hearts/myocytes, an effect that is abrogated in the presence of PKG inhibitor, but is absent in hearts/myocytes lacking eNOS or 3 adrenergic receptor (3AR) coupled to eNOS.33,35 This antiadrenergic effect might be at-tributable to troponin I phosphorylation by PKG35 and/or inhi-bition of the L-type Ca2+ channel (phosphorylation by PKG).33,36 The antiadrenergic response to PDE5 inhibitor is also observed in humans.37 Importantly, normal subcellular localization of PDE5 to the Z-band is the key to this regulation. The anti-adrenergic effect of PDE5 inhibition is absent in failing canine myocytes in which PDE5 is diffusely distributed.27

    Importantly, cGMP derived from pGC (GCA) by exoge-nous ANP has no effect on cardiac systolic function or the -adrenergic contractility response, suggesting the existence of compartmentalized regulation.13,38,39 On the other hand, car-diac diastolic function is modified by the NP-pGC pathway similarly to the NO-sGC pathway. Exogenous BNP enhances the speed and extent of ventricular relaxation in normal and heart failure dogs.39 The PDE5 inhibitor, sildenafil, alone or in combination with BNP, increases left ventricular diastolic properties (distensibility) in both normal and old hypertensive dogs.40 This might be at least partially attributable to phosphory-lation of titin, a sarcomeric spring that regulates myocardial passive stiffness.40 Reduction of titin-stiffness by cGMP-PKG was also demonstrated in the human myocardium.41

    Table. PDEs in the Heart

    PDE isoenzymeKm (M) Heart

    expressioncAMP cGMP

    PDE1 Ca2+/CaM regulated, Dual specificity

    PDE1A: 73120, PDE1B: 1024, PDE1C: 0.31.2

    PDE1A: 2.65, PDE1B: 1.25.9, PDE1C: 0.62.2

    Yes

    PDE2 cGMP-stimulated, Dual specificity

    3050 1030 Yes

    PDE3 cGMP-inhibited, Dual specificity

    0.020.15 0.18 Yes

    PDE4 cAMP-specific 2.910 Yes

    PDE5 cGMP-specific 16.2 Yes

    PDE8 cAMP-specific 0.15 Yes

    PDE9 cGMP-specific 0.170.39 Yes

    PDEs, phosphodiesterases.

    Advance Publication by-J-STAGE

  • TAKIMOTO E

    cGMP affects cAMP signaling via cross-talk regulation by cGMP-regulated PDEs (PDE2 or PDE3), and modulates car-diac function. Using a real-time cyclic nucleotide imaging tech-nique, Mongillo et al reported that catecholamine stimulates cGMP via 3AR coupled to eNOS, and activates PDE2-cAMP hydrolysis, attenuating localized accumulation of cAMP.19 A more recent study has suggested that PDE2 activity is con-fined to the PKA RII isoforms residing compartment that is functionally coupled to phospholamban or troponin I phos-phorylation.42 cGMP-cAMP cross-talk involving PDE3 was implicated in pathological right hearts. In a rat model of right ventricular hypertrophy, acute PDE5 inhibition increased myocardial cAMP (as well as cGMP) and cardiac contraction, associated with reduced PDE3 activity.22 A similar positive inotropic effect by PDE5 inhibition was observed in human failing right hearts.24

    cGMP Regulation of Cardiac Hypertrophy and RemodelingThe heart develops hypertrophy in response to pathological stress.43 Although cardiac hypertrophy provides the initial func-tional compensation response to stress, it evolves into mal-adaptive remodeling/dilation with functional de-compensation, and can result in overt failure. cGMP signaling plays a brake-like role (negative regulator) in the cardiac hypertrophy re-modeling process. Gq-agonist-induced hypertrophy in cultured cardiac myocytes is suppressed by ANP, NO, cGMP or PKG.1 Although an early study using conventional PKGI null ani-mals with vascular re-introduction of PKGI failed to demon-strate a significant role for PKGI in cardiac hypertrophy or remodeling, this could be partly because their pressure-load protocol was not severe enough to induce the pathological signal activation that cGMP-PKG targets.44 The 2 most recent studies clearly demonstrated the role of PKGI as a key nega-tive regulator of cardiac hypertrophy and remodeling.45,46 Mice with reduced myocyte PKG activity by cardiac myocyte PDE5 overexpression develop exacerbated hypertrophy and remodeling in response to pressure overload, and normalizing PKG activity by turning off PDE5 overexpression ameliorates the once established remodeling.45 Mice with cardiac-specific deletion of PKGI revealed worse remodeling to angiotensin II or pressure overload.46 Studies of animals with a modified NP-cGMP pathway have shown consistent results.10,47 Mice lacking NPRA throughout the body show arterial hypertension with a disproportionate degree of cardiac hypertrophy, and mice with cardiac-specific NPRA deletion develop exagger-ated hypertrophy in response to pressure overload.10 Converse-ly, cardiac overexpression of constitutive active GC inhibits pressure-overload hypertrophy.47

    Most importantly, small molecule cGMP-PDE inhibitors ameliorate hypertrophy and remodeling. We first reported that the PDE5 inhibitor, sildenafil, activates myocardial PKG and ameliorates cardiac hypertrophy/remodeling in a mouse model of pressure overload.25 Sildenafil efficacy in volume overload remodeling was recently demonstrated in a rat model of chronic mitral valve regurgitation.48 Miller et al reported that a PDE1 inhibitor (IC86340) has antihypertrophic effects in cultured cardiac myocytes exposed to Gq-agonist and in chron-ic isoproterenol-induced hypertrophy in mice.49 PDE1 and PDE5 inhibitors show additive antihypertrophic effects, sug-gesting that these enzymes do not hydrolyze identical intracel-lular cGMP pools.49

    Several mechanisms have been suggested for the antihyper-trophic/remodeling effects of the cGMP signals. The cardiac hypertrophic response involves activation of various intracel-lular signaling cascades. Gq-coupled signaling, and in particu-

    lar, calcineurin, plays a major role in this process.50,51 Fiedler et al first demonstrated in cultured myocytes that PKG deacti-vates calcineurin via inhibition of the L-type Ca2+ channel, blunting the cellular hypertrophy response.52 Recently, studies have implicated a role for transient receptor potential canonical (TRPC) 6, a receptor-operated Ca2+ channel, in this regula-tion.53 PKG phosphorylates TRPC6 at Thr 69, 70 and Ser 322, and suppresses its conductance, resulting in inhibition of cal-cineurin-NFAT signaling. However, calcineurin inhibition is not the sole mechanism, as mice lacking calcineurin A can still develop moderate cardiac hypertrophy in response to pressure overload that is inhibited by PDE5 inhibitor.54 Stud-ies have demonstrated a central role for Regulator of G protein Signaling (RGS2 and RGS4) in the cGMP-mediated anti-hy-pertrophy/remodeling mechanism.55,56 RGS proteins de-acti-vate G protein-coupled signaling via GTPase activating prop-erties. PKGI phosphorylates (activates) RGS2 and RGS4, and terminates the Gq signal, inhibiting hypertrophy. We reported that PKG activation by PDE5 inhibitor fails to de-activate multiple hypertrophy signaling cascades in hearts lacking RGS2, resulting in a lack of the antihypertrophic/remodeling effect,55 and Tokudome et al reported that RGS4 overexpres-sion ameliorates cardiac spontaneous hypertrophy in NPRA (GCA) null animals.56 Recent study suggested that PKG-RGS2 is also involved in the ANP-GCA antihypertrophy.57

    Lastly, it is worthwhile mentioning the differential response between the right and left heart to PDE5 inhibitor in hypertro-phy/remodeling. Interestingly, PDE5 inhibitor fails to exert a direct antihypertrophic effect on right heart hypertrophy in-duced by surgically induced pressure overload.58 This differ-ence might be attributable to fundamental anatomical, bio-logical and physiological differences between the right and left sides of the heart.

    Cardiac Protection by cGMP Signaling: Ischemic Injury and CardiomyopathyThe cGMP-PKG pathway has been implicated in cardiac pro-tection against cell death-inducing stress, such as ischemic injury and doxorubicin toxicity.59,60 NO, BNP and 8Br-cGMP all ameliorate cell death in cultured cardiac myocytes exposed to ischemia-reoxygenation,61 and mice lacking PKGI develop a larger infarct than controls after ischemia-reperfusion.62 In vivo and ex vivo studies have revealed that NO donors limit infarct size and improve post-ischemic functional recovery. Such beneficial effects of NO, however, were not consistently observed, which might be attributable to impaired bioavail-ability of NO because of reactive oxygen species generated during early reperfusion.59 Inhibition of NOS during ischemic preconditioning or postconditioning abolishes its cardiac pro-tective effects, suggesting the essential role of NOS-cGMP-PKG in the cardiac protection conferred by preconditioning or post-conditioning.59 On the other hand, NPs also limit the in-farct size from ischemia-reperfusion injury in animal models and in humans. Kitakaze et al reported that patients with acute myocardial infarction who were given carperitide (recombi-nant human ANP)63 as an adjunct to reperfusion therapy had smaller infarcts, fewer reperfusion injuries and better clinical outcomes than controls.64 Recent studies have revealed potent cardioprotection by PDE5 inhibitors. The infarct-limiting ef-fect of PDE5 inhibitor in myocardial ischemia-reperfusion injury has been reported in mouse, rat and rabbit hearts.60

    The underlying mechanisms are complex, involving mito-chondrial KATP channel (mitoKATP) regulation and stress respon-sive (survival) signaling cascades.59,60 Ischemia-reperfusion induces mitochondrial permeability transition (MPT) pore

    Advance Publication by-J-STAGE

  • cGMP-Dependent Signaling in Cardiac Myocyte

    formation that induces a permeability change of mitochondria, leading to cell death. Ischemic preconditioning potently inhib-its this process, which is mediated by transient opening of mi-toKATP.65 Opening the mitoKATP partially compensates the mem-brane potential (m), which enables additional protons to be pumped out to form an H+ gradient for both ATP synthesis and Ca2+ transport.65 Costa et al reported the mechanism that links PKG to mitoKATP and MPT: PKG increases opening of mito-KATP by PKC via indirect activation.66 Rapid influx of K+ increases the matrix pH and increases H2O2 production from complex I, to further activate PKC coupled to MPT, resulting in MPT inhibition. In addition to direct PKG mitochondrial regulation, other mechanisms might be at play in cGMP-PKG mediated cardioprotection. Phosphorylation of ERK and gly-cogen synthase kinase 3 (GSK3) and upregulation of anti-apoptotic bcl2 were reported in ischemia re-perfused hearts treated with PDE5 inhibitor.67 Although ERK activation lies upstream of this regulation,68 mitochondrial effects might be directly mediated by GSK3 inactivation (phosphorylation) and upregulated bcl2, the former inhibiting MPT pore formation69 and the latter inhibiting mitochondrial apoptosis. Fiedler et al reported PKG inhibition of pro-apoptotic p38 MAPK.62 Isch-emia-reperfusion activates p38 via TAB1 (TAK1 binding pro-tein), independently of upstream kinases MKK3 or 6, inducing apoptosis. PKGI physically associates with p38 MAPK and prevents TAB1 interaction, resulting in reduced p38 activation and ischemia-reperfusion injury.62 Another protective mecha-nism involves sarcolemmal Na+/K+ ATPase regulation. Mad-hani et al reported that sildenafil or PKG phosphorylates phos-pholemman and activates Na+/K+ ATPase, to limit Na+ overload and thus Ca2+ overload during reperfusion.70

    The cardioprotective effects of cGMP signaling has been demonstrated in some types of cardiomyopathy. Doxorubicin (Dox), an anticancer drug, has cardiac toxicity that ultimately leads to cardiomyopathy. PDE5 inhibitor (sildenafil or tadala-fil) improved cardiac function and survival in a rodent model of Dox-cardiomyopathy by attenuating oxidative stress and apoptosis, which might be attributable to upregulation of bcl2 and mitochondrial superoxide dismutase (MnSOD).60 A ge-netic mouse model of Duchenne muscular dystrophy lacking dystrophin (mdx) develops cardiomyopathy as do human mus-cular dystrophy patients. Enhancing cGMP signal by cardiac overexpression of sGC or by PDE5 inhibitor, sildenafil, ame-liorates the cardiac phenotype of mdx, including cardiac con-tractile performance, metabolic status and sarcolemmal integ-rity,71 via mechanisms that might involve inhibition of MPT formation.72 PDE5 inhibitor is effective also in diabetic car-diomyopathy.60 Tadalafil improves cardiac function in leptin-deficient mice, an animal model for type II diabetes, which is associated with an altered proteomic profile of cytoskeletal rearrangement and redox regulation.

    ConclusioncGMP Signal Modification as Therapy for Human Heart DiseaseAccumulating evidence suggests that cGMP-PKG signaling plays a cardioprotective role against pathological stress. Among several strategies to enhance this signaling, PDE5 inhibition has been gaining great attention, with 3 inhibitors (sildenafil, vardenafil and tadalafil) already in clinical use to treat erectile dysfunction and pulmonary hypertension. Recent small human studies revealed beneficial cardioprotective effects from chron-ic sildenafil treatment, while it also provides acute hemody-namic benefits.73,74 Guazzi et al reported that 1-year sildenafil

    treatment improved cardiac function and exercise performance, associated with reverse remodeling of left atrial volume index and LV mass index, in heart failure patients (New York Heart Association class IIIII).75 Giannetta et al reported anti-remod-eling effects and improved cardiac kinetics from 3 months silde-nafil treatment in diabetic cardiomyopathy patients.76 These results suggest that PDE5 inhibition is a promising new ap-proach to treating various human heart diseases. A large pla-cebo-controlled multicenter trial (RELAX, NCT00763867) led by NIH is ongoing to test sildenafil efficacy in heart failure patients with preserved ejection fraction. A single center study is currently testing sildenafil efficacy in cardiomyopathy with Duchenne and Becker muscular dystrophies (REVERS-DBMD, NCT01168908). These studies will soon provide more infor-mation on the clinical benefit of PDE5 inhibitor treatment in human heart disease. On the other hand, BNP (nesiritide) was introduced into clinical practice in 2001 for early relief of dyspnea in acute heart failure patients, because of its vasodila-tory properties. However, recent evaluation in a large random-ized trial showed minimal benefit and an increased risk of hypotension.77 The efficacy of combined use of tadalafil and nesiritide is being tested in subclinical heart failure patients, in whom the expected benefit is renal function improvement rather than a direct cardiac effect (NCT01544998). Consider-ing that the enzymes for cGMP synthesis, including NOS, sCG and GC-A, could be uncoupled from cGMP synthesis under chronic stress conditions, stimulation of sGC with a redox-insensitive activator and/or re-coupling of NOS with tetrahy-drobiopterin (BH4) might have intriguing potential as a strat-egy to enhance cGMP signaling other than inhibiting cGMP degradation.

    In summary, the cardiac cGMP system plays a key role in protection against various cardiac pathologies, including hy-pertrophy, heart failure, ischemic injury and cardiomyopathy. PDE5 inhibitors, in particular, are a promising therapeutic modality to enhance this signal, and efficacy has been demon-strated in human heart failure and diabetic cardiomyopathy. Ongoing and future large-scale clinical trials will determine whether the therapy will have long-term beneficial effects on cardiac remodeling and a reduction in major cardiac events. NP (nesiritide) failed to show benefit in acute decompensated heart failure, but its cardioprotective effect, particularly against ischemic injury, warrants further investigation.78

    AcknowledgmentsThis work was supported by National Institute of Health (RO1 093432) and American Heart Association (11GRNT7700071).

    References 1. Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular patho-

    physiology and therapeutics. Pharmacol Ther 2009; 122: 216 238. 2. Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent pro-

    tein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev 2010; 62: 525 563.

    3. Fischmeister R, Castro LR, bi-Gerges A, Rochais F, Jurevicius J, Leroy J, et al. Compartmentation of cyclic nucleotide signaling in the heart: The role of cyclic nucleotide phosphodiesterases. Circ Res 2006; 99: 816 828.

    4. Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in car-diovascular signaling. Circ Res 2010; 106: 633 646.

    5. Nakahira A, Minamiyama Y, Takemura S, Hirai H, Sasaki Y, Okada S, et al. Coadministration of carvedilol attenuates nitrate tolerance by preventing cytochrome p450 depletion. Circ J 2010; 74: 1711 1717.

    6. Ziolo MT, Kohr MJ, Wang H. Nitric oxide signaling and the regula-tion of myocardial function. J Mol Cell Cardiol 2008; 45: 625 632.

    7. Friebe A, Koesling D. The function of NO-sensitive guanylyl cy-

    Advance Publication by-J-STAGE

  • TAKIMOTO E

    clase: What we can learn from genetic mouse models. Nitric Oxide 2009; 21: 149 156.

    8. Heo KS, Fujiwara K, Abe J. Disturbed-flow-mediated vascular reac-tive oxygen species induce endothelial dysfunction. Circ J 2011; 75: 2722 2730.

    9. Takimoto E, Kass DA. Role of oxidative stress in cardiac hypertro-phy and remodeling. Hypertension 2007; 49: 241 248.

    10. Kishimoto I, Tokudome T, Nakao K, Kangawa K. Natriuretic pep-tide system: An overview of studies using genetically engineered animal models. FEBS J 2011; 278: 1830 1841.

    11. Kuhn M. Structure, regulation, and function of mammalian mem-brane guanylyl cyclase receptors, with a focus on guanylyl cyclase-A. Circ Res 2003; 93: 700 709.

    12. Kalra PR, Clague JR, Bolger AP, Anker SD, Poole-Wilson PA, Struthers AD, et al. Myocardial production of C-type natriuretic pep-tide in chronic heart failure. Circulation 2003; 107: 571 573.

    13. Wollert KC, Yurukova S, Kilic A, Begrow F, Fiedler B, Gambaryan S, et al. Increased effects of C-type natriuretic peptide on contractil-ity and calcium regulation in murine hearts overexpressing cyclic GMP-dependent protein kinase I. Br J Pharmacol 2003; 140: 1227 1236.

    14. Schlossmann J, Ammendola A, Ashman K, Zong X, Huber A, Neubauer G, et al. Regulation of intracellular calcium by a signalling complex of IRAG, IP3 receptor and cGMP kinase Ibeta. Nature 2000; 404: 197 201.

    15. Francis SH, Blount MA, Corbin JD. Mammalian cyclic nucleotide phosphodiesterases: Molecular mechanisms and physiological func-tions. Physiol Rev 2011; 91: 651 690.

    16. Johnson WB, Katugampola S, Able S, Napier C, Harding SE. Profil-ing of cAMP and cGMP phosphodiesterases in isolated ventricular cardiomyocytes from human hearts: Comparison with rat and guinea pig. Life Sci 2012; 90: 328 336.

    17. Vandeput F, Wolda SL, Krall J, Hambleton R, Uher L, McCaw KN, et al. Cyclic nucleotide phosphodiesterase PDE1C1 in human car-diac myocytes. J Biol Chem 2007; 282: 32749 32757.

    18. Vandeput F, Krall J, Ockaili R, Salloum FN, Florio V, Corbin JD, et al. cGMP-hydrolytic activity and its inhibition by sildenafil in normal and failing human and mouse myocardium. J Pharmacol Exp Ther 2009; 330: 884 891.

    19. Mongillo M, Tocchetti CG, Terrin A, Lissandron V, Cheung YF, Dostmann WR, et al. Compartmentalized phosphodiesterase-2 activ-ity blunts beta-adrenergic cardiac inotropy via an NO/cGMP-depen-dent pathway. Circ Res 2006; 98: 226 234.

    20. Zaccolo M, Movsesian MA. cAMP and cGMP signaling cross-talk: Role of phosphodiesterases and implications for cardiac pathophys-iology. Circ Res 2007; 100: 1569 1578.

    21. Sun B, Li H, Shakur Y, Hensley J, Hockman S, Kambayashi J, et al. Role of phosphodiesterase type 3A and 3B in regulating platelet and cardiac function using subtype-selective knockout mice. Cell Signal 2007; 19: 1765 1771.

    22. Nagendran J, Archer SL, Soliman D, Gurtu V, Moudgil R, Haromy A, et al. Phosphodiesterase type 5 is highly expressed in the hypertro-phied human right ventricle, and acute inhibition of phosphodiester-ase type 5 improves contractility. Circulation 2007; 116: 238 248.

    23. Pokreisz P, Vandenwijngaert S, Bito V, Van den BA, Lenaerts I, Busch C, et al. Ventricular phosphodiesterase-5 expression is increased in patients with advanced heart failure and contributes to adverse ventricular remodeling after myocardial infarction in mice. Circula-tion 2009; 119: 408 416.

    24. Shan X, Quaile MP, Monk JK, French B, Cappola TP, Margulies KB. Differential expression of PDE5 in failing and nonfailing human myocardium. Circ Heart Fail 2012; 5: 79 86.

    25. 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: 214 222.

    26. Lu Z, Xu X, Hu X, Lee S, Traverse JH, Zhu G, et al. Oxidative stress regulates left ventricular PDE5 expression in the failing heart. Cir-culation 2010; 121: 1474 1483.

    27. Senzaki H, Smith CJ, Juang GJ, Isoda T, Mayer SP, Ohler A, et al. Cardiac phosphodiesterase 5 (cGMP-specific) modulates beta-adren-ergic signaling in vivo and is down-regulated in heart failure. FASEB J 2001; 15: 1718 1726.

    28. Takimoto E, Champion HC, Belardi D, Moslehi J, Mongillo M, Mergia E, et al. cGMP catabolism by phosphodiesterase 5A regulates car-diac adrenergic stimulation by NOS3-dependent mechanism. Circ Res 2005; 96: 100 109.

    29. Burgoyne JR, Madhani M, Cuello F, Charles RL, Brennan JP, Schroder E, et al. Cysteine redox sensor in PKGIa enables oxidant-induced activation. Science 2007; 317: 1393 1397.

    30. Prysyazhna O, Rudyk O, Eaton P. Single atom substitution in mouse

    protein kinase G eliminates oxidant sensing to cause hypertension. Nat Med 2012; 18: 286 290.

    31. Wegener JW, Nawrath H, Wolfsgruber W, Kuhbandner S, Werner C, Hofmann F, et al. cGMP-dependent protein kinase I mediates the negative inotropic effect of cGMP in the murine myocardium. Circ Res 2002; 90: 18 20.

    32. Layland J, Li JM, Shah AM. Role of cyclic GMP-dependent protein kinase in the contractile response to exogenous nitric oxide in rat cardiac myocytes. J Physiol 2002; 540: 457 467.

    33. Wang H, Kohr MJ, Traynham CJ, Ziolo MT. Phosphodiesterase 5 restricts NOS3/Soluble guanylate cyclase signaling to L-type Ca2+ current in cardiac myocytes. J MolCell Cardiol 2009; 47: 304 314.

    34. Wang H, Viatchenko-Karpinski S, Sun J, Gyorke I, Benkusky NA, Kohr MJ, et al. Regulation of myocyte contraction via neuronal nitric oxide synthase: Role of ryanodine receptor S-nitrosylation. J Physi-ol 2010; 588: 2905 2917.

    35. Lee DI, Vahebi S, Tocchetti CG, Barouch LA, Solaro RJ, Takimoto E, et al. PDE5A suppression of acute beta-adrenergic activation requires modulation of myocyte beta-3 signaling coupled to PKG-mediated troponin I phosphorylation. Basic Res Cardiol 2010; 105: 337 347.

    36. Yang L, Liu G, Zakharov SI, Bellinger AM, Mongillo M, Marx SO. Protein kinase G phosphorylates Cav1.2 alpha1c and beta2 subunits. Circ Res 2007; 101: 465 474.

    37. Borlaug BA, Melenovsky V, Marhin T, Fitzgerald P, Kass DA. Silde-nafil inhibits beta-adrenergic-stimulated cardiac contractility in hu-mans. Circulation 2005; 112: 2642 2649.

    38. Takimoto E, Belardi D, Tocchetti CG, Vahebi S, Cormaci G, Ketner EA, et al. Compartmentalization of cardiac beta-adrenergic inotropy modulation by phosphodiesterase type 5. Circulation 2007; 115: 2159 2167.

    39. Lainchbury JG, Burnett JC Jr, Meyer D, Redfield MM. Effects of natriuretic peptides on load and myocardial function in normal and heart failure dogs. Am J Physiol Heart Circ Physiol 2000; 278: H33 H40.

    40. Bishu K, Hamdani N, Mohammed SF, Kruger M, Ohtani T, Ogut O, et al. Sildenafil and B-type natriuretic peptide acutely phosphorylate titin and improve diastolic distensibility in vivo. Circulation 2011; 124: 2882 2891.

    41. Kruger M, Kotter S, Grutzner A, Lang P, Andresen C, Redfield MM, et al. Protein kinase G modulates human myocardial passive stiffness by phosphorylation of the titin springs. Circ Res 2009; 104: 87 94.

    42. Stangherlin A, Gesellchen F, Zoccarato A, Terrin A, Fields LA, Berrera M, et al. cGMP signals modulate cAMP levels in a compart-ment-specific manner to regulate catecholamine-dependent signaling in cardiac myocytes. Circ Res 2011; 108: 929 939.

    43. Frey N, Olson EN. Cardiac hypertrophy: The good, the bad, and the ugly. Annu Rev Physiol 2003; 65: 45 79.

    44. Kass DA, Takimoto E. Regulation and role of myocyte cyclic GMP-dependent protein kinase-1. Proc Natl Acad Sci USA 2010; 107: E98; author reply E99.

    45. Zhang M, Takimoto E, Hsu S, Lee DI, Nagayama T, Danner T, et al. Myocardial remodeling is controlled by myocyte-targeted gene reg-ulation of phosphodiesterase type 5. J Am Coll Cardiol 2010; 56: 2021 2030.

    46. Frantz S, Klaiber M, Baba HA, Oberwinkler H, Volker K, Gabetaner B, et al. Stress-dependent dilated cardiomyopathy in mice with car-diomyocyte-restricted inactivation of cyclic GMP-dependent protein kinase I. Eur Heart J 2011 December 23 [Epub ahead of print].

    47. Zahabi A, Picard S, Fortin N, Reudelhuber TL, Deschepper CF. Ex-pression of constitutively active guanylate cyclase in cardiomyocytes inhibits the hypertrophic effects of isoproterenol and aortic constric-tion on mouse hearts. J Biol Chem 2003; 278: 47694 47699.

    48. Kim KH, Kim YJ, Ohn JH, Yang J, Lee SE, Lee SW, et al. Long-term effects of sildenafil in a rat model of chronic mitral regurgita-tion: Benefits of ventricular remodeling and exercise capacity. Cir-culation 2012; 125: 1390 1401.

    49. Miller CL, Oikawa M, Cai Y, Wojtovich AP, Nagel DJ, Xu X, et al. Role of Ca2+/calmodulin-stimulated cyclic nucleotide phosphodies-terase 1 in mediating cardiomyocyte hypertrophy. Circ Res 2009; 105: 956 964.

    50. Dorn GW 2nd, Force T. Protein kinase cascades in the regulation of cardiac hypertrophy. J Clin Invest 2005; 115: 527 537.

    51. Fiedler B, Wollert KC. Interference of antihypertrophic molecules and signaling pathways with the Ca2+-calcineurin-NFAT cascade in cardiac myocytes. Cardiovasc Res 2004; 63: 450 457.

    52. Fiedler B, Lohmann SM, Smolenski A, Linnemuller S, Pieske B, Schroder F, et al. Inhibition of calcineurin-NFAT hypertrophy sig-naling by cGMP-dependent protein kinase type I in cardiac myo-cytes. Proc Natl Acad Sci USA 2002; 99: 11363 11368.

    53. Koitabashi N, Aiba T, Hesketh GG, Rowell J, Zhang M, Takimoto E,

    Advance Publication by-J-STAGE

  • cGMP-Dependent Signaling in Cardiac Myocyte

    et al. Cyclic GMP/PKG-dependent inhibition of TRPC6 channel ac-tivity and expression negatively regulates cardiomyocyte NFAT ac-tivation Novel mechanism of cardiac stress modulation by PDE5 in-hibition. J Mol Cell Cardiol 2010; 48: 713 724.

    54. Hsu S, Nagayama T, Koitabashi N, Zhang M, Zhou L, Bedja D, et al. Phosphodiesterase 5 inhibition blocks pressure overload-induced car-diac hypertrophy independent of the calcineurin pathway. Cardio-vasc Res 2009; 81: 301 309.

    55. Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, et al. Regulator of G protein signaling 2 mediates cardiac compen-sation to pressure overload and antihypertrophic effects of PDE5 in-hibition in mice. J Clin Invest 2009; 119: 408 420.

    56. Tokudome T, Kishimoto I, Horio T, Arai Y, Schwenke DO, Hino J, et al. Regulator of G-protein signaling subtype 4 mediates antihyper-trophic effect of locally secreted natriuretic peptides in the heart. Circulation 2008; 117: 2329 2339.

    57. Klaiber M, Kruse M, Volker K, Schroter J, Feil R, Freichel M, et al. Novel insights into the mechanisms mediating the local antihyper-trophic effects of cardiac atrial natriuretic peptide: Role of cGMP-dependent protein kinase and RGS2. Basic Res Cardiol 2010; 105: 583 595.

    58. Schafer S, Ellinghaus P, Janssen W, Kramer F, Lustig K, Milting H, et al. Chronic inhibition of phosphodiesterase 5 does not prevent pres-sure-overload-induced right-ventricular remodelling. Cardiovasc Res 2009; 82: 30 39.

    59. Burley DS, Ferdinandy P, Baxter GF. Cyclic GMP and protein ki-nase-G in myocardial ischaemia-reperfusion: Opportunities and ob-stacles for survival signaling. Br J Pharmacol 2007; 152: 855 869.

    60. Kukreja RC, Salloum FN, Das A, Koka S, Ockaili RA, Xi L. Emerg-ing new uses of phosphodiesterase-5 inhibitors in cardiovascular dis-eases. Exp Clin Cardiol 2011; 16: e30 e35.

    61. Gorbe A, Giricz Z, Szunyog A, Csont T, Burley DS, Baxter GF, et al. Role of cGMP-PKG signaling in the protection of neonatal rat car-diac myocytes subjected to simulated ischemia/reoxygenation. Basic Res Cardiol 2010; 105: 643 650.

    62. Fiedler B, Feil R, Hofmann F, Willenbockel C, Drexler H, Smolenski A, et al. cGMP-dependent protein kinase type I inhibits TAB1-p38 mitogen-activated protein kinase apoptosis signaling in cardiac myo-cytes. J Biol Chem 2006; 281: 32831 32840.

    63. Takahashi R, Uchiyama A, Iguchi N, Mashimo T, Fujino Y. Effects of continuous venovenous hemofiltration on the pharmacology of carperitide, a recombinant human atrial natriuretic peptide. Circ J 2010; 74: 1888 1894.

    64. Kitakaze M, Asakura M, Kim J, Shintani Y, Asanuma H, Hamasaki T, et al. Human atrial natriuretic peptide and nicorandil as adjuncts to reperfusion treatment for acute myocardial infarction (J-WIND): Two randomised trials. Lancet 2007; 370: 1483 1493.

    65. Ardehali H, ORourke B. Mitochondrial K(ATP) channels in cell survival and death. J Mol Cell Cardiol 2005; 39: 7 16.

    66. Costa AD, Garlid KD, West IC, Lincoln TM, Downey JM, Cohen MV, et al. Protein kinase G transmits the cardioprotective signal from cy-

    tosol to mitochondria. Circ Res 2005; 97: 329 336.67. Das A, Xi L, Kukreja RC. Protein kinase G-dependent cardioprotec-

    tive mechanism of phosphodiesterase-5 inhibition involves phos-phorylation of ERK and GSK3beta. J Biol Chem 2008; 283: 29572 29585.

    68. Das A, Salloum FN, Xi L, Rao YJ, Kukreja RC. ERK phosphoryla-tion mediates sildenafil-induced myocardial protection against isch-emia-reperfusion injury in mice. Am.J Physiol Heart Circ Physiol 2009; 296: H1236 H1243.

    69. Sanada S, Komuro I, Kitakaze M. Pathophysiology of myocardial reperfusion injury: Preconditioning, postconditioning, and transla-tional aspects of protective measures. Am J Physiol Heart Circ Physi-ol 2011; 301: H1723 H1741.

    70. Madhani M, Hall AR, Cuello F, Charles RL, Burgoyne JR, Fuller W, et al. Phospholemman Ser69 phosphorylation contributes to sildena-fil-induced cardioprotection against reperfusion injury. Am J Physiol Heart Circ Physiol 2010; 299: H827 H836.

    71. Khairallah M, Khairallah RJ, Young ME, Allen BG, Gillis MA, Danialou G, et al. Sildenafil and cardiomyocyte-specific cGMP sig-naling prevent cardiomyopathic changes associated with dystrophin deficiency. Proc Natl Acad Sci USA 2008; 105: 7028 7033.

    72. Ascah A, Khairallah M, Daussin F, Bourcier-Lucas C, Godin R, Allen BG, et al. Stress-induced opening of the permeability transition pore in the dystrophin-deficient heart is attenuated by acute treatment with sildenafil. Am J Physiol Heart Circ Physiol 2011; 300: H144 H153.

    73. Lewis GD, Lachmann J, Camuso J, Lepore JJ, Shin J, Martinovic ME, et al. Sildenafil improves exercise hemodynamics and oxygen uptake in patients with systolic heart failure. Circulation 2007; 115: 59 66.

    74. Lindman BR, Zajarias A, Madrazo JA, Shah J, Gage BF, Novak E, et al. Effects of phosphodiesterase type 5 inhibition on systemic and pulmonary hemodynamics and ventricular function in patients with severe symptomatic aortic stenosis. Circulation 2012; 125: 2353 2362.

    75. Guazzi M, Vicenzi M, Arena R, Guazzi MD. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geom-etry, and clinical status in patients with stable systolic heart failure: Results of a 1-year, prospective, randomized, placebo-controlled study. Circ Heart Fail 2011; 4: 8 17.

    76. Giannetta E, Isidori AM, Galea N, Carbone I, Mandosi E, Vizza CD, et al. Chronic inhibition of cyclic GMP phosphodiesterase 5a im-proves diabetic cardiomyopathy: A randomized, controlled clinical trial using magnetic resonance imaging with myocardial tagging. Circulation 2012; 125: 2323 2333.

    77. OConnor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V, et al. Effect of nesiritide in patients with acute de-compensated heart failure. N Engl J Med 2011; 365: 32 43.

    78. Sezai A, Wakui S, Akiyama K, Hata M, Yoshitake I, Unosawa S, et al. Myocardial protective effect of human atrial natriuretic peptide in cardiac surgery: hANP shot in clinical safety trial. Circ J 2011; 75: 2144 2150.

    Advance Publication by-J-STAGE