Molecular Pathways for Cardiac Hypertrophy and Heart Failure Progression

Molecular Pathways for Cardiac Hypertrophy and Heart Failure Progression

S E C T I O N 3 Cardiac Muscle Diseases chapter1 5 Molecular Pathways for Cardiac Hypertrophy and Heart Failure Progression Masahiko Hoshijima Susu...

2MB Sizes 0 Downloads 77 Views

S E C T I O N 3

Cardiac Muscle Diseases

chapter1 5

Molecular Pathways for Cardiac Hypertrophy and Heart Failure Progression Masahiko Hoshijima Susumu Minamisawa Hideo Yasukawa Kenneth R. Chien

Heart failure is emerging as a leading cause of human morbidity and mortality and is predicted to reach epidemic proportions in the early 21st century.1 Unfortunately, current therapy for heart failure is largely based on a complex regimen of drugs that are primarily targeted at symptoms of the disease rather than the fundamental processes that drive disease progression.2–4 One of the major difficulties has been the complexity of the disease, which is chronic, progressive, and propelled by a combination of environmental stimuli and genetic susceptibility.5,6 Studies of rare familial forms of heart failure have documented a trend toward incomplete penetrance with a highly variable genotype-phenotype correlation, suggesting the presence of strong modifiers and associated genetic background effects.7,8 At the same time, diverse environmental stimuli, including postinfarction injury, cardiotoxic side effects of chemotherapeutic agents, viral infection, and chronic exposure to volume and pressure overload can all serve to initiate and modify disease progression. Although decreases in cardiac contractility and relaxation are conserved features of many forms of heart failure, these acute physiologic end points do not always correlate with long-term mortality, again underscoring the complex nature of the heart failure phenotype. Heart failure is a growing, unmet clinical problem, representing both a major scientific opportunity and a clinical challenge for cardiovascular physicians and scientists alike. Many laboratories now view heart failure as the “cancer” of the cardiovascular system, because it is a classic, multifactorial, polygenic, and chronic disease that progresses to a terminal stage. Intriguingly, a growing body of work now suggests another important parallel between the biology of cancer and the biology of heart failure. The massive, abnormal growth in cardiac muscle that accompanies heart failure shares several biologic principles and molecular pathways with cancer biology. The biologic precepts of cell growth, apoptosis, and cell survival are as important in the onset of heart failure as they are in tumor progression, and the molecular signals for cell proliferation

and cardiac myocyte hypertrophy are highly conserved. Both diseases are inexorable and progressive in their nature, and a “multihit” hypothesis applies for both cancer and heart failure progression, largely based on the interplay between genetic susceptibility and environmental stimuli. This view, coupled with the integration of multiple experimental approaches including in vivo somatic gene transfer, bioinformatics, and computational biology, is beginning to provide new insights into the disease process. Insights from genetically based studies in mice and humans are now suggesting new therapeutic targets and strategies for intervening in the disease. With the completion of the human and mouse genome databases and physical mapping of annotated genes that provide a road map, great strides in unraveling the logic of heart failure progression can be made. This chapter highlights several advances in the understanding of the molecular pathways that drive the onset of cardiac hypertrophy and heart failure; these advances are forming the basis for a new wave of biologically targeted therapy.

BIPHASIC TEMPORAL SIGNALING CASCADES DIRECTLY ACTIVATED BY MECHANICAL STRESS: POSITIVE AND NEGATIVE REGULATORS Mechanical stress is one of the most important stimuli for triggering the initial steps toward heart failure. A diverse group of biochemical signals can be rapidly activated during in vivo pressure overload; many can be activated within a few minutes of exposure to dynamic or passive stretch that accompanies increases in aortic pressure or volume overload.9–12 To date, the precise identity of the molecular machinery that initially senses cardiac mechanical stress has remained largely unknown. A series of studies have indicated that a subset of ion-channel or ion exchangers, such as a stretch-activated channel or a sodium-proton exchanger, are regulated by deformation of 273

274

CARDIAC MUSCLE DISEASES

the plasma membrane and are found in cardiomyocytes exposed to mechanical or osmotic load.13,14 These mechanical stress-related changes in ionic current have rarely been linked to the onset of hypertrophy in individual cardiomyocytes.15 However, several transmembrane signaling pathways, including MAP kinases, Janus kinase (JAK)/signal transducers and activators of transcription (STAT), and phosphoinositide 3-kinase (PI3K)/Akt (protein kinase B) cascades, have been suggested to connect mechanical load to the onset of cardiac hypertrophy.16–19 Following mechanical stretch of cultured cardiomyocytes, most of these cardiac signaling kinase cascades are activated, in part via the cell adhesion complex,which include integrins and integrin-associated proteins that are linked to the extracellular matrix (ECM).20,21 Cardiac conditional knockout mice of the β1 integrin gene display a blunted MAP kinase activation following in vivo pressure overload.22 In addition, ablation of melusin, an integrin-binding protein, has resulted in blunted response to pressure-overload stimulation to activate Akt/glycogen synthase kinase-3β (GSK3β) signaling and to induce cardiac hypertrophy.23 However, further investigation will be necessary to determine the precise role of the ECM-integrin-cytoskeleton complex in cardiac mechanical stress sensing. In addition, hormonal stimuli, such as adrenergic agonists, angiotensin, endothelin, and gp130 cytokine receptor agonists also acti-

vate these signaling kinases, thereby triggering hypertrophic responses in vitro and in vivo.16–19 Many of these insights in the signaling pathways for cardiac hypertrophy have originated from studies in cultured neonatal rat cardiomyocytes in vitro, followed by studies of normal and genetically engineered mouse models that have been exposed to the pressure overload that accompanies in vivo aortic banding. The acute response to pressure-overload peaks within hours and includes the activation of ERK1/2, JNK, p38, Akt, and gp130/STAT and the induction of early stress-response genes, such as the suppressor of cytokine signaling-3 (SOCS3) (Fig. 15-1,Table 15-1). This early phase response is followed by a hypertrophic response, which starts within 48 hours after aortic banding and reaches a peak within 7 days.This later hypertrophic response is clearly marked by an increase in the left ventricle to body weight ratio and the induction of a series of embryonic gene markers such as ANF, skeletal actin, and β-myosin heavy chain.9,31,32 Interestingly, there is a second peak of activation for many of signaling cascade kinases in this subacute hypertrophic response phase,10,11 suggesting that there may be phasic and discrete roles for a subset of signaling pathways that respond to mechanical stress. The double peak of early mechanical stress responses implicates the presence of inhibitory regulators that act

FIGURE 15-1. Mechanical stress-induced transmembrane signaling and integrated phenotypes of a failing heart. The activation profiles of various signaling cascades and time-dependent changes in phenotypic features of heart failure are illustrated based on our previous studies10 and unpublished observations. Lower panels show biphasic induction of representative stress-responding marker genes and upregulation of left ventricular to body weight (LV/BW) ratio after transverse thoracic aortic constriction in normal adult mice. (Modified from Yasukawa H, Hoshijima M, Gu Y, et al: Suppressor of cytokine signaling-e is a biomechanical stress-inducible gene that suppresses gp130-mediated cardiac myocyte hypertrophy and survival pathways. J Clin Invest 2001;108:1459–1467 and Hoshijima M, Chien KR: Mixed signals in heart failure: Cancer rules. J Clin Invest 2002;109:849–855.)

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION

275

TABLE 15-1 MECHANICAL STRESS-RELATED CHANGES IN GENE EXPRESSION DURING HEART FAILURE PROGRESSION IN HUMAN AND EXPERIMENTAL STUDIES Acute (<24 hr) Up c-fos c-jun junB egr-1 nur77 BNP SOCS3

Hypertrophic–Failing (Days-Weeks-Years) Up Secreted proteins ANF, lipocortin I, ET-1 HB-EGF,TGFβ1, BNP Osteoblast specific factor 2 Cytoskeletal proteins αMHC, βMHC MLC1a/v, MLC2a MLC2v, tropomyosin Troponin C, Myomesin Smooth muscle α–actin Skeletal α–actin α–cardiac actin FHL1(HCM), sarcosin Desmin, gelsolin Extracellular matrix Fibulin, fibronectin Laminin, collagen Ion-channels/carriers Na+/Ca2+ exchanger, Kv1.4 Voltage dependent anion channel-1

Down Metabolism/translational proteins Ubiquitin, pyruvate dehydrogenase α NADH ubiquinone oxidoreductase Creatine kinase, Myoglobin Superoxide dismutase 2 Aldose reductase, EF-1a, EF-2, IF-4AII 28S, 60S ribosomal L3 Signaling Gsα, βARK, adenylyl cyclase VII A-kinase, C-kinase inhibitor-1, ILK Rap1B, SOCS3, Id-1, GATA-4 SP1/3, PGD/D2 synthase Others Heat shock 70kD proteins 1, 6, 8 Quaking protein, CARP

Cytoskeletal proteins FHL1 (failing heart) Nonsarcomeric MLC2 Ion-channels/carriers L-type Ca2+ channel SERCA2 Phospholamban Kv4.2, 4.3 Kv1.5 KChIP2 Signaling β1-AR, type-A like Ephrin receptor Others α1-Antichymotrypsin αB-Crystallin Plasminogen activator inhibitor-1 TIM17 Connexin43

Modified from Hoshijima M, Chien KR: Mixed signals in heart failure: Cancer rules. J Clin Invest 2002;109:849–855, and references 24 to 30.

as a negative feedback loop to prevent the continuous activation of any single pathway. Naturally, there must be negative regulators of the response that leads to attenuation of the signal so that the acute response is properly terminated and an orderly compensatory hypertrophic response ensues, as opposed to unbridled reactive growth, which is often seen in mouse models of cardiac hypertrophy induced by cardiac overexpression of signaling molecules.19 A number of studies have recently identified negative feedback regulation of these intracellular signaling pathways. SOCS3 provides an example of a biomechanical stress-inducible negative regulator, which acts to inhibit the gp130 stress-inducible pathway for myocyte survival.10,33 Within minutes of aortic constriction, either by the action of gp130 ligands (e.g., cardiotrophin-1 and LIF) or via unknown mechanisms of direct activation, JAKs phosphorylate a transcriptional regulator STAT3 leading to the translocation of STAT3 into the nucleus, which directly activates the expression of genes that can confer myocyte hypertrophy34,35 and survival (e.g., BCL-x),36–38 and SOCS3.10 SOCS3 is induced in the myocardium within 1 hour of aortic banding, peaking for several hours and suppressing JAK kinase activity by selectively binding to gp130. By inhibiting JAKs, SOCS3 negatively regulates stress-inducible gp130 activation (Fig. 15-2). It is likely that this negative regulatory circuit prevents hyperstimulation of this pathway that is induced by sustained expression of gp130-coupled cytokines, which may have independent pathologic effects on cardiac function. In this manner, the delicate balance between the activation of gp130/JAK signaling and the induction of its negative feedback regulator SOCS3 might be important in the transition between car-

diac hypertrophy and failure.5,10 Disrupting the balance between this positive and negative regulatory loop by chronic exposure to gp130 ligands might lead to secondary effects that impair cardiac function. A number of other novel negative regulators of cardiac signaling have recently been described. Recent studies have identified myocyte-enriched calcineurin-interacting protein-1 (MCIP1), a protein that inhibits cardiac hypertrophy by attenuating calcineurin activity.39 The MCIP1 gene contains NFAT (nuclear factors of activated T cells) binding motifs that mediate its upregulation by calcineurin signals, thereby suggesting that MCIP1 participates in the negative feedback loop of calcineurin signaling in the myocardium.40 A separate negative feedback regulatory system has been found for cardiac MAP kinase signaling: forced expression of MAP kinase phosphatase-1 (MKP-1) has been shown to negatively regulate the cardiac hypertrophic response by downregulating the three branches of MAP kinase cascade (p38s, JNK1/2, and ERK1/2).41 Given this complex temporal pattern of signaling pathways that are activated following in vivo pressure overload, it is not surprising that studies of the effects of the overexpression of diverse components of cardiac signaling pathways have been shown to result in cardiac hypertrophy or failure. It will become increasingly important to examine the temporal sequence of activation of these signaling pathways and to connect them to specific phenotypic features that are associated with discrete physiologic end points associated with the disease, such as defects in contractility, action potential, arrhythmogenesis, and so forth. A more meticulous gene switching in model animals42 and high-efficiency somatic gene delivery systems have become available toward this end.43,44

276

CARDIAC MUSCLE DISEASES

FIGURE 15-2. PI3K/Akt signaling in cardiomyocytes. Activation of variety of downstream molecules leads to hypertrophic and survival effects in cardiomyocytes.

HYPERTROPHY RESPONSE: QUANTITATIVE EFFECTS OF INCREASING TRANSLATIONAL EFFICIENCY AND QUALITATIVE EFFECTS ON INDUCING AN EMBRYONIC GENE PROGRAM PI3K/Akt Pathway Cardiac hypertrophy can be broken down into two central phenotypic end points: (1) quantitative effects on increasing the level of sarcomeric and other constitutive proteins and their transcripts and (2) qualitative changes in the cardiac gene program that lead to a return to the fetal phenotype. Interestingly, both of these phenotypic effects can be found during growth stimulation of diverse differentiated cell types. In this regard, insulin-like growth factor-1 (IGF-1) is a potent stimulus for both cardiac45,46 and skeletal muscle hypertrophy47 in vivo. Recent studies have clearly identified that IGF-1 increases translational efficiency of preexisting mRNAs, which underlies the hypertrophic response of skeletal muscle hypertrophy.48–50 A number of independent approaches have convincingly shown a central role of Akt in enhancing translational efficiency by phosphorylating and thereby inhibiting GSK3.48,49,51 Skeletal muscle atrophy leads to a decrease in the Akt/mTOR (mammalian target of rapamycin) pathway, and IGF-1 unexpectedly acts via Akt to antagonize calcineurin signaling during skeletal myotube hypertrophy. Taken together, the activation of the Akt/mTOR pathway and its downstream targets, p70S6K and PHAS-1/4E-BP1,3,52 appear to play a central role in skeletal muscle and perhaps cardiac muscle hypertrophy and atrophy (Fig. 15-2).53 IGF-1 activates PI3K,54,55 thereby activating Akt and phosphoinositide-dependent

kinase 1 (PDK-1), which interact with each other and cooperatively activate downstream signaling.52,56,57 The tumor suppressor phosphatase and tensin homolog on chromosome 10 (PTEN) antagonizes this pathway by facilitating dephosphorylation of PtdIns(3,4,5)P3 (phosphatidylinositol-3,4,5-triphosphate),58,59 During in vivo pressure overload, an increase in translational efficiency has also been documented, and myocardial Akt is particularly activated during the subacute hypertrophic response phase.10 Taken together, a similar role for Akt signaling in heart failure-related myocardial remodeling is likely, and activation of Akt is noted in the failing human heart.60 Transgenic mouse models of cardiac IGF-1,61 PI3K,62 and Akt overexpression63–65 have been reported to induce cardiac hypertrophy and/or hyperplasia, whereas cardiac overexpression of GSK3β suppresses pressure overload or β-adrenergic stimulation-dependent hypertrophy.53,66 In addition, in vitro overexpression of catalytically inactive mutant of PTEN induces cardiomyocyte hypertrophy,67 and muscle-specific ablation of PTEN enhances in vivo mouse cardiac hypertrophy.68 Taken together, PI3K/Akt and their downstream signaling appear to be an important in vivo determinant of cardiac size. Recently, further complexity of P13K signaling was documented. PI3Kα is linked to receptor tyrosine kinase pathways that regulate cardiac myocyte size, and PI3Kγ, which binds to G-protein βγ subunits,69 negatively regulates cardiomyocyte contractility through β2-adrenergic receptor/cAMP-dependent signaling.68

Calcium Signaling Numerous studies in cardiac muscle now support an important role for calcium signaling in the activation of a hypertrophic response. Earlier, activation of calmodulin

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION

(CaM)-dependent kinase (CaMK) was reported to be involved in the regulation of a fetal gene program that is coactivated during cardiac hypertrophy.70 CaMK activity is elevated in failing human hearts.71 Subsequently, a series of studies using transgenic mouse models documented that cardiac overexpression of calcineurin, a calcium-CaM dependent protein phosphatase, can induce massive cardiac hypertrophy.72 A family of transcriptional factors NFATs are located in the downstream of calcineurin signaling.73 In addition, the cardiac overexpression of CaMKIV, a neural CaMK isotype, results in a concentric form of cardiac hypertrophy.74 Myocyte enhancer factor-2 (MEF2), a MADS-box transcription factor highly expressed in myocytes, is a putative downstream target molecule of the CaMK pathway, and dual activation of CaMK and calcineurin-dependent pathways by calcium has been proposed to regulate cardiac hypertrophy.19,73 In this regard, MEF2 associates with NFAT and GATA to facilitate transcriptional activation of hypertrophy-responsive genes75 (Fig. 15-3). From studies in skeletal muscle and neural tissue, the molecular mechanism of CaMK-dependent MEF2 regulation has been proposed as follows76: activated CaMK phosphorylates class II histone deacetylases (HDACs) (4, 5, 7, and 9: abundant in striated muscle and brain) or perhaps MEF2-interacting transcriptional facator-2 (MITR), which is a splicing variant of HDAC and lacks the catalytic domain. Phosphorylated HDACs or MITR binds to an intracellular chaperone protein 14-3-3, and this interaction exposes a nuclear export sequence (NES) on HDACs, which leads to CRM1-dependent translocation of HDAC/14-3-3 complex to the cytoplasm. On the other hand, MITR phosphorylation leads to the redistribution of the MITR/14-3-3 complex in the nucleus

277

because MITR lacks the NES. The dislocation of HDACs or MITR releases MEF2–interaction-dependent suppression of muscle gene activation. However, this CaMK/HDAC/MEF2 activation sequence has not yet been proved in cardiomyocytes. Interestingly, in cardiomyocytes the CaMK phosphorylation sites of HDAC/MITR seems to be catalyzed by other kinases, because the HDAC phosphorylation activity in cardiac extracts from calcineurin-transgenic mice or mice with cardiac hypertrophy induced by pressure-overload is insensitive to chemical CaMK inhibitors and EGTA, a divalent cation chelator.77 So far, the molecular identity of the putative cardiac HDAC kinase(s) remains unclear. On the other hand, p38 phosphorylates and directly activates MEF2.78 It also antagonizes nuclear translocalization of NFAT.79 In addition, GSK3 negatively regulates GATA480 and NFAT.66 As described, the transcriptional factor complex, including MEF2, NFAT, and GATA, is localized in the downstream of a web of multiple kinase cascades. Pharmacologic inhibition of calcineurin using cyclosporin A or FK506 has provided mixed results for their antihypertrophic effect in various experimental models.81–83 However, more compelling studies, using cardiac specific expression of dominant negative forms of calcineurin84 and endogenous calcineurin inhibitors,32,85 have confirmed a pivotal role for the calcineurin pathway in certain in vitro and in vivo assay systems. Subsequent studies in calcineurin Aβ-deficient mice revealed that changes in heart weight to tibia length ratio are diminished following acute pressure overload or chronic angiotensin II/isoproterenol infusion.86 Herein, the calcineurin Aβ form dominantly contributes to cardiac protein phosphatase 2B activity.In addition,targeted disruption of NFATc3,but not

FIGURE 15-3. Activation of the embryonic gene program in the hypertrophic heart via diverse membrane-nucleus signaling pathways. The signaling meshwork includes Gq, small GTP-biding proteins, MAP kinases, and calcium-calcineurin/CaM. The well-characterized ANF promoter lesion is shown in the lower panel with positioning of respective transactivators, such as MEF2,Tb×5, Nk×2.5, and GATA4.

278

CARDIAC MUSCLE DISEASES

NTATc4, was shown to attenuate both pressure-overload and calcineurin–overexpression-mediated mouse cardiac hypertrophy.87 On the other hand, calcineurin inhibition aggravated cardiac dysfunction in a hypertrophic cardiomyopathy (HCM) model carrying a point mutation of the myosin heavy chain gene,88 which corresponds to the human inherited form of cardiomyopathy89 and points out the need to further probe the clinical significance of calcineurin signaling in cardiac diseases. The recent notion that the constitutive activation of CaMKIIdelta(B), the major cardiac CaMK isotype, results in a dilated form of cardiomyopathy90 suggests another level of complexity regarding calcium-dependent signaling in the regulation of cardiac hypertrophy. The compartmental source for calcium that triggers biochemical calcium signaling has not been identified. It is uncertain whether the pool of calcium that activates components of the hypertrophic response is derived from the beatby-beat transient increase in free cytoplasmic calcium that underlies excitation-contraction coupling91 or whether this reflects a specific intracellular compartmentalization (such as specialized endoplasmic reticulum,92,93 myofilaments,94 or nuclear membrane) of calcium. Mice harboring a targeted disruption of calreticulin display embryonic cardiomyopathy, and the calreticulin-deficient myocytes show reduced NFAT translocation into the nucleus, suggesting the possibility that calcium mobilization from the distinctive cardiac cytoplasmic membrane structure where calreticulin localizes may activate calcineurin-dependent nuclear signaling.95 Another possibility is distinctive calciumdependent signaling pathways that dictate the particular frequency or concentration range of intracellular calcium oscillation.96–98 The pCa of calcineurin activation is also one order of magnitude lower than that required for CaMK.99,100 Thus, it appears that a cohesive, integrated view of the role of calcium signaling in hypertrophy is beginning to appear, although an extensive amount of work will be required. In addition, it will be of interest to determine how this calcium signaling is integrated into other calcium-independent signaling pathways for hypertrophy.

Gq Pathways A growing body of clinical and scientific evidence supports the viewpoint that Gq/G11 pathways are one of the most proximal steps in the hypertrophic response.18 By microinjection in cultured neonatal cardiomyocytes, the Gq protein was initially shown to be critical to the induction of morphologic hypertrophy and ANF expression.101 Subsequently, studies using either in vitro overexpression of Gq via an adenovirus vector or in vivo overexpression of Gq in transgenic mice resulted in cardiomyocyte hypertrophy.102 Excessive expression of Gq provokes cardiomyocyte apoptosis and associated cardiac dysfunction in both experimental systems. Herein, induction of a Bcl-2 family proapoptotic mitochondriatargeted protein Nix/Bni3L by Gq overexpression may play a cytopathic role via cytochrome c release and caspase-3 activation.103 The overexpression of the inhibitory peptide for Gq suppressed in vivo pressure-overload-

induced cardiac hypertrophy in mice,104,105 which was confirmed by a subsequent study using mice that harbor a double cardiac-restricted ablation of both Gq and G11.106 The downstream signaling of Gq/G11 activation is diverse and includes activations of small molecular GTP binding proteins, such as ras, rho, and rac; multiple MAP kinases; rho kinase; phosphatidyl inositol (PtdIns) hydrolysis; PKC; and CaMK16–19 (Fig. 15-3). The extent of crosstalk between calcium signaling, PI3K/Akt, ECMadhesion complex, and Gq/G11 pathways and their differential role in triggering downstream phenotypic features of cardiac hypertrophy are currently unclear. Other membrane G-proteins, including G12/G13, are also involved in ligand-dependent cardiac hypertrophy.107 A new Gqdependent pathway has recently been uncovered, which involves the Gq-dependent transactivation of EGF receptor through cleavage of heparin-binding EGF via the extracellular metalloprotease 12 (ADAM12).108 Further investigation will be needed to confirm that this intracellular or intercellular transactivation of tyrosine kinase receptor is indeed downstream of Gq-dependent signaling in the myocardium.

Protein Kinase C (PKC) Signaling PKC was originally identified in bovine brain extracts as a calcium/phospholipid-dependent protein kinase, and its diverse physiologic functions include regulation of cell proliferation, differentiation, motility, and intermediary metabolism.109 The in vitro effect of activated PKC on cardiomyocyte hypertrophy was noted relatively early (using direct PKC-activating agents; e.g., phorbol esters or diacylglycerols110) and was later confirmed in in vivo cardiac overexpression studies of various isoforms of PKC in transgenic mice.111–114 A combination of biochemical and molecular cloning strategies have now identified more than 12 members of the PKC family,which are categorized into four groups based on structural similarities: classic (cPKC), novel (nPKC), atypical (aPKC), and related genes.115 In cardiomyocytes, cPKCα, nPKCδ, and nPKCε are relatively abundant and consistently identified; aPKCζ and aPLCλ/ι are also detectable. The presence of cPKCβ1/2 in the heart has been inconsistent between laboratories.17,116–118 A two-step model of PKC activation has been proposed. PDK-1 accesses to the activation loop of PKCs for conformational changes associated with phosphorylation,119 which is sufficient for the activation of aPKCs, whereas the primed cPKCs need subsequent coupling with PtdIns hydrolysis.50,115 Gq-coupled agonists can lead to the translocation of nPKCδ and nPKCε to the cardiac membrane fraction from the cytoplasm,120–122 and these PKC isoforms display crosstalk and partly constitute a molecular complex with MAP kinases.123,124 Perhaps through c-Raf dependent ERK1/2 activation,125 nPKCε has been implicated in both cardiac hypertrophy and cardioprotective effects, whereas nPKCδ is known to be proapoptotic115 and may be linked to JNK and p38 pathways.126 Overexpression of an nPKCε-activating peptide ψεRACK1 in mice results in mild cardiac hypertrophy,127 whereas an nPKCε-inhibitory peptide εV1 accelerates induction of a heart failure phenotype in Gq-transgenic mice.128 Intriguingly, nPKCε-null mice, which have normal

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION

basal cardiac function, are blunted for sphingosine1-phosphate and ganglioside GM-1 mediated cardiac protection against ischemia-reperfusion injury.129 The cardioprotective action of nPKCε may be related to its partial colocalization with MAP kinases near cardiac mitochondria,130 which induces phosphorylation and inactivation of a proapoptotic protein Bad.130,131 On the other hand, earlier immunohistologic studies using isotype-specific antibodies have documented that nPKCε can rapidly translocate from the cytosol to the Z-disc accompanied by kinase activation,132 thereby raising the possibility that nPKCε is directly involved in remodeling of the cardiac cytoskeletal architecture or Z-disc related mechanical stress sensing machinery (see later).Two possible groups of proteins may recruit nPKCε to the Z-disc related structure: (1) β′COP, a selective binding protein (RACK) for activated nPKCε colocalizes in cardiomyocytes with crosstriation133 and (2) some PDZ-Lim domain proteins including ENH (Enigma-homolog)134,135 and ZASP/Cypher/ Oracle136–138 are known to interact with PKC134,136 and colocalize at the Z-disc.135,136

gp130 Signaling The first link between gp130 signaling and cardiac hypertrophy arose via studies of a novel cytokine that was capable of activating an increase in cardiomyocyte cell size via an expression cloning approach.34 A mouse embryonic stem cell cDNA library was screened for its activity to induce rat neonatal cardiomyocyte hypertrophy. This cytokine was named cardiotrophin-1 (CT-1) and was characterized as a member of the gp130 receptor cytokine family peptides.34,35 Separately, an inflammatory peptide IL-6, another gp130 cytokine, was documented to induce in vivo and in vitro cardiomyocyte hypertrophy by associating with the soluble form of the sIL-6 receptor.139 The gp130 peptide is a single transmembrane domain peptide that forms heterodimer complexes with various α subunit ligand-binding peptides. A LIF-binding subunit β (LIFR)-gp130 heterodimer was identified as CT-1 receptor on the surface of cardiomyocytes using a neutralizing antibody against LIFR.35 Dimerized gp130 cytokine receptor activates JAKs, which then induce phosphorylation of a tyrosine residue of gp130, followed by the phosphorylation of the family of STATs. Phosphorylated and active forms of STATs result in homodimerization or heterodimerization and are translocated into the nucleus, leading to target gene expression.140 Cardiac overexpression of STAT3 induces mild hypertrophy141 and also promotes cardiac hypervascularization, which might be linked to VEGF induction in the myocardium.142 Other kinase signaling cascades, including ERK1/2,143 MEK5/ERK5,144 and Akt,145 have also been linked to gp130 activation. In an in vitro cultured rat neonatal cardiomyocyte system, CT-1/LIF-treated cardiomyocytes showed unique polymorphic features of cardiomyocyte hypertrophy,146 which are distinct from Gq-coupling receptor agonists but often are observed in the myocytes isolated from the heart with eccentric hypertrophy. Accordingly, CT-1 induces longitudinal growth of myofilaments rather than lateral thickening of actin-myosin bundles. Protein and

279

RNA synthesis were also enhanced, Several characteristic markers for the embryonic gene program were reactivated: myofilaments were newly formed and cell surface area was enlarged by CT-1 treatment in cultured cardiomyocytes. The molecular mechanisms that lead to this distinct assembly of sarcomeres in series is not well understood at present but may include MEK5/ERK5 signaling144; Rho/Rho kinase dependent nonsarcomeric myosin light chain phosphorylation;147 and/or formation of nascent Z-discs with segregation of α-Actinin and other associated proteins such as ALP, a Z-disc PDZ-Lim protein.148

MAP Kinase Signaling Cytoplasmic MAP kinase, MAP kinase kinase (MKK), and MKK kinase signaling cascades are activated by diverse receptor-linked membrane signaling proteins including Gq and are known to have extensive crosstalk and interaction with many of the other previously mentioned signaling cascades.17,124 This intricate signaling meshwork includes three major nodal points, including ERK1/2, JNK1/2, and p38s (Fig. 15-3). Despite effort to determine the physiologic significance and precise downstream cardiac targets that are selectively triggered by these MAPK signaling molecules, a complete understanding of their exact role in cardiac hypertrophy and failure has been elusive because of their complexity and the inherent difficulty in distinguishing between primary and secondary effects. Endogenous activation of many of these molecules induce cardiac hypertrophy and cytoprotective effects, and enhanced activation results in toxicity with apoptotic or necrotic cell death in many cases. Synthetic small molecule kinase inhibitors are often less selective, and overexpression of dominant negative forms of kinases are uncertain for their specificity for molecular compartmentalization; however, more sophisticated genetic approaches, such as conditional gene ablation, are starting to shed light on this problem. Recent studies suggest that the MEK1/2-ERK1/2 cascade might be a central player in this kinase cascade meshwork and primarily induces a concentric form of cardiac hypertrophy and cardioprotection.124 Cardiac-restricted MEK1 transgenic mice have mild concentric form of hypertrophy with enhanced left-ventricular contractility.149 A significant ERK1/2 downstream nuclear target in the cardiomyocyte may be GATA4, which interacts with other cardiac specific transcriptional factors to regulate muscle gene expression150 (see later). In contrast, cardiac p38 activation is primarily cytopathic in vivo,151 which is partly due to its direct proapoptotic effects and also partly due to reduction in myofilament calcium sensitivity.152 TAK1, a p38 upstream kinase, also promotes cardiac hypertrophy with subsequent heart failure in vivo.153 JNK signaling was originally related to cardiomyocyte hypertrophy in vitro154; however, the significance of JNK signaling in in vivo cardiac hypertrophy was less clear in subsequent studies. MEKK1, a JNK upstream regulating kinase, was genetically ablated and has been tested in two in vivo mouse systems. Under conditions in which JNK activation was blunted by MEKK1 ablation, pressure-overload-induced in vivo

280

CARDIAC MUSCLE DISEASES

hypertrophy was not affected, although there was mild induction of cytopathic changes in the myocardium.155 On the other hand, cardiomyopathic changes in Gq transgenic mice were largely diminished by MEKK1 ablation.156 Part of these MEKK1/JNK signaling effects have been linked to JNK-dependent phosphorylation of a gapjunction molecule, connexin43.157 Interestingly, the significance of respective MAP kinase cascade activation has been studied in humans tissue, demonstrating the in vivo activation of JNK1/2 and p38. On the other hand, levels of activated ERK1/2 were found unchanged in the failing heart in these studies.60 Although these studies in human tissue are supportive, they have inherent difficulties related to individual variability, difficulty in obtaining sufficient control specimens, and hazards in extrapolating from a one-time measurement at the end stage of the disease to the pathways that trigger the initial steps of the disease-related pathways.

PATHWAYS FOR SURROGATE FAILING HEART PHENOTYPES

versus diastolic dysfunction, and outflow tract obstruction, leading to three clinical subtypes: HCM, dilated cardiomyopathy (DCM), and restrictive cardiomyopathy (RCM).168,169 Genotyping of human cardiomyopathy was initiated with the surprising discovery of mutations in the β-Myosin heavy chain gene in HCM cases in large pedigrees170; this was subsequently expanded to include other sarcomeric gene mutations in human HCM patients, including myosin binding protein-C,171,172 myosin light chains,173 cardiac actin,174 troponin-T,171,175 troponin-I,176 α-Tropomyosin,171,175 and titin.177 On the other hand, parallel analyses of candidate genes for DCM were more difficult, because of the relatively small pedigrees, incomplete penetrance, and phenotypic diversity. Eventually, studies in genetically engineered mice revealed an initial role for cytoskeletal defects in DCM via studies of mutant mice that are deficient in the cardiac Z-disc–associated cytoskeletal muscle-specific LIM protein (MLP)178 (Fig. 15-4). Later, a missense mutation of MLP was identified in a subset of DCM patients who reside in northern Europe.179 Different MLP mutations were also linked to familial cases of HCM.180 Extensive links between other cytoskeletal proteins and familial forms of human DCM were subsequently established.6,168,169 Based on these early findings, a generalized concept was formulated that suggested that sarcomeric mutations lead to HCM, whereas cytoskeletal mutations result in DCM, thereby reflecting specific defects in the hardwiring within cardiac muscle cells that govern these two distinct phenotypes. Thus, the phenotypic diversity of familial cardiomyopathies appeared to be primarily driven by the disease genotype. However, recent experimental and clinical studies suggest a more complex genotype-phenotype relationship of cardiomyopathies168,169 (Table 15-2). It now appears that mutations in a given sarcomeric gene can lead to a spectrum of cardiomyopathic phenotypes, often overlapping between the clinical subsets of DCM, HCM, and RCM. Although there is little doubt that the disease genotype plays a critical role in initiating the cardiomyopathic process, the ultimate clinical phenotype seems to represent the integrated effect of multiple interacting factors.This view is supported by a host of circumstantial evidence, including the poor penetrance of many cardiomyopathic genotypes; the influence of hemodynamic stress on disease progression; the secondary effects of the loss of cardiac myocyte survival and subsequent replacement fibrosis; strong modifying effects of calcium cycling and calcium signaling; and clear evidence of genetic background effects, which is also apparent in gene-targeted mouse models of cardiomyopathy.5,204

Genotype-Phenotype Relationships in Cardiomyopathy

Chamber Dilation: Diverse Cytoskeletal Pathways

The initial precepts that guided the understanding of human cardiomyopathies were based on the presumption that the primary determinant of the clinical phenotype is the molecular genotype. This clinical viewpoint is supported by noninvasive analyses, such as echocardiography, that can quantitatively assess differences in chamber volume, wall thickness, hypertrophy, systolic

Changes in organ size, shape, and histologic phenotype are central features of cardiomyopathy and related heart failure. Chronic increases in ventricular volume and filling pressure can lead to dilation of the cardiac chamber and subsequent thinning of the ventricular wall in the advancement of heart failure, which results in large increases in diastolic and systolic wall stress in both the

Transcriptional Regulation of Cardiac Genes The activation of a fetal gene program is a highly conserved feature of the hypertrophic response and has been extensively studied as a means to identify pathways that might be involved in the in vivo response. Although the initial steps of the induction of embryonic genes are clearly reversible, chronic changes in the cardiac gene program may trigger pathologic changes in the myocardium that can be associated with irreversible dysfunction. Much of this work has focused on the inducibility of ANF gene, which is reactivated in ventricular muscle early during the hypertrophic response (Fig. 15-3). A host of cardiac transcription factors, including NKX 2.5,158 a zinc-finger protein GATA4,159,160 NFATs,72 TBX5,161,162 dHAND,163 CBP/p300,163,164 MEF2,74 and another MADS box protein serum response factor (SRF),160 work together to control the expression of ANF and a panel of downstream target genes in the embryonic gene program. The recent discovery of a new series of cardiac restricted transcriptional cofactors such as Myocardin165 and HOP166,167 have suggested the combinatorial control of the cardiac gene program in response to defined upstream signals. As mentioned previously, a dual enzyme system composed of histone acetyltransferases (HATs) and HDACs is associated with this transcriptional complex and serves as an epigenetic control point of the embryonic cardiac program.76,163,164

FIGURE 15-4. Lim domain proteins and cardiomyopathy. Cardiac-enriched Lim domain proteins are categorized based on their domain structure. MLP-null mice were initially reported to display DCM phenotypes, followed by the identification of human MLP mutations linked to cardiomyopathy. Later, cardiomyopathic phenotypes were also noted in a series of experimental model animals in which Lim domain proteins are ablated.

TABLE 15-2 MOLECULAR DEFECTS LINKED TO HUMAN CARDIOMYOPATHIES DEFECTS IN HUMANS Genomic Defects

HCM

DCM

Missense171,181,182 Missense173 Missense173 Missense174 Missense/deletion171,175 Missense176 Missense171,175 Missense/deletion171,172

Missense185

Missense177

Missense/deletion188 Missense179

RCM

Sarcomere Myosin heavy chain Myosin essential light chain Myosin regulatory light chain Cardiac actin Troponin-T Troponin-I α-tropomyosin Myosin binding protein-C

Missense186 Deletion185 Missense203 Missense187

Titin/titin-related protein Titin Telethonin (T-cap) Z-disk-associated proteins Missense179

MLP Sarcolemmal cytoskeleton

Deletion189–191 Deletion/duplication192 Missense193 Missense194 Deletion195,196

Dystrophin β-Sarcoglycan δ-Sarcoglycan α-Dystrobrevin Metavinculin Intermediate filaments

Missense197,198 Missense199

Desmin Lamin A/C Miscellaneous Mitochondrial respiratory chain G4.5 (tafazzin) Phospholamban

Nonsense183/ promoter184

Missense200 Nonsense201 Missense/nonsense183–202

282

CARDIAC MUSCLE DISEASES

right and left ventricular chambers. Based on the discovery of a cytoskeletal network of DCM-related genes in both experimental and clinical studies, distinct classes of cytoskeletal defects have been uncovered that include components of the macromolecular titin complex, Z-disc and intercalated disc proteins, components of the dystrophin/dystroglycan/sarcoglycan complex, and mutations in the nuclear and intermediate filaments (Table 15-2). These diverse cytoskeletal defects have been proposed to lead to chamber dilation via multiple potential mechanisms, such as abnormalities in force transmission,178,186 force generation,185 sarcolemmal integrity,205 sarcomeric organization/assembly defects,144 and intercalated disc stability.206 In addition to these structural roles of the cardiac cytoskeleton, selective cytoskeletal mutations and associated defects may exert their effects on heart failure via specific effects on intracellular signaling pathways triggered by biomechanical or hormonal stimuli to promote cardiac remodeling.179 MLP was demonstrated to directly bind to telethonin (T-cap), a 19-kd muscle-specific titin-interacting protein, and MLP deficiency causes T-cap dislocation from Z-disc structure. Physiologic analysis of MLP-deficient myocardium, before the gross development of DCM, has revealed a selective defect in the cardiac mechanical stress sensor, which is related to abnormalities in the titin-related complex179 (Fig. 15-5). MLP is downregulated at the protein level in the myocardium of both sporadic DCM and ischemic cardiomyopathy patients.207 In this manner, cardiac cytoskeletal defects in mechanical stress sensing might also trigger chamber dilation in acquired forms of heart failure, extending the observations obtained in rare forms of DCM to more common forms of heart failure. The recent association of cytoskeletal components with distinct cardiac channels (min K)208 and the identification of a mutation of ankyrin-B (a membrane cytoskeletal protein and an associated protein of the sarcolemmal sodium-potassium ATPase) in long QT syndrome type 4209 have further raised the possibility of divergent roles of cytoskeletal components in the direct regulation of cardiac channels involved in the control of cardiac repolarization and/or excitation–contraction coupling. An expanding number of novel musclespecific cytoskeletal components have been found localized in the cardiac Z-disc, thereby implying that the Z-disc may play a specialized function beyond its known role as a structural element210 (Fig. 15-6). At the Z-disc, αactinins are organized into a series of antiparallel dimers that crosslink polymerized actin filaments (thin filaments). The Z-disc also anchors titin, a 2- to 3-Md muscle-specific protein. Titin contains two α-actinin binding domains at its N-terminus and spans the length of each half-sarcomere, reaching to the M line in the middle of the A-band. T-cap interacts with titin at the Z-disc. As mentioned previously, a series of signaling molecules including several PKC isoforms, are known to translocate to Z-disc after activation; accordingly, Z-disc proteins with the PDZ-Lim structure including ENH134,135 and ZASP/Cypher/Oracle136–138 and the calcineurin-binding proteins Calsarcin/FATZ211–213 may serve as mechanicalto-biologic signal transducers.

FIGURE 15-5. MLP/T-cap/titin complex and the cardiac stretch sensor. At peak diastole, as ventricular filling pressure rises, titin elastic segments are reshaped, generating diastolic wall stress. The change in intrinsic elasticity functions to trigger the mechanical stress sensor. In DCM (e.g., the MLP-null mouse heart), ventricular chamber dilation and wall thinning act to enhance diastolic wall stress; the increased wall stress destabilizes the function of the titin/Z-disc complex (e.g., in the MLP-null mouse heart,T-cap, which is mechanically stabilized by MLP, gradually dislocates from Z-disc); the titin/Z-disc related mechanical stress sensor fails in translating mechanical stress to the cytoplasmic signaling cascade. The diastolic wall stress reduction appears to be critical for the prevention and rescue of heart failure progression by stabilizing titin/Z-disc complex. For example, in the MLP/phospholamban double knockout (MLP/PLN double KO) mouse heart, Z-disc abnormality, which is apparent in MLP-null myocardium, is genetically rescued.179 (Modified from Knoll R, Hoshijima M, Hoffman HM, et al: The cardiac mechanical stretch sensor machinery involves a Z disc complex that is defective in a subset of human dilated cardiomyopathy. Cell 2002;111:943–955.)

Excitation-Contraction Coupling in Heart Failure Progression: A Common Pathologic Endpoint Associated with Defects in Calcium Cycling During the course of cardiac chamber dilation, a progressive decrease in cardiac contractility and relaxation ensues, which ultimately leads to chronic heart failure. The precise link between chamber dilation and cardiac dysfunction is not completely clear, because there are numerous defects that have been associated with the

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION

283

FIGURE 15-6. Cardiac cytoskeletal proteins and cardiomyopathy. Representative cytoskeletal proteins constituting cardiac Z-disc and connecting myofilaments to the costamere are illustrated. (Modified from Chien KR: Genomic circuits and the integrative biology of cardiac diseases. Nature 2000;407:227–232.) *Proteins linked to human and experimental cardiomyopathies.

impairment of cardiac contractile function in heart failure. However, recent studies point to the central role of defects in calcium cycling in heart failure progression. These defects arise from a decrease in the activity of the SR calcium ATPase type 2 (SERCA2) that regulates diastolic and systolic function via the resequestration of calcium into the SR lumen.214–216 During the onset of heart failure, there is a decrease in the content of SERCA2 and its activity relative to the unrestrained inhibitory effect of phospholamban (PLN), an endogenous SERCA2 inhibitory peptide.215 In the normal heart, the inhibitory activity of PLN is under the control of the β-adrenergic receptor (β-AR)-adenylyl cyclase-cAMP dependent PKA pathway in which phosphorylation of the S16 residue of PLN leads to an inactivation of its suppressor effect on SERCA2,217, 218 (Fig. 15-7). In human and experimental models of heart failure, PLN is hypophosphorylated,219–221 which most likely reflects desensitization of the β-adrenergic pathway resulting from the increase in the activity of the β-adrenergic receptor kinase (βARK: GRK2) in the failing heart.222 Recently, a PLN mutation (R9CPLN) was found in patients with familial DCM, which blocks the PKA-dependent site-specific phosphorylation in both R9CPLN and co-oligomerized wild-type PLNs.202 Together with the experimental finding that cardiac restricted R9CPLN overexpression leads to progressive heart fail-

ure,202 SR calcium uptake suppression appears to be a critical step in the progression of heart failure. Although clinical and experimental studies document that simply enhancing cAMP during heart failure can lead to cardiac injury and an associated increase in mortality,223–225 genetic complementation studies and somatic gene transfer in heart failure model animals have suggested that it is possible to markedly improve cardiac diastolic and systolic function independent of effects on cAMP levels via directly enhancing SR calcium cycling. The clear therapeutic effects of β-adrenergic receptor blockade, even at advanced stages of heart failure, suggests that any new therapy will have to be compatible with chronic β-blocker therapy.226 Percutaneous or intramuscular delivery of an adenovirus vector (Adeno) carrying β2-AR (Adeno/β2-AR) delivery results in enhanced contractility in normal rabbit or cardiomyopathic hamsters.227,228 βAR kinase inhibitor peptide (βARKct)-transgenic overexpression partially rescues cardiomyopathic phenotypes of MLP-null mice229 and R403Q mutant α-myosin heavy chain (αMHC) transgenic mice.230 Adeno/βARKct also improves contractility and delays heart failure development in postmyocardial infarction rabbits.231,232 Adenylyl cyclase type VI (AC-type VI) transgenic overexpression rescues the heart failure phenotype in Gq cardiomyopathic mice,233 and percutaneous

284

CARDIAC MUSCLE DISEASES

FIGURE 15-7. Regulatory pathways and key molecules in the control of SR calcium cycling. The G-protein coupling receptor (GPCR)/cAMP/PKA axis is the main regulating system and serves as a pharmacologic and genetic target at multiple steps in the cascade to improve cardiac contractility and relaxation in heart failure. (Modified from Hoshijima M, Chien KR: Mixed signals in heart failure: Cancer rules. J Clin Invest 2002;109:849–855.)

Adeno/AC-type VI delivery enhances cardiac contractility in normal pigs.234 SERCA2 transgenic mice have enhanced cardiac function,235 and SERCA2 transgenic overexpression partially rescues MLP-null cardiac dysfunction,236 whereas Adeno/SERCA2 gene delivery improves cardiac contractility and prognosis of postaortic-banded rats.237,238 PLN ablation induces a hyperkinetic phenotype in the normal mouse heart239 and dominantly complements cardiomyopathic changes in the MLP-null animals240 The genetic rescue effect of PLN ablation might be modeldependent, because it is not obvious in Gq transgenic mice (an apoptotic heart failure model) and homozygous myosin binding protein-C mutant animals (a neonatalonset progressive ventricular dilation model)241; however, molecular intervention to target PLN-SERCA2 interaction demonstrates clear beneficial effects, which are indeed chronic.44 Enhancement of isolated cardiomyocytes from endstage heart failure patients is also promoted by suppression of PLN expression.242 Diastolic leakage of calcium from SR through a SR calcium release channel (ryanodine receptor: RyR) has been linked to the PKA-dependent hyper-phosphorylation of RyR and subsequent dissociation of FKBP12.6, a stabilizing protein of RyR from the RyR/FKBP12.6 complex243; enhanced diastolic SR calcium leakage was later confirmed in FKBP12.6-null mice.244 Thus, long-term beneficial effects of β-blocker therapy may be partly attributed

to the stabilization of the RyR/FKBP12.6 complex.245,246 The evidence that human RyR mutations247–249 can lead to polymorphic ventricular tachycardia and associated sudden death250 further supports this possibility. The in vitro transfection of Adeno/FKBP12.6 in cultured cardiomyocyte increases contractility,251 suggesting that RyR stabilization may also improve cardiac contractile in the failing hearts.

Molecular Substrates for Arrhythmogenesis in Heart Failure: Acquired Defects in Conduction Lineages, Intercellular Coupling, Repolarization, and Action Potential Duration (APD) One of the major causes of mortality in heart failure is the onset of life-threatening malignant arrhythmias. The incidence of cardiac sudden death and associated cardiac rhythm abnormalities correlates with the severity of dysfunction, but the mechanism that serves as the substrate for this markedly increased susceptibility is unclear. In addition to the abnormal delayed inward depolarization current that is partly associated with diastolic SR calcium leak (see previous discussion), a growing body of electrophysiologic, genetic, experimental, and clinical data is supporting the concept that hetero-

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION

geneity of APD throughout the myocardium of the failing heart may partly account for the arrhythmogenic substrate that creates susceptibility for reentrant ventricular tachyarrrhythmias.252,253 Much of this insight has been gleaned from genetically based forms of arrhythmias in clinical and animal model systems. Several distinct classes of mutations have been shown to be associated with cardiac arrhythmogenesis. Studies of long QT syndrome, an autosomal dominant disease associated with an increased incidence of cardiac sudden death, have indicated that the disease is due to mutations in channel pore-forming proteins and interacting proteins of iontransporters that can lead to prolongation of the QT interval and associated APD prolongation.209,252 A second class of arrhythmogenic defects has recently been uncovered, which are related to abnormalities in cardiac intercellular conduction that is associated with the loss of cardiac connexins.157,254,255 The third class of mutations associated with malfunctions in the conduction system and cardiac sudden death have been discovered in human patients and from studies using genetically engineered mouse models. The mutations occur because of defects in cardiac transcription factors that control muscle gene expression in both ventricular muscle and conduction system cell lineages.162,256,257 Recent studies have further implicated a glycogen-storage abnormality associated with HCM that contributes to progressive conduction defects within the conduction system itself.258–260 Because it is unlikely that somatic mutations account for most cases of cardiac sudden death associated with progressive heart failure, the generalizable mechanisms that accompany arrhythmogenesis in this acquired form

FIGURE 15-8. Ion transporter abnormalities and the substrate for malignant ventricular arrhythmia in KChIP2-null (KChIP-/-) myocardium. The absence of KChIP2 blocks the translocation of Kv4.2/4.3 pore-forming subunits of membrane potassium channels,leading to a loss of the cardiac Ito current. The transmural gradient of Ito current, which is normally high in the outer layer, is lost in the KChIP2-null myocardium,resulting in the heterogeneity of repolarization. This repolarization heterogeneity can create multiple reentry circuits with unidirectional conduction block, which provide a substrate for malignant ventricular arrhythmia. (Modified from Kuo H, Cheng CF, Clark RB, et al: A defect in the Kv Channel-interacting protein 2 (KCHIP2) gene leads to a complete loss of I (to) and confers susceptibility to ventricular tachycardia. Cell 2001;107:801–813.)

285

of the disease warrant investigation. There are several conserved electrophysiologic features of human heart failure, which include elongation of APD, enhanced inward current associated with sarcolemmal sodiumcalcium exchanger activity, and the loss of a transmural gradient of phase 1 Ito potassium current.214 The mechanisms that contribute to the loss of the Ito gradient are unclear, and the relative importance of changes in this specific current versus the multitude of other changes in the electrophysiologic phenotype of the failing heart is unknown. Recent studies have clearly identified that the loss of Ito can independently increase the dispersion of repolarization and confer marked susceptibility to lethal forms of ventricular tachycardia.261 These observations are based on the discovery of a cytoplasmic accessory protein (KChIP2) that binds to the Kv4.2 and 4.3 channel proteins and that is markedly downregulated early during pressure-overload hypertrophy.261 This negative regulation mimics the reactivation of the fetal gene program, because the KChIP2 gene is not expressed in the fetal heart until the late embryonic stage and is postnatally activated in the adult heart. Interestingly, KChIP2 quantitatively regulates the Ito current, because heterozygotes for the mutant allele display a precise 50% decrease in current density. Mice that lack KChIP2 can display a loss of the normal Ito current gradient across the ventricular wall and sustained polymorphic ventricular tachycardia with a single, extra stimulus in programmed stimulation studies261 (Fig. 15-8). Because human and canine hearts express the KChIP2 gene in a gradient (epicardium greater than endocardium) that matches with the distribution of Ito current (the Kv4.2 and 4.3 genes are uniformly expressed throughout the

286

CARDIAC MUSCLE DISEASES

ventricular wall262,263), it is likely that the KChIP2 gene quantitatively regulates the Ito current in the human heart. Therefore, it will be interesting to determine whether there is a downregulation of KChIP2 gene expression in the failing human heart that might account for the loss of the Ito gradient in human heart failure. If such is the case, then defects in KChIP2 may represent a new class of genetic modifiers that can confer risk for both inheritable and acquired forms of ventricular arrhythmias. Because the expression of KChIP2 is downregulated during cardiac remodeling,261 strategies to maintain Ito by promoting the level of KChIP2 expression in the failing heart may represent a new therapeutic approach for combating the prevalent lethal arrhythmias associated with heart failure. The roles of other channel accessory proteins that may be important in the regulation of other cardiac currents and how they confer susceptibility to arrhythmogenesis in the setting of heart failure should be investigated.

TOWARD NEW THERAPEUTIC PATHWAYS Genomic databases, which provide a universal language that can be used to identify the conserved functions of cardiovascular genes in multiple species and organ systems, are ushering in a new era of integrative biology.6 For example, numerous parallels exist between cardiac muscle and neuromuscular disorders (e.g., excitability, conduction, terminal differentiation, and innervation). Unsuspected connections between neural and cardiac developmental gene programs have been uncovered in gene-targeted mice, and conserved myocyte and neuronal cell survival factors have been discovered, which suggest the existence of other factors. Thus, major opportunities for understanding cardiac diseases may lie in working at the interface of cardiovascular science and neuroscience. To evaluate the real clinical potential of disease pathways found in mouse models, parallel studies in larger animals will be necessary. The difficulty of germ-line gene manipulation in larger animals suggests the value of new strategies using peptide inhibitors, neutralizing antibodies, and somatic gene transfer with smart vectors that allow long-term expression. The many quantitative end points for complex in vivo cardiac phenotypes should empower the next frontier that lies at the boundaries of genomic databases, physiology, and human disease. We are entering a renaissance of integrative biology of the intact organism, and the heart may again play a key role in defining the circuitry for complex in vivo physiologic traits.

REFERENCES 1. Cohn JN, Bristow MR, Chien KR, et al: Report of the National Heart, Lung, and Blood Institute Special Emphasis Panel on Heart Failure Research. Circulation 1997;95:766–770. 2. McMurray J, Pfeffer MA: New therapeutic options in congestive heart failure. I. Circulation 2002;105:2099–2106. 3. McMurray J, Pfeffer MA: New therapeutic options in congestive heart failure. II. Circulation 2002;105:2223–2228.

4. Angeja BG, Grossman W: Evaluation and management of diastolic heart failure. Circulation 2003;107:659–663. 5. Chien KR: Stress pathways and heart failure. Cell 1999;98: 555–558. 6. Chien KR: Genomic circuits and the integrative biology of cardiac diseases. Nature 2000;407:227–232. 7. Franz WM, Muller OJ, Katus HA: Cardiomyopathies: From genetics to the prospect of treatment. Lancet 2001;358:1627–1637. 8. Charron P, Komajda M: Are we ready for pharmacogenomics in heart failure? Eur J Pharmacol 2001;417:1–9. 9. Rockman HA, Ross RS, Harris AN et al: Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy. Proc Natl Acad Sci USA 1991;88:8277–8281. 10. Yasukawa H, Hoshijima M, Gu Y 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 Invest 2001;108:1459–1467. 11. Fischer TA, Ludwig S, Flory E et al: Activation of cardiac c-Jun NH(2)-terminal kinases and p38-mitogen-activated protein kinases with abrupt changes in hemodynamic load. Hypertension 2001;37:1222–1228. 12. http://cardiogenomics.med.harvard.edu/public-data 13. Hu H, Sachs F: Stretch-activated ion channels in the heart. J Mol Cell Cardiol 1997;29:1511–1523. 14. Ruwhof C, van der Laarse A: Mechanical stress-induced cardiac hypertrophy: Mechanisms and signal transduction pathways. Cardiovasc Res 2000;47:23–37. 15. Yamazaki T, Komuro I, Kudoh S, et al: Role of ion channels and exchangers in mechanical stretch-induced cardiomyocyte hypertrophy. Circ Res 1998;82:430–437. 16. Sadoshima J, Izumo S:The cellular and molecular response of cardiac myocytes to mechanical stress. Annu Rev Physiol 1997;59: 551–571. 17. Sugden PH, Clerk A: Cellular mechanisms of cardiac hypertrophy. J Mol Med 1998;76:725–746. 18. Molkentin JD, Dorn IG 2nd: Cytoplasmic signaling pathways that regulate cardiac hypertrophy. Annu Rev Physiol 2001;63: 391–426. 19. Frey N, Olson EN: Cardiac hypertrophy: The good, the bad, and the ugly. Annu Rev Physiol 2003;65:45–79. 20. Choquet D, Felsenfeld DP, Sheetz MP: Extracellular matrix rigidity causes strengthening of integrin-cytoskeleton linkages. Cell 1997; 88:39–48. 21. Ross RS, Borg TK: Integrins and the myocardium. Circ Res 2001;88:1112–1119. 22. Shai SY, Harpf AE, Babbitt CJ, et al: Cardiac myocyte-specific excision of the beta1 integrin gene results in myocardial fibrosis and cardiac failure. Circ Res 2002;90:458–464. 23. Brancaccio M, Fratta L, Notte A, et al: Melusin, a muscle-specific integrin beta1-interacting protein, is required to prevent cardiac failure in response to chronic pressure overload. Nat Med 2003;9:68–75. 24. Hunter JJ, Grace A, Chien KR: Molecular and cellular biology of cardiac hypertrophy and failure. In Chien KR (ed): Molecular Basis of Cardiovascular Disease. Philadelphia, WB Saunders, 1999, pp 211–250. 25. Hwang DM, Dempsey AA,Wang RX, et al:A genome-based resource for molecular cardiovascular medicine: Toward a compendium of cardiovascular genes. Circulation 1997;96: 4146–4203. 26. Yang J, Moravec CS, Sussman MA, et al: Decreased SLIM1 expression and increased gelsolin expression in failing human hearts measured by high-density oligonucleotide arrays. Circulation 2000;102:3046–3052. 27. Friddle CJ, Koga T, Rubin EM, Bristow J: Expression profiling reveals distinct sets of genes altered during induction and regression of cardiac hypertrophy. Proc Natl Acad Sci USA 2000;97:6745–6750. 28. Redfern CH, Degtyarev MY, Kwa AT, et al: Conditional expression of a Gi-coupled receptor causes ventricular conduction delay and a lethal cardiomyopathy. Proc Natl Acad Sci USA 2000; 97:4826–4831. 29. Lim DS, Roberts R, Marian AJ: Expression profiling of cardiac genes in human hypertrophic cardiomyopathy: Insight into the pathogenesis of phenotypes. J Am Coll Cardiol 2001;38: 1175–1180.

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION 30. Johnatty SE, Dyck JR, Michael LH, et al: Identification of genes regulated during mechanical load-induced cardiac hypertrophy. J Mol Cell Cardiol 2000;32:805–815. 31. Chien KR, Knowlton KU, Zhu H, Chien S: Regulation of cardiac gene expression during myocardial growth and hypertrophy: Molecular studies of an adaptive physiologic response. FASEB J 1991;5:3037–5046. 32. Hunter JJ, Chien KR: Signaling pathways for cardiac hypertrophy and failure. N Engl J Med 1999;341:1276–1283. 33. Yasukawa H, Sasaki A, Yoshimura A: Negative regulation of cytokine signaling pathways. Annu Rev Immunol 2000;18: 143–164. 34. Pennica D, King KL, Shaw KJ et al: Expression cloning of cardiotrophin 1, a cytokine that induces cardiac myocyte hypertrophy. Proc Natl Acad Sci USA 1995;92:1142–1146. 35. Wollert KC, Chien KR: Cardiotrophin-1 and the role of gp130dependent signaling pathways in cardiac growth and development. J Mol Med 1997;75:492–501. 36. 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;122:419–428. 37. Fujio Y, Kunisada K, Hirota H, et al: Signals through gp130 upregulate bcl-x gene expression via STAT1-binding cis-element in cardiac myocytes. J Clin Invest 1997;99:2898–2905. 38. Yamauchi-Takihara K, Kishimoto T:A novel role for STAT3 in cardiac remodeling. Trends Cardiovasc Med 2000;10:298–303. 39. Rothermel BA, McKinsey TA,Vega RB, et al: Myocyte-enriched calcineurin-interacting protein, MCIP1, inhibits cardiac hypertrophy in vivo. Proc Natl Acad Sci USA 2001;98:3328–3333. 40. Yang J, Rothermel B, Vega RB, et al: Independent signals control expression of the calcineurin inhibitory proteins MCIP1 and MCIP2 in striated muscles. Circ Res 2000;87:E61–68. 41. Bueno OF, De Windt LJ, Lim HW, et al: The dual-specificity phosphatase MKP-1 limits the cardiac hypertrophic response in vitro and in vivo. Circ Res 2001;88:88–96. 42. Ruiz-Lozano P, Chien KR: Cre-constructing the heart. Nat Genet 2003;33:8–9. 43. Ikeda Y, Gu Y, Iwanaga Y, et al: Restoration of deficient membrane proteins in the cardiomyopathic hamster by in vivo cardiac gene transfer. Circulation 2002;105:502–508. 44. Hoshijima M, Ikeda Y, Iwanaga 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;8:864–871. 45. Duerr RL, Huang S, Miraliakbar HR, et al: Insulin-like growth factor1 enhances ventricular hypertrophy and function during the onset of experimental cardiac failure. J Clin Invest 1995;95: 619–627. 46. Tanaka N, Ryoke T, Hongo M et al: Effects of growth hormone and IGF-I on cardiac hypertrophy and gene expression in mice. Am J Physiol 1998;275:H393–99. 47. Barton ER, Morris L, Musaro A, et al: Muscle-specific expression of insulin-like growth factor I counters muscle decline in mdx mice. J Cell Biol 2002;157:137–148. 48. Rommel C, Bodine SC, Clarke BA, et al: Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol 2001;3:1009–1013. 49. Bodine SC, Stitt TN, Gonzalez M, et al:Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 2001;3:1014–1019. 50. Glass DJ: Signalling pathways that mediate skeletal muscle hypertrophy and atrophy. Nat Cell Biol 2003;5:87–90. 51. Cohen P, Frame S: The renaissance of GSK3. Nat Rev Mol Cell Biol 2001;2:769–776. 52. Scheid MP,Woodgett JR: Pkb/akt: Functional insights from genetic models. Nat Rev Mol Cell Biol 2001;2:760–768. 53. Hardt SE, Sadoshima J: Glycogen synthase kinase-3beta: A novel regulator of cardiac hypertrophy and development. Circ Res 2002;90:1055–1063. 54. Fujio Y, Nguyen T, Wencker D, et al: Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. Circulation 2000;101:660–667. 55. Wu W, Lee WL, Wu YY, et al: Expression of constitutively active phosphatidylinositol 3-kinase inhibits activation of caspase 3 and apoptosis of cardiac muscle cells. J Biol Chem 2000;275: 40113–40119.

287

56. Vanhaesebroeck B, Alessi DR: The PI3K-PDK1 connection: More than just a road to PKB. Biochem J 2000;346(Pt 3):561–576. 57. Toker A, Newton AC: Cellular signaling: Pivoting around PDK-1. Cell 2000;103:185–188. 58. Vanhaesebroeck B, Leevers SJ,Ahmadi K, et al: Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem 2001;70:535–602. 59. Backman S, Stambolic V, Mak T: PTEN function in mammalian cell size regulation. Curr Opin Neurobiol 2002;12:516–522. 60. Haq S, Choukroun G, Lim H et al: Differential activation of signal transduction pathways in human hearts with hypertrophy versus advanced heart failure. Circulation 2001;103:670–677. 61. Reiss K, Cheng W, Ferber A, et al: Overexpression of insulin-like growth factor-1 in the heart is coupled with myocyte proliferation in transgenic mice. Proc Natl Acad Sci USA 1996;93:8630–8635. 62. Shioi T, Kang PM, Douglas PS et al:The conserved phosphoinositide 3-kinase pathway determines heart size in mice. EMBO J 2000;19:2537–2548. 63. Shioi T, McMullen JR, Kang PM, et al: Akt/protein kinase B promotes organ growth in transgenic mice. Mol Cell Biol 2002;22:2799–809. 64. Condorelli G, Drusco A, Stassi G et al: Akt induces enhanced myocardial contractility and cell size in vivo in transgenic mice. Proc Natl Acad Sci USA 2002;99:12333–12338. 65. Matsui T, Li L,Wu JC, et al: Phenotypic spectrum caused by transgenic overexpression of activated Akt in the heart. J Biol Chem 2002;277:22896–22901. 66. Antos CL, McKinsey TA, Frey N, et al: Activated glycogen synthase-3 beta suppresses cardiac hypertrophy in vivo. Proc Natl Acad Sci USA 2002;99:907–912. 67. Schwartzbauer G, Robbins J: The tumor suppressor gene PTEN can regulate cardiac hypertrophy and survival. J Biol Chem 2001;276:35786–35793. 68. Crackower MA, Oudit GY, Kozieradzki I, et al: Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell 2002;110:737–749. 69. Toker A, Cantley LC: Signalling through the lipid products of phosphoinositide-3-OH kinase. Nature 1997;387:673–676. 70. Ramirez MT, Zhao XL, Schulman H, Brown JH: The nuclear deltaB isoform of Ca2+/calmodulin-dependent protein kinase II regulates atrial natriuretic factor gene expression in ventricular myocytes. J Biol Chem 1997;272:31203–31208. 71. Kirchhefer U, Schmitz W, Scholz H, Neumann J:Activity of cAMPdependent protein kinase and Ca2+/calmodulin-dependent protein kinase in failing and nonfailing human hearts. Cardiovasc Res 1999;42:254–261. 72. Molkentin JD, Lu JR, Antos CL, et al: A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell 1998; 93:215–228. 73. Frey N, McKinsey TA, Olson EN: Decoding calcium signals involved in cardiac growth and function. Nat Med 2000;6: 1221–1227. 74. Passier R, Zeng H, Frey N et al: CaM kinase signaling induces cardiac hypertrophy and activates the MEF2 transcription factor in vivo. J Clin Invest 2000;105:1395–1406. 75. Han J, Molkentin JD: Regulation of MEF2 by p38 MAPK and its implication in cardiomyocyte biology. Trends Cardiovasc Med 2000;10:19–22. 76. McKinsey TA, Zhang CL, Olson EN: Control of muscle development by dueling HATs and HDACs. Curr Opin Genet Dev 2001;11:497–504. 77. Zhang CL, McKinsey TA, Chang S, et al: Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell 2002;110:479–488. 78. Zhao M, New L, Kravchenko VV, et al. Regulation of the MEF2 family of transcription factors by p38. Mol Cell Biol 1999;19: 21–30. 79. Yang TT, Xiong Q, Enslen H, et al: Phosphorylation of NFATc4 by p38 mitogen-activated protein kinases. Mol Cell Biol 2002;22: 3892–3904. 80. Morisco C, Seta K, Hardt SE et al: Glycogen synthase kinase 3beta regulates GATA4 in cardiac myocytes. J Biol Chem 2001; 276:28586–28597. 81. Olson EN, Molkentin, JD: Prevention of cardiac hypertrophy by calcineurin inhibition: Hope or hype? Circ Res 1999;84:623–632.

288

CARDIAC MUSCLE DISEASES

82. Sugden PH: Signaling in myocardial hypertrophy: Life after calcineurin? Circ Res 1999;84:633–646. 83. Wilkins BJ, Molkentin JD: Calcineurin and cardiac hypertrophy: Where have we been? Where are we going? J Physiol 2002; 541:1–8. 84. Zou Y, Hiroi Y, Uozumi H, et al: Calcineurin plays a critical role in the development of pressure overload-induced cardiac hypertrophy. Circulation 2001;104:97–101. 85. De Windt LJ, Lim HW, Bueno OF, et al: Targeted inhibition of calcineurin attenuates cardiac hypertrophy in vivo. Proc Natl Acad Sci USA 2001;98:3322–3327. 86. Bueno OF, Wilkins BJ, Tymitz KM, et al: Impaired cardiac hypertrophic response in calcineurin Abeta-deficient mice. Proc Natl Acad Sci USA 2002;99:4586–4591. 87. Wilkins BJ, De Windt LJ, Bueno OF, et al: Targeted disruption of NFATc3, but not NFATc4, reveals an intrinsic defect in calcineurin-mediated cardiac hypertrophic growth. Mol Cell Biol 2002;22:7603–7613. 88. Fatkin D, McConnell BK, Mudd JO, et al: An abnormal Ca(2+) response in mutant sarcomere protein-mediated familial hypertrophic cardiomyopathy. J Clin Invest 2000;106:1351–1359. 89. Geisterfer-Lowrance AA, Christe M, Conner DA, et al: A mouse model of familial hypertrophic cardiomyopathy. Science 1996; 272:731–734. 90. Zhang T, Johnson EN, Gu Y et al: The cardiac-specific nuclear delta(B) isoform of Ca2+/calmodulin-dependent protein kinase II induces hypertrophy and dilated cardiomyopathy associated with increased protein phosphatase 2A activity. J Biol Chem 2002; 277:1261–1267. 91. Bers DM: Cardiac excitation-contraction coupling. Nature 2002;415:198–205. 92. Mesaeli N, Nakamura K, Opas M, Michalak M: Endoplasmic reticulum in the heart, a forgotten organelle? Mol Cell Biochem 2001; 225:1–6. 93. Allen BG, Katz S: Calreticulin and calsequestrin are differentially distributed in canine heart. J Mol Cell Cardiol 2000;32: 2379–2384. 94. Semsarian C,Ahmad I, Giewat M, et al: The L-type calcium channel inhibitor diltiazem prevents cardiomyopathy in a mouse model. J Clin Invest 2002;109:1013–1020. 95. Mesaeli N, Nakamura K, Zvaritch E et al: Calreticulin is essential for cardiac development. J Cell Biol 1999;144:857–868. 96. Dolmetsch RE, Lewis RS, Goodnow CC, Healy JI: Differential activation of transcription factors induced by Ca2+ response amplitude and duration. Nature 1997;386:855–858. 97. Zhang J, Warwick PJ, Wolfrum E, et al: Saturated output of a Ge. Physical Rev A 1996;54:R4653–R4656. 98. Timmerman LA, Clipstone NA, Ho SN, et al: Rapid shuttling of NFAT in discrimination of Ca2+ signals and immunosuppression. Nature 1996;383:837–840. 99. Lisman J:The CaM kinase II hypothesis for the storage of synaptic memory. Trends Neurosci 194;17:406–412. 100. Bayer KU, De Koninck P, Schulman H: Alternative splicing modulates the frequency-dependent response of CaMKII to Ca(2+) oscillations. EMBO J 2002;21:3590–3597. 101. LaMorte VJ, Thorburn J, Absher D, et al: Gq- and ras-dependent pathways mediate hypertrophy of neonatal rat ventricular myocytes following alpha 1-adrenergic stimulation. J Biol Chem 1994;269:13490–13496. 102. Adams JW, Pagel AL, Means CK et al: Cardiomyocyte apoptosis induced by Galphaq signaling is mediated by permeability transition pore formation and activation of the mitochondrial death pathway. Circ Res 2000;87:1180–1187. 103. Yussman MG,Toyokawa T, Odley A, et al: Mitochondrial death protein Nix is induced in cardiac hypertrophy and triggers apoptotic cardiomyopathy. Nat Med 2002;8:725–730. 104. Akhter SA, Luttrell LM, Rockman HA, et al:Targeting the receptorGq interface to inhibit in vivo pressure overload myocardial hypertrophy. Science 1998;280:574–577. 105. Esposito G, Rapacciuolo A, Naga Prasad SV, et al: Genetic alterations that inhibit in vivo pressure-overload hypertrophy prevent cardiac dysfunction despite increased wall stress. Circulation 2002;105:85–92. 106. Wettschureck N, Rutten H, Zywietz A, et al: Absence of pressure overload induced myocardial hypertrophy after conditional inac-

107. 108.

109. 110. 111. 112. 113. 114.

115. 116. 117. 118. 119. 120. 121.

122.

123. 124. 125.

126.

127. 128.

tivation of Galphaq/Galpha11 in cardiomyocytes. Nat Med 2001;7:1236–1240. Maruyama Y, Nishida M, Sugimoto Y, et al: Galpha(12/13) mediates alpha(1)-adrenergic receptor-induced cardiac hypertrophy. Circ Res 2002;91:961–969. Asakura M, Kitakaze M,Takashima S, et al: Cardiac hypertrophy is inhibited by antagonism of ADAM12 processing of HB-EGF: Metalloproteinase inhibitors as a new therapy. Nat Med 2002; 8:35–40. Nishizuka Y: Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 1992;258:607–614. Dunnmon PM, Iwaki K, Henderson SA, et al: Phorbol esters induce immediate-early genes and activate cardiac gene transcription in neonatal rat myocardial cells. J Mol Cell Cardiol 1990;22:901–910. Bowman JC, Steinberg SF, Jiang T, et al: Expression of protein kinase C beta in the heart causes hypertrophy in adult mice and sudden death in neonates. J Clin Invest 1997;100:2189–2195. Wakasaki H, Koya D, Schoen FJ, et al: Targeted overexpression of protein kinase C beta2 isoform in myocardium causes cardiomyopathy. Proc Natl Acad Sci USA 1997;94:9320–9325. Takeishi Y, Ping P, Bolli R, et al:Transgenic overexpression of constitutively active protein kinase C epsilon causes concentric cardiac hypertrophy. Circ Res 2000;86:1218–1223. Pass JM, Zheng Y, Wead WB, et al: PKCepsilon activation induces dichotomous cardiac phenotypes and modulates PKCepsilonRACK interactions and RACK expression. Am J Physiol Heart Circ Physiol 2001;280:H946–55. Dempsey EC, Newton AC, Mochly-Rosen D, et al: Protein kinase C isozymes and the regulation of diverse cell responses. Am J Physiol Lung Cell Mol Physiol 2000;279:L429–438. Steinberg SF, Goldberg M, Rybin VO: Protein kinase C isoform diversity in the heart. J Mol Cell Cardiol 1995;27:141–153. Mackay K, Mochly-Rosen D: Localization, anchoring, and functions of protein kinase C isozymes in the heart. J Mol Cell Cardiol 2001;33:1301–1307. Malhotra A, Kang BP, Opawumi D, et al: Molecular biology of protein kinase C signaling in cardiac myocytes. Mol Cell Biochem 2001;225:97–107. Dutil EM, Newton AC: Dual role of pseudosubstrate in the coordinated regulation of protein kinase C by phosphorylation and diacylglycerol. J Biol Chem 2000;275:10697–10701. Puceat M, Hilal-Dandan R, Strulovici B, et al: Differential regulation of protein kinase C isoforms in isolated neonatal and adult rat cardiomyocytes. J Biol Chem 1994;269:16938–16944. Clerk A, Bogoyevitch MA, Anderson MB, Sugden PH: Differential activation of protein kinase C isoforms by endothelin-1 and phenylephrine and subsequent stimulation of p42 and p44 mitogen-activated protein kinases in ventricular myocytes cultured from neonatal rat hearts. J Biol Chem 1994;269: 32848–32857. Clerk A, Gillespie-Brown J, Fuller SJ, Sugden PH: Stimulation of phosphatidylinositol hydrolysis, protein kinase C translocation, and mitogen-activated protein kinase activity by bradykinin in rat ventricular myocytes: Dissociation from the hypertrophic response. Biochem J 1996;317(Pt 1):109–118. Clerk A, Sugden PH: Untangling the Web: Specific signaling from PKC isoforms to MAPK cascades. Circ Res 2001;89:847–849. Bueno OF, Molkentin JD: Involvement of extracellular signal-regulated kinases 1/2 in cardiac hypertrophy and cell death. Circ Res 2002;91:776–781. Bogoyevitch MA, Marshall CJ, Sugden PH: Hypertrophic agonists stimulate the activities of the protein kinases c-Raf and A-Raf in cultured ventricular myocytes. J Biol Chem 1995;270: 26303–26310. Heidkamp MC, Bayer AL, Martin JL, Samarel AM: Differential activation of mitogen-activated protein kinase cascades and apoptosis by protein kinase C epsilon and delta in neonatal rat ventricular myocytes. Circ Res 2001;89:882–890. Mochly-Rosen D,Wu G, Hahn H et al: Cardiotrophic effects of protein kinase C epsilon: Analysis by in vivo modulation of PKCepsilon translocation. Circ Res 2000;86:1173–1179. Wu G,Toyokawa T, Hahn H, Dorn GW 2nd: Epsilon protein kinase C in pathological myocardial hypertrophy. Analysis by combined transgenic expression of translocation modifiers and Galphaq. J Biol Chem 2000;275:29927–29930.

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION 129. Jin ZQ, Zhou HZ, Zhu P, et al: Cardioprotection mediated by sphingosine-1-phosphate and ganglioside GM-1 in wild-type and PKC epsilon knockout mouse hearts. Am J Physiol Heart Circ Physiol 2002;282:H1970–1977. 130. Baines CP, Zhang J,Wang GW, et al: Mitochondrial PKCepsilon and MAPK form signaling modules in the murine heart: Enhanced mitochondrial PKCepsilon-MAPK interactions and differential MAPK activation in PKCepsilon-induced cardioprotection. Circ Res 2002;90:390–397. 131. Valks DM, Cook SA, Pham FH et al: Phenylephrine promotes phosphorylation of Bad in cardiac myocytes through the extracellular signal-regulated kinases 1/2 and protein kinase A. J Mol Cell Cardiol 2002;34:749–763. 132. Huang XP, Pi Y, Lokuta AJ, et al: Arachidonic acid stimulates protein kinase C-epsilon redistribution in heart cells. J Cell Sci 1997;110(Pt 14):1625–1634. 133. Csukai M, Chen CH, De Matteis MA, Mochly-Rosen D: The coatomer protein beta′-COP, a selective binding protein (RACK) for protein kinase Cepsilon. J Biol Chem 1997;272:29200–29206. 134. Kuroda S, Tokunaga C, Kiyohara Y, et al: Protein-protein interaction of zinc finger LIM domains with protein kinase C. J Biol Chem 1996;271:31029–31032. 135. Nakagawa N, Hoshijima M, Oyasu M, et al: ENH, containing PDZ and LIM domains, heart/skeletal muscle-specific protein, associates with cytoskeletal proteins through the PDZ domain. Biochem Biophys Res Commun 2000;272:505–512. 136. Zhou Q, Ruiz-Lozano P, Martone ME, Chen J: Cypher, a striated muscle-restricted PDZ and LIM domain-containing protein, binds to alpha-actinin-2 and protein kinase C. J Biol Chem 1999; 274:19807–19813. 137. Faulkner G, Pallavicini A, Formentin E, et al: ZASP: A new Z-band alternatively spliced PDZ-motif protein. J Cell Biol 1999;146: 465–475. 138. Passier R, Richardson JA, Olson EN: Oracle, a novel PDZ-LIM domain protein expressed in heart and skeletal muscle. Mech Dev 2000;92:277–284. 139. 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;92:4862–4866. 140. Kishimoto T, Taga T, Akira S: Cytokine signal transduction. Cell 1994;76:253–262. 141. Kunisada K, Negoro S,Tone E, et al: Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicin- induced cardiomyopathy. Proc Natl Acad Sci USA 2000;97: 315–319. 142. Osugi T, Oshima Y, Fujio Y, et al: Cardiac-specific activation of signal transducer and activator of transcription 3 promotes vascular formation in the heart. J Biol Chem 2002;277:6676–6681. 143. Kodama H, Fukuda K, Pan J, et al: Significance of ERK cascade compared with JAK/STAT and PI3-K pathway in gp130-mediated cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2000; 279:H1635–1644. 144. Nicol RL, Frey N, Pearson G, et al: Activated MEK5 induces serial assembly of sarcomeres and eccentric cardiac hypertrophy. EMBO J 2001;20:2757–2767. 145. Oh H, Fujio Y, Kunisada K, et al:Activation of phosphatidylinositol 3-kinase through glycoprotein 130 induces protein kinase B and p70 S6 kinase phosphorylation in cardiac myocytes. J Biol Chem 1998;273:9703–9710. 146. Wollert KC, Taga T, Saito M, 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;271:9535–9545. 147. Hoshijima M, Sah VP, Wang Y, et al: The low molecular weight GTPase Rho regulates myofibril formation and organization in neonatal rat ventricular myocytes: Involvement of Rho kinase. J Biol Chem 1998;273:7725–7730. 148. Pashmforoush M, Pomies P, Peterson KL, et al:Adult mice deficient in actinin-associated LIM-domain protein reveal a developmental pathway for right ventricular cardiomyopathy. Nat Med 2001; 7:591–597. 149. Bueno OF, De Windt LJ, Tymitz KM, et al: The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J 2000;19:6341–6350.

289

150. Liang Q, Wiese RJ, Bueno OF, et al: The transcription factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes. Mol Cell Biol 2001;21:7460–7469. 151. Liao P, Georgakopoulos D, Kovacs A, et al: The in vivo role of p38 MAP kinases in cardiac remodeling and restrictive cardiomyopathy. Proc Natl Acad Sci USA 2001;98:12283–12288. 152. Liao P, Wang SQ, Wang S, et al: p38 Mitogen-activated protein kinase mediates a negative inotropic effect in cardiac myocytes. Circ Res 2002;90:190–196. 153. Zhang D, Gaussin V, Taffet GE, et al: TAK1 is activated in the myocardium after pressure overload and is sufficient to provoke heart failure in transgenic mice. Nat Med 2000;6:556–563. 154. Wang Y, Su B, Sah VP, et al: Cardiac hypertrophy induced by mitogen-activated protein kinase kinase 7, a specific activator for c-Jun NH2-terminal kinase in ventricular muscle cells. J Biol Chem 1998;273:5423–5426. 155. Sadoshima J, Montagne O, Wang Q, et al: The MEKK1-JNK pathway plays a protective role in pressure overload but does not mediate cardiac hypertrophy. J Clin Invest 2002;110:271–279. 156. Minamino T, Yujiri T, Terada N, et al: MEKK1 is essential for cardiac hypertrophy and dysfunction induced by Gq. Proc Natl Acad Sci USA 2002;99:3866–3871. 157. Petrich BG, Gong X, Lerner DL, et al: c-Jun N-terminal kinase activation mediates downregulation of connexin43 in cardiomyocytes. Circ Res 2002;91:640–647. 158. Kasahara H, Usheva A, Ueyama T, et al: Characterization of homoand heterodimerization of cardiac Csx/Nkx2.5 homeoprotein. J Biol Chem 2001;276:4570–4580. 159. Morin S, Charron F, Robitaille L, Nemer M: GATA-dependent recruitment of MEF2 proteins to target promoters. EMBO J 2000;19:2046–2055. 160. Morin S, Paradis P,Aries A, Nemer M: Serum response factor-GATA ternary complex required for nuclear signaling by a G-proteincoupled receptor. Mol Cell Biol 2001;21:1036–1044. 161. Hiroi Y, Kudoh S, Monzen K, et al: Tbx5 associates with Nkx2-5 and synergistically promotes cardiomyocyte differentiation. Nat Genet 2001;28:276–280. 162. Bruneau BG, Nemer G, Schmitt JP, et al: A murine model of HoltOram syndrome defines roles of the T-box transcription factor Tbx5 in cardiogenesis and disease. Cell 2001;106:709–721. 163. Dai YS, Cserjesi P, Markham BE, Molkentin JD: The transcription factors GATA4 and dHAND physically interact to synergistically activate cardiac gene expression through a p300-dependent mechanism. J Biol Chem 2002;277:24390–24398. 164. Gusterson RJ, Jazrawi E, Adcock IM, Latchman DS: The transcriptional co-activators 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;278:6838–6847. 165. Wang D, Chang PS,Wang Z, et al:Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor. Cell 2001;105:851–862. 166. Shin CH, Liu ZP, Passier R, et al: Modulation of cardiac growth and development by HOP, an unusual homeodomain protein. Cell 2002;110:725–735. 167. Chen F, Kook H, Milewski R, et al: Hop is an unusual homeobox gene that modulates cardiac development. Cell 2002;110: 713–723. 168. Seidman JG, Seidman C: The genetic basis for cardiomyopathy: From mutation identification to mechanistic paradigms. Cell 2001;104:557–567. 169. Towbin JA, Bowles NE: The failing heart. Nature 2002;415: 227–233. 170. Seidman C, Seidman JG: Molecular genetics of inherited cardiomyopathies. In Chien KR, Breslow JL, Leiden JM, et al (eds): Molecular Basis of Cardiovascular Disease. Philadelphia, WB Saunders, 1999, pp 251–263. 171. Bonne G, Carrier L, Richard P, et al: Familial hypertrophic cardiomyopathy: From mutations to functional defects. Circ Res 1998;83:580–593. 172. Bonne G, Carrier L, Bercovici J, et al: Cardiac myosin binding protein-C gene splice acceptor site mutation is associated with familial hypertrophic cardiomyopathy. Nat Genet 1995;11:438–440. 173. Poetter K, Jiang H, Hassanzadeh S, et al: Mutations in either the essential or regulatory light chains of myosin are associated with

290

174. 175. 176. 177.

178. 179.

180. 181. 182.

183.

184. 185. 186. 187. 188. 189.

190. 191. 192. 193. 194. 195. 196.

CARDIAC MUSCLE DISEASES a rare myopathy in human heart and skeletal muscle. Nat Genet 1996;13:63–69. Mogensen J, Klausen IC, Pedersen AK, et al: Alpha-cardiac actin is a novel disease gene in familial hypertrophic cardiomyopathy. J Clin Invest 1999;103:R39–43. Thierfelder L,Watkins H,MacRae C,et al:Alpha-tropomyosin and cardiac troponin T mutations cause familial hypertrophic cardiomyopathy:A disease of the sarcomere. Cell 1994;77: 701–712. Kimura A, Harada H, Park JE, et al: Mutations in the cardiac troponin I gene associated with hypertrophic cardiomyopathy. Nat Genet 1997;16:379–382. Satoh M,Takahashi M, Sakamoto T, et al: Structural analysis of the titin gene in hypertrophic cardiomyopathy: Identification of a novel disease gene. Biochem Biophys Res Commun 1999;262: 411–417. Arber S, Hunter JJ, Ross J Jr, et al: MLP-deficient mice exhibit a disruption of cardiac cytoarchitectural organization, dilated cardiomyopathy, and heart failure. Cell 1997;88:393–403. Knoll R, Hoshijima M, Hoffman HM, et al: The cardiac mechanical stretch sensor machinery involves a Z disc complex that is defective in a subset of human dilated cardiomyopathy. Cell 2002; 111:943–955. Geier C, Perrot A, Ozcelik C, et al. Mutations in the human muscle LIM protein gene in families with hypertrophic cardiomyopathy. Circulation 2003;107:1390–1395. Tanigawa G, Jarcho JA, Kass S, et al: A molecular basis for familial hypertrophic cardiomyopathy: An alpha/beta cardiac myosin heavy chain hybrid gene. Cell 1990;62:991–998. Geisterfer-Lowrance AA, Kass S, Tanigawa G, et al: A molecular basis for familial hypertrophic cardiomyopathy: A beta cardiac myosin heavy chain gene missense mutation. Cell 1990; 62:999–1006. Haghighi K, Kolokathis F, Pater L, et al: Human phospholamban null results in lethal dilated cardiomyopathy revealing a critical difference between mouse and human. J Clin Invest 2003;111: 869–876. Minamisawa S, Sato Y, Tatsuguchi Y, et al: Mutation of the phospholamban promoter associated with hypertrophic cardiomyopathy. Biochem Biophys Res Commun 2003;304:1–4. Kamisago M, Sharma SD, DePalma SR, et al: Mutations in sarcomere protein genes as a cause of dilated cardiomyopathy. N Engl J Med 2000;343:1688–1696. Olson TM, Michels VV, Thibodeau SN, et al: Actin mutations in dilated cardiomyopathy, a heritable form of heart failure. Science 1998;280:750–752. Olson TM, Kishimoto NY, Whitby FG, Michels VV: Mutations that alter the surface charge of alpha-tropomyosin are associated with dilated cardiomyopathy. J Mol Cell Cardiol 2001;33:723–732. Gerull B, Gramlich M,Atherton J, et al: Mutations of TTN, encoding the giant muscle filament titin, cause familial dilated cardiomyopathy. Nat Genet 2002;30:201–204. Towbin JA, Hejtmancik JF, Brink P, et al: X-linked dilated cardiomyopathy. Molecular genetic evidence of linkage to the Duchenne muscular dystrophy (dystrophin) gene at the Xp21 locus. Circulation 1993;87:1854–1865. Muntoni F, Cau M, Ganau A, et al: Brief report: Deletion of the dystrophin muscle-promoter region associated with X-linked dilated cardiomyopathy. N Engl J Med 1993;329:921–925. Melacini P, Fanin M, Danieli GA, et al: Myocardial involvement is very frequent among patients affected with subclinical Becker’s muscular dystrophy. Circulation 1996;94:3168–3175. Barresi R, Di Blasi C, Negri T, et al: Disruption of heart sarcoglycan complex and severe cardiomyopathy caused by beta sarcoglycan mutations. J Med Genet 2000;37:102–107. Tsubata S, Bowles KR,Vatta M, et al: Mutations in the human deltasarcoglycan gene in familial and sporadic dilated cardiomyopathy. J Clin Invest 2000;106:655–662. Ichida F, Tsubata S, Bowles KR, et al: Novel gene mutations in patients with left ventricular noncompaction or Barth syndrome. Circulation 2001;103:1256–1263. Maeda M, Holder E, Lowes B, et al: Dilated cardiomyopathy associated with deficiency of the cytoskeletal protein metavinculin. Circulation 1997;95:17–20. Olson TM, Illenberger S, Kishimoto NY, et al: Metavinculin mutations alter actin interaction in dilated cardiomyopathy. Circulation 2002;105:431–437.

197. Goldfarb LG, Park KY, Cervenakova L, et al: Missense mutations in desmin associated with familial cardiac and skeletal myopathy. Nat Genet 1998;19:402–403. 198. Li D, Tapscoft T, Gonzalez O, et al: Desmin mutation responsible for idiopathic dilated cardiomyopathy. Circulation 1999;100: 461–464. 199. Fatkin D, MacRae C, Sasaki T, et al: Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease. N Engl J Med 1999;341: 1715–1724. 200. DiMauro S, Hirano M: Mitochondria and heart disease. Curr Opin Cardiol 1998;13:190–197. 201. Bione S, D’Adamo P, Maestrini E, et al: A novel X-linked gene, G4.5. is responsible for Barth syndrome. Nat Genet 1996;12: 385–389. 202. Schmitt JP, Kamisago M,Asahi M, et al: Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Science 2003;299:1410–1413. 203. Mogensen J, Kubo T, Duque M, et al: Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin I mutations. J Clin Invest 2003;111:209–216. 204. Hoshijima M, Chien KR: Mixed signals in heart failure: Cancer rules. J Clin Invest 2002;109:849–855. 205. Durbeej M, Campbell KP: Muscular dystrophies involving the dystrophin-glycoprotein complex: An overview of current mouse models. Curr Opin Genet Dev 2002;12:349–361. 206. Ehler E, Horowits R, Zuppinger C, et al:Alterations at the intercalated disk associated with the absence of muscle LIM protein. J Cell Biol 2001;153:763–772. 207. Zolk O, Caroni P, Bohm M: Decreased expression of the cardiac LIM domain protein MLP in chronic human heart failure. Circulation 2000;101:2674–2677. 208. Furukawa T, Ono Y, Tsuchiya H, et al: Specific interaction of the potassium channel beta-subunit minK with the sarcomeric protein T-cap suggests a T-tubule-myofibril linking system. J Mol Biol 2001;313:775–784. 209. Mohler PJ, Schott JJ, Gramolini AO, et al: Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. Nature 2003;421:634–639. 210. Clark KA, McElhinny AS, Beckerle MC, Gregorio CC: Striated muscle cytoarchitecture: An intricate web of form and function. Annu Rev Cell Dev Biol 2002;18:637–706. 211. Frey N, Richardson JA, Olson EN: Calsarcins, a novel family of sarcomeric calcineurin-binding proteins. Proc Natl Acad Sci USA 2000;97:14632–14637. 212. Faulkner G, Pallavicini A, Comelli A, et al: FATZ, a filamin-, actinin-, and telethonin-binding protein of the Z-disc of skeletal muscle. J Biol Chem 2000;275:41234–41242. 213. Frey N, Olson EN: Calsarcin-3, a novel skeletal muscle-specific member of the calsarcin family, interacts with multiple Z-disc proteins. J Biol Chem 2002;277:13998–4004. 214. Tomaselli GF, Marban E: Electrophysiological remodeling in hypertrophy and heart failure. Cardiovasc Res 1999;42:270–283. 215. Hasenfuss G, Pieske B: Calcium cycling in congestive heart failure. J Mol Cell Cardiol 2002;34:951–969. 216. Houser SR, Margulies KB: Is depressed myocyte contractility centrally involved in heart failure? Circ Res 2003;92:350–358. 217. Brittsan AG, Kranias EG: Phospholamban and cardiac contractile function. J Mol Cell Cardiol 2000;32:2131–2139. 218. Tada M,Toyofuku T: Molecular regulation of phospholambin function and expression. Trends in Cardiovascular medicine 1998;8:330–340. 219. Schwinger RH, Munch G, Bolck B, et al: Reduced Ca(2+)-sensitivity of SERCA 2a in failing human myocardium due to reduced serin-16 phospholamban phosphorylation. J Mol Cell Cardiol 1999;31:479–491. 220. Sande JB, Sjaastad I, Hoen IB, et al: Reduced level of serine(16) phosphorylated phospholamban in the failing rat myocardium: A major contributor to reduced SERCA2 activity. Cardiovasc Res 2002;53:382–391. 221. Frank KF, Bolck B, Brixius K, et al: Modulation of SERCA: Implications for the failing human heart. Basic Res Cardiol 2002;97(Suppl 1):I72–78. 222. Rockman HA, Koch WJ, Lefkowitz RJ: Seven-transmembranespanning receptors and heart function. Nature 2002;415: 206–212.

MOLECULAR PATHWAYS FOR CARDIAC HYPERTROPHY AND HEART FAILURE PROGRESSION 223. Lohse MJ, Engelhardt S: Protein kinase a transgenes: The many faces of cAMP. Circ Res 2001;89:938–940. 224. Stevenson LW: Inotropic therapy for heart failure. N Engl J Med 1998;339:1848–1850. 225. Antos CL, Frey N, Marx SO, et al: Dilated cardiomyopathy and sudden death resulting from constitutive activation of protein kinase a. Circ Res 2001;89:997–1004. 226. Bristow MR: beta-adrenergic receptor blockade in chronic heart failure. Circulation 2000;101:558–569. 227. Shah AS, Lilly RE, Kypson AP, et al: Intracoronary adenovirus-mediated delivery and overexpression of the beta(2)-adrenergic receptor in the heart: Prospects for molecular ventricular assistance. Circulation 2000;101:408–414. 228. Tomiyasu K, Oda Y, Nomura M, et al: Direct intra-cardiomuscular transfer of beta2-adrenergic receptor gene augments cardiac output in cardiomyopathic hamsters. Gene Ther 2000;7:2087–2093. 229. Rockman HA, Chien KR, Choi DJ, 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;95:7000–7005. 230. Freeman K, Lerman I, Kranias EG, et al: Alterations in cardiac adrenergic signaling and calcium cycling differentially affect the progression of cardiomyopathy. J Clin Invest 2001;107:967–974. 231. White DC, Hata JA, Shah AS, et al: Preservation of myocardial betaadrenergic receptor signaling delays the development of heart failure after myocardial infarction. Proc Natl Acad Sci USA 2000;97:5428–5433. 232. Shah AS, White DC, Emani S, et al: In vivo ventricular gene delivery of a beta-adrenergic receptor kinase inhibitor to the failing heart reverses cardiac dysfunction. Circulation 2001;103: 1311–1316. 233. Roth DM, Gao MH, Lai NC, et al: Cardiac-directed adenylyl cyclase expression improves heart function in murine cardiomyopathy. Circulation 1999;99:3099–3102. 234. Lai NC, Roth DM, Gao MH, et al: Intracoronary delivery of adenovirus encoding adenylyl cyclase VI increases left-ventricular function and cAMP-generating capacity. Circulation 2000;102: 2396–2401. 235. He H, Giordano FJ, Hilal-Dandan R, et al: Overexpression of the rat sarcoplasmic reticulum Ca2+ ATPase gene in the heart of transgenic mice accelerates calcium transients and cardiac relaxation. J Clin Invest 1997;100:380–389. 236. (unpublished data by Hoshijima M, Minamisawa S, Dillmann WH, Ross J, and Chien KR) 237. Miyamoto MI, del Monte F, Schmidt U, et al: Adenoviral gene transfer of SERCA2a improves left-ventricular function in aorticbanded rats in transition to heart failure. Proc Natl Acad Sci USA 2000;97:793–798. 238. del Monte F,Williams E, Lebeche D, et al: Improvement in survival and cardiac metabolism after gene transfer of sarcoplasmic reticulum Ca(2+)-ATPase in a rat model of heart failure. Circulation 2001;104:1424–1429. 239. Kiriazis H, Kranias EG: Genetically engineered models with alterations in cardiac membrane calcium-handling proteins. Annu Rev Physiol 2000;62:321–351. 240. Minamisawa S, Hoshijima M, Chu G, et al: Chronic phospholamban-sarcoplasmic reticulum calcium ATPase interaction is the critical calcium cycling defect in dilated cardiomyopathy. Cell 1999;99:313–322. 241. Song Q, Schmidt AG, Hahn HS, et al: Rescue of cardiomyocyte dysfunction by phospholamban ablation does not prevent ventricular failure in genetic hypertrophy. J Clin Invest 2003;111:859–867. 242. del Monte F, Harding SE, Dec GW, et al:Targeting phospholamban by gene transfer in human heart failure. Circulation 2002;105: 904–907. 243. Marx SO, Reiken S, Hisamatsu Y et al: PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): Defective regulation in failing hearts. Cell 2000;101: 365–376. 244. Xin HB, Senbonmatsu T, Cheng DS, et al: Oestrogen protects FKBP12.6 null mice from cardiac hypertrophy. Nature 2002; 416:334–338. 245. Reiken S, Gaburjakova M, Gaburjakova J, et al: Beta-adrenergic receptor blockers restore cardiac calcium release channel (ryanodine receptor) structure and function in heart failure. Circulation 2001;104:2843–2848.

291

246. Doi M, Yano M, Kobayashi S, et al: Propranolol prevents the development of heart failure by restoring FKBP12.6-mediated stabilization of ryanodine receptor. Circulation 2002;105:1374–1379. 247. Laitinen PJ, Brown KM, Piippo K, et al: Mutations of the cardiac ryanodine receptor (RyR2) gene in familial polymorphic ventricular tachycardia. Circulation 2001;103:485–490. 248. Priori SG, Napolitano C,Tiso N, et al: Mutations in the cardiac ryanodine receptor gene (hRyR2) underlie catecholaminergic polymorphic ventricular tachycardia. Circulation 2001;103:196–200. 249. Tiso N, Stephan DA, Nava A, et al: Identification of mutations in the cardiac ryanodine receptor gene in families affected with arrhythmogenic right ventricular cardiomyopathy type 2 (ARVD2). Hum Mol Genet 2001;10:189–194. 250. Marks AR: Ryanodine receptors/calcium release channels in heart failure and sudden cardiac death. J Mol Cell Cardiol 2001;33: 615–624. 251. Prestle J, Janssen PM, Janssen AP, et al: Overexpression of FK506binding protein FKBP12.6 in cardiomyocytes reduces ryanodine receptor-mediated Ca(2+) leak from the sarcoplasmic reticulum and increases contractility. Circ Res 2001;88:188–194. 252. Keating MT, Sanguinetti MC: Molecular and cellular mechanisms of cardiac arrhythmias. Cell 2001;104:569–580. 253. Marban E: Cardiac channelopathies. Nature 2002;415:213–218. 254. Gutstein DE, Morley GE, Tamaddon H, et al: Conduction slowing and sudden arrhythmic death in mice with cardiac-restricted inactivation of connexin43. Circ Res 2001;88:333–339. 255. Barker RJ, Gourdie, RG: JNK bond regulation: Why do mammalian hearts invest in connexin43? Circ Res 2002;91:556–558. 256. Nguyen-Tran VT, Kubalak SW, Minamisawa S, et al:A novel genetic pathway for sudden cardiac death via defects in the transition between ventricular and conduction system cell lineages. Cell 2000;102:671–682. 257. Schott JJ, Benson DW, Basson CT, et al: Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science 1998;281:108–111. 258. Gollob MH, Green MS, Tang AS, et al: Identification of a gene responsible for familial Wolff-Parkinson-White syndrome. N Engl J Med 2001;344:1823–1831. 259. Blair E, Redwood C,Ashrafian H, et al: Mutations in the gamma(2) subunit of AMP-activated protein kinase cause familial hypertrophic cardiomyopathy: Evidence for the central role of energy compromise in disease pathogenesis. Hum Mol Genet 2001;10: 1215–1220. 260. Arad M, Benson DW, Perez-Atayde AR, et al: Constitutively active AMP kinase mutations cause glycogen storage disease mimicking hypertrophic cardiomyopathy. J Clin Invest 2002;109:357–362. 261. Kuo HC, Cheng CF, Clark RB, et al:A defect in the Kv channel-interacting protein 2 (KChIP2) gene leads to a complete loss of I(to) and confers susceptibility to ventricular tachycardia. Cell 2001;107:801–813. 262. Rosati B, Pan Z, Lypen S, et al: Regulation of KChIP2 potassium channel beta subunit gene expression underlies the gradient of transient outward current in canine and human ventricle. J Physiol 2001;533:119–125. 263. Rosati B, Grau F, Rodriguez S, et al: Concordant expression of KChIP2 mRNA, protein and transient outward current throughout the canine ventricle. J Physiol 2003;548:815–822.

EDITOR’S CHOICE Intravenous nesiritide vs nitroglycerin for treatment of decompensated congestive heart failure: A randomized controlled trial. Jama 2002;287:1531–1540. BNP finds a home as a therapy for acute, decompensated heart failure. Chien KR, Ross J Jr, Hoshijima M: Calcium and heart failure:The cycle game. Nat Med 2003;9:508–509. Promoting calcium cycling via phospholamban inhibition is new therapeutic target to halt heart failure progression. del Monte F, Hajjar RJ:Targeting calcium cycling proteins in heart failure through gene transfer. J Physiol 2003;546:49–61. Overview of gene therapy strategies for heart failure via promting calcium cycling.

292

CARDIAC MUSCLE DISEASES

Haghighi K, Kolokathis F, Pater L, et al: Human phospholamban null results in lethal dilated cardiomyopathy revealing a critical difference between mouse and human. J Clin Invest 2003;111:869–876. Human mutation results in a truncated phospholamban peptide and is associated with genetically base dilated cardiomyopathy, contains cytoplasmic inhibitory domain but also is unstable, may represent a mixed genetic effect of poison peptide and hypomorphic allele. Hajjar RJ, MacRae CA: Adrenergic-receptor polymorphisms and heart failure. N Engl J Med 2002;347:1196–1199. Nice review of the association of human variants of the beta-adrenergic receptor and heart failure. MacLennan DH, Kranias EG: Phospholamban: A crucial regulator of cardiac contractility. Nat Rev Mol Cell Biol 2003;4:566–577. Excellent review of emerging topic of clinical importance. Maisel AS, Krishnaswamy P, Nowak RM, et al: Rapid measurement of Btype natriuretic peptide in the emergency diagnosis of heart failure. N Engl J Med 2002;347:161–167. BNP as a biomarker for heart failure; rapidly finding a place as standard diagnostic test in the ER and elsewhere. Olson EN, Schneider MD: Sizing up the heart: Development redux in disease. Genes Dev 2003;17:1937–1956. Thought leaders provide excellent overview of complex field. Prasad SV, Perrino C, Rockman HA: Role of phosphoinositide 3-kinase in cardiac function and heart failure. Trends Cardiovasc Med 2003;13:206–212. Pl-3 pathways may represent a new signaling pathway to regulate cardiac contractility.

Schmitt JP, Kamisago M, Asahi M, et al: Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Science 2003;299:1410–1413. Seminal paper provides first clear genetic evidence that the augmentation of phospholamban inhibitory activity is sufficient by itself to cause human dilated cardiomyopathy and heart failure; human validation of earlier studies in mice that suggested that phospholamban is a valid therapeutic target for heart failure. Small KM, Wagoner LE, Levin AM, et al: Synergistic polymorphisms of beta1-and alpha2C-adrenergic receptors and the risk of congestive heart failure. N Engl J Med 2002;347:1135–1142. Genetic susceptibility to heart failure based on variants of adrenergic receptors. Wehrens XH, Marks AR: Myocardial disease in failing hearts: Defective excitation-contraction coupling. Cold Spring Harb Symp Quant Biol 2002;67:533–541. Leader in the molecular biology of ryanodine receptors provides nice overview of large body of work. Wencker D, Chandra M, Nguyen K, et al:A mechanistic role for cardiac myocyte apoptosis in heart failure. J Clin Invest 2003;111:1497–1504. Makes the case for a pivotal role of apoptosis in heart failure progression. Yussman MG, Toyokawa T, Odley A, et al: Mitochondrial death protein Nix is induced in cardiac hypertrophy and triggers apoptotic cardiomyopathy. Nat Med 2002;8:725–730. New pathway for cell death in heart muscle cells could play important role in heart failure progression during chronic stimulation of receptors linked to Gq pathways.