Skip main navigation

Hypertrophic Cardiomyopathy

Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy
Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.117.311059Circulation Research. 2017;121:749–770

    Abstract

    Hypertrophic cardiomyopathy (HCM) is a genetic disorder that is characterized by left ventricular hypertrophy unexplained by secondary causes and a nondilated left ventricle with preserved or increased ejection fraction. It is commonly asymmetrical with the most severe hypertrophy involving the basal interventricular septum. Left ventricular outflow tract obstruction is present at rest in about one third of the patients and can be provoked in another third. The histological features of HCM include myocyte hypertrophy and disarray, as well as interstitial fibrosis. The hypertrophy is also frequently associated with left ventricular diastolic dysfunction. In the majority of patients, HCM has a relatively benign course. However, HCM is also an important cause of sudden cardiac death, particularly in adolescents and young adults. Nonsustained ventricular tachycardia, syncope, a family history of sudden cardiac death, and severe cardiac hypertrophy are major risk factors for sudden cardiac death. This complication can usually be averted by implantation of a cardioverter-defibrillator in appropriate high-risk patients. Atrial fibrillation is also a common complication and is not well tolerated. Mutations in over a dozen genes encoding sarcomere-associated proteins cause HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin-binding protein C, respectively, are the 2 most common genes involved, together accounting for ≈50% of the HCM families. In ≈40% of HCM patients, the causal genes remain to be identified. Mutations in genes responsible for storage diseases also cause a phenotype resembling HCM (genocopy or phenocopy). The routine applications of genetic testing and preclinical identification of family members represents an important advance. The genetic discoveries have enhanced understanding of the molecular pathogenesis of HCM and have stimulated efforts designed to identify new therapeutic agents.

    Definition and Epidemiology

    Hypertrophic cardiomyopathy (HCM) is a genetic disorder of cardiac myocytes that is characterized by cardiac hypertrophy, unexplained by the loading conditions; a nondilated left ventricle; and a normal or increased ejection fraction. Cardiac hypertrophy is usually asymmetrical with greatest involvement most commonly of the basal interventricular septum subjacent to the aortic valve. It is occasionally restricted to other myocardial regions, such as the apex, the midportion, and the posterior wall of the left ventricle. At the cellular level, cardiac myocytes are hypertrophied, disorganized, and separated by areas of interstitial fibrosis.

    HCM is a disorder without a distinct geographic, ethnic, or sex pattern of distribution. Prevalence of HCM has been estimated at 0.16% to 0.29% (≈ 1:625–1:344 individuals) in the general adult population.14 In adults, HCM may be diagnosed by the presence of left ventricular end diastolic wall thickness >13 mm on an echocardiogram or other imaging technique.1 The European Society of Cardiology guidelines recommend using a left ventricular wall thickness of ≥15 mm in the diagnostic criteria.5 Estimating the prevalence of HCM based on detection of cardiac hypertrophy, athough clinically valuable, has many limitations. Notable among them is the age-dependent expression of cardiac hypertrophy; the latter is present in approximately one half of patients with the underlying causal mutations by the third decade of life and in approximately three fourths by the sixth decade.6,7 Given the age-dependent expression of HCM mutations, its prevalence is expected to be higher in older subjects. Indeed, HCM has been recognized in 0.29% (1:333) of 60-year-old individuals undergoing echocardiography for cardiovascular evaluation in the United States.2 A similar prevalence has been reported in other parts of the world.3,4 When more sensitive imaging methods are used, when more family members are evaluated, and when genetic testing is more widely used (see below), a much higher estimate of 0.6% (1:167) has been suggested.810

    Cardiac hypertrophy may be of late onset, and <13 mm, the diagnostic cut point for the diagnosis of HCM.7,11 Hence, HCM may be underdiagnosed in such individuals. Conversely, cardiac hypertrophy could result from the phenocopy conditions (see below), which might account for 5% to 10% of the clinically diagnosed HCM cases in children.1214 Moreover, the presence of concomitant conditions that may cause myocardial hypertrophy, such as arterial hypertension or aortic stenosis, may make the differentiation of primary (HCM) from secondary hypertrophy challenging. However, the asymmetrical shape of the ventricle with greatest hypertrophy of the basal interventricular septum and genetic testing of the subjects and their families (see below) may be helpful in this population.

    Molecular Genetic Basis

    HCM is an archetypical single gene disorder with an autosomal dominant pattern of inheritance,15 whereby a single mutation is usually sufficient to cause the disease, albeit with variable penetrance and expression. The variability of the phenotype is due, at least in part, to the causal mutation acting in concert with many other genetic and of nongenetic influences. Approximately 60% of patients with HCM have a clearly recognizable familial disease. Autosomal recessive and X-linked modes of inheritance have been described but are rare.16,17 An X-linked inheritance typically raises the possibility of a phenocopy condition, such as Fabry disease.18 A phenocopy condition also occurs in syndromic conditions, such as the Noonan syndrome and in storage diseases, such as Anderson–Fabry disease.1820 The phenocopy (genocopy) conditions are not discussed.

    Pioneering studies by Christine and Jonathan Seidman have led to partial elucidation of the molecular genetic basis of HCM. The discovery of the p.Arg403Glu mutation in the MYH7 gene, encoding sarcomere protein MYH7(β-myosin heavy chain) in the French-Canadian family described by Pare et al,21 paved the way for important subsequent discoveries.22 The identification of multiple separate mutations in the principal causal genes, all encoding sarcomere proteins (Figure 1), established HCM as a genetically heterogeneous disease. Among the known causal genes, MYH7 and MYBPC3 (myosin-binding protein C) are the 2 most common, together being responsible for approximately half of the patients with familial HCM.2326 Mutations TNNT2, TNNI3 (cardiac troponin I), and TPM1 (α-tropomyosin) are relatively uncommon causes of HCM and together are responsible for <10% of cases.2528 Mutations in ACTC1 (cardiac α-actin), MYL2 (myosin light chain 2), MYL3 (myosin light chain 3), and CSRP3 (cysteine and glycine-rich protein 3) are also established, albeit uncommon, causes of HCM.2931 Evidence for the causal role of the above 9 genes in HCM is the strongest (Table 1).32,33

    Figure 1.

    Figure 1. Hypertrophic cardiomyopathy (HCM) as a disease of sarcomere proteins. A schematic structure of a sarcomere composed of thick and thin filaments and Z discs is depicted along with its protein constituents involved in HCM. Established causal genes for HCM and their population frequencies are listed.

    Table 1. Genes Implicated in HCM

    Gene Protein Function Tolerance to Variation
    Missense (Z Score) LoF (pLI)
    Established causal gene HCM (large families)
    MYH7 β-Myosin heavy chain ATPase activity, force generation 6.54 0.00
    MYBPC3 Myosin-binding protein C Cardiac contraction 0.69 0.00
    TNNT2 Cardiac troponin T Regulator of actomyosin interaction 1.54 0.01
    TNNI3 Cardiac troponin I Inhibitor of actomyosin interaction 1.88 0.17
    TPM1 α-Tropomyosin Places the troponin complex on cardiac actin 3.42 0.80
    ACTC1 Cardiac α-actin Actomyosin interaction 5.25 0.95
    MYL2 Regulatory myosin light chain Myosin heavy chain 7–binding protein 0.86 0.02
    MYL3 Essential myosin light chain Myosin heavy chain 7–binding protein 0.75 0.89
    CSRP3 Cysteine- and glycine-rich protein 3 Muscle LIM protein (MLP), a Z disk protein −0.66 0.00
    Likely causal genes for HCM (small families)
    FHL1 Four-and-a-half LIM domains 1 Muscle development and hypertrophy 1.29 0.92
    MYOZ2 Myozenin 2 (calsarcin 1) Z disk protein 0.03 0.02
    PLN Phospholamban Regulator of sarcoplasmic reticulum calcium 0.57 0.11
    TCAP Tcap (telethonin) Titin capping protein 0.45 0.08
    TRIM63 Muscle ring finger protein 1 E3 ligase of proteasome ubiquitin system 0.02 0.00
    TTN Titin Sarcomere function −5.48 0.00
    Genes associated with HCM (small families and sporadic cases)
    ACTN2 Actinin, α2 Z disk protein 1.76 1.0
    ANKRD1 Ankyrin repeat domain 1 A negative regulator of cardiac genes −0.01 0.00
    CASQ2 Calsequestrin 2 Calcium-binding protein −1.08 0.00
    CAV3 Caveolin 3 A caveolae protein 1.19 0.34
    JPH2 Junctophilin-2 Intracellular calcium signaling 3.93 0.01
    LDB3 Lim domain binding 3 Z disk protein 0.32 0.00
    MYH6 Myosin heavy chain α Sarcomere protein expressed at low levels in the adult human heart 2.87 0.00
    MYLK2 Myosin light chain kinase 2 Phosphorylate myosin light chain 2 0.73 0.22
    NEXN Nexilin Z disc protein −1.32 0.00
    TNNC1 Cardiac troponin C Calcium-sensitive regulator of myofilament function 2.22 0.51
    VCL Vinculin Z disk protein 3.11 0.99

    The Z score for each gene reflects deviation of the observed variants in the ExAC database from the expected number. A higher positive Z score indicates that the gene is intolerant to variation. Likewise, pLI indicates probability of intolerance to LoF variants with 1 indicating total intolerance. HCM indicates hypertrophic cardiomyopathy; and LoF, loss of function.

    Mutations in TTN (titin), TCAP (telethonin), MYOZ2 (myozenin 2), TRIM63 (ubiquitin E3 ligase tripartite motif protein 63 or MuRF1), and FHL1 (four-and-a-half LIM domains 1) also have been implicated as causes of HCM but occur typically in sporadic cases and small families.3441FHL1 is located on the X chromosome, and mutations might affect hemizygous men disproportionately. Finally, mutations in TNNC1 (cardiac troponin C), MYH6 (myosin heavy chain or α-myosin heavy chain), PLN (phospholamban), CAV3 (caveolin 3), ALPK3 (α kinase 3), and JPH2 (junctophilin-2) have also been reported in patients with HCM,4247 but their causal role in HCM is less certain and has not been established unambiguously (see below).

    A subset of pathogenic variants that exert large phenotypic effects, exhibit high penetrance, and cosegregate with the phenotype in large HCM families are considered the causal mutations.4850 Causality of such mutations is robustly established through cosegregation and linkage analysis. This is the case for the common HCM genes, such as MYH7 and MYBPC3, whose causal role in HCM is unambiguous. Nevertheless, not all genetic variants in the established causal genes for HCM actually cause HCM.50 The ExAc database show that MYH7, MYBPC3, TNNT2, TNNI3, and TPM1 are highly constrained and not tolerant to missense and loss-of-function (LoF) genetic variants (http://exac.broadinstitute.org/). This means that missense and LoF variants in these genes are rare in the general population but not totally absent. Therefore, not all missense and LoF variants in the above genes cause HCM. In contrast, ACTC1 is exceedingly intolerant to missense and LoF variants, as no LoF variant in the ACTC1 gene was observed in ≈ 60 000 unrelated individuals (http://exac.broadinstitute.org/). Consequently, missense and LoF variants in the ACTC1 gene typically cause HCM (or dilated cardiomyopathy).

    Genetic variants with smaller phenotypic effects show incomplete penetrance. Penetrance and phenotypic effects of such variants depend on the presence of other genetic and environmental factors. Many low-to-moderate penetrance genetic variants are found in patients with sporadic HCM and in small families with HCM. Establishing the causal role of such variants in HCM is challenging. Factors that contribute to difficulty in ascertaining causality include human genetic diversity, population-specific frequency of the variants, and the presence of several thousand pathogenic coding variants in each exome, which collectively make the distinction between a causal and incidental variant difficult.4953

    HCM is caused by rare mutations. These mutations typically affect domains in genes that code for sarcomere and sarcomere-associated proteins. Accordingly, the prevalence of specific mutations in an HCM population is low. Two notable exceptions are the p.Arg502Trp mutation in MYBPC3, which has been reported to occur in ≈ 1.5% to 3% of HCM patients5456 and the MYBPC3 mutation p.Val762Asp, which has been identified in 3.9% of the Japanese population.57 Almost all other mutations occur at a frequency of <1% in the HCM population and approximately half are found in a single proband or family.56 The high frequencies of these mutations might reflect hot spots for mutations or likely a founder effect.55 The high frequency is not a feature of mutations occurring in genetically independent HCM populations and might be restricted to the reported study populations. Accordingly, with the possible exception of the above 2 mutations, there is no clear hot spot for mutation in any of the known genes, despite the initial reports.5860

    In general, there is no clear distinction between the location of the mutations in the domain of the encoded proteins and the HCM phenotype. However, mutations in the MYH7 gene show a strong predilection toward the globular head and hinge region of the myosin heavy chain protein, albeit mutations causing changes in the rod domain have also been described.6164 Phenotypically, mutations in the converter domain and a flat surface area in the globular head of MYH7 have been associated with an earlier disease onset.64 Overall, MYH7 and MYBPC3 are not only the 2 most common causal genes in the usual form of HCM with involvement of the basal septum but are also the 2 most common genes in apical HCM (see below).65,66

    The majority of the causal mutations in HCM are missense mutations with the exception of the mutations in MYBPC3, which exhibit a predilection to insertion/deletion and premature truncation mutations because of a frameshift.23,25,26 The missense mutation may alter protein structure and function by changing the amino acid composition of the encoding protein. The insertion/deletion mutations, which induce a frameshift in the encoded protein, are commonly targeted for degradation by the nonsense-mediated decay. Likewise, the premature truncated proteins are subsequently degraded by the ubiquitin-proteasome system, leading to haploinsufficiency (see below). Rare deletion mutations in MYH7, TNNT2, and others also have been reported.61,62

    Missing Causal Genes

    The difficulty in identifying the remaining causal genes, which are sometimes referred to as the missing causal genes, is typically relevant to HCM occurring in sporadic cases or in small families. The missing causal gene in HCM might be in part because of the difficulty in ascertaining the causality of the genetic variants, in an unambiguous manner, in the sporadic cases and small families.50

    In general, genetic variants exert a gradient of effect sizes, ranging from large and causal to clinically negligible.48,49,67 As described earlier, genetic variants that exert large effect sizes are considered to be mutations responsible for autosomal dominant single gene disorders, including HCM, in large families. Such mutations have been mapped and identified through robust genetic approaches, such as cosegregation and linkage analyses. Examples include genetic variants in the MYH7 and MYBPC3 genes. On the other end of the spectrum of effect sizes are the genetic variants that have clinically small or indiscernible effects. Such variants, whenever functional, influence the phenotype, albeit modestly, but do not cause monogenic disorders. In the center of the spectrum is a subset of genetic variants that exert intermediary to large effect sizes and exhibit incomplete penetrance, that is, their effects are influenced by other genetic and nongenetic factors. It is speculated that such variants cause HCM but with incomplete or low penetrance. In such situations, there is no clear cosegregation of the genetic variant with the phenotype and hence, linkage cannot be robustly established, particularly when the family size is small and obviously in the sporadic cases. The difficulty is in part because of the plethora of variants in each genome.

    An approach to identify the missing causal genes is via large-scale association studies followed by analysis of rare variants. The demonstration of an association between a candidate gene and the HCM phenotype in a discovery population is considered provisional (ie, hypothesis-generating) and requires testing for replication in an independent population.

    Possible Digenic/Oligogenic Pathogeneis of HCM

    A subset of HCM patients, ≈5%, exhibits 2 (digenic) or more (oligogenic) causal mutations in the same gene or causal mutations in different genes.26,56,57,6874 The severity of ventricular hypertrophy in subjects with such mutations seems to be more pronounced.57,6871 Double mutations have been limited primarily to variants in well-known HCM genes identified in members of small families. Consequently, it has been difficult to show cosegregation and to establish unambiguously the causal role of each variant. Nevertheless, these observations raise the intriguing possibility that the missing causal genes may be explained, in part, because of the digenic or oligogenic nature in some patients with HCM. Thus, a subset of patients with HCM may not fit into the classic definition of a single gene disorder. In addition, these findings shift the focus from genetic causality of a single dominant mutation to identification of the pathogenic mutations in sporadic HCM and HCM occurring in small families. There are inadequate data to determine differences in the phenotypic expression of HCM caused by 2 mutations on a single gene and 1 mutation on each of 2 causal genes.

    Pathogenesis

    A diverse array of mechanisms, mirroring the diversity of the causal genes and mutations, are implicated in the pathogenesis of HCM (Figure 2). The mechanistic events in HCM might be categorized into 4 sets of interlocking mechanisms. The primary defect is the mutation. Initial or proximal phenotypes are defined as those resulting from the direct effects of the mutations on the structure and function of the sarcomere proteins. The intermediary (or secondary) phenotypes include the molecular changes that occur in response to the changes in the sarcomere protein structure and function. Examples of the latter include altered gene expression and activation of the signaling pathways, such as the MAPK and TGFB1 pathways. The tertiary effects are the ensuing histological and pathological phenotypes, which are the consequence of perturbation of a myriad of secondary molecular events in the myocardium, such as activation of the hypertrophic signaling pathways. These molecular and histological changes lead to the clinical phenotypes of HCM (quaternary). It is important to note that there is a mechanistic distinction between cases of HCM caused by sarcomere protein mutation and the phenocopy conditions because ventricular hypertrophy in the latter may, at least in part, result from storage of material, such as glycogen, and in part because of functional defects in myocytes, such as impaired contraction.

    Figure 2.

    Figure 2. Pathogenesis of hypertrophic cardiomyopathy (HCM). The primary defect is the mutation in the sarcomere, composed of thick and thin filaments and the Z disks. A change in the amino acid sequence in the sarcomere protein or the deficiency of a sarcomere protein (the primary defect) instigates a series of initial (or proximal) defects, such as altered levels of the sarcomere protein, calcium sensitivity, or ATPase activity. These initial defects activate expression of a series of intermediary molecular or secondary changes, such as altered transcriptomics or signaling pathways. The latter set of the molecular changes induce histological and morphological changes in the myocardium, such as myocyte hypertrophy and fibrosis, which could be considered tertiary phenotypes. These molecular and histological changes lead to the clinical or quaternary phenotypes of HCM, such as cardiac arrhythmias and heart failure.

    Initial (Proximal) Defects in HCM

    The initial defects in HCM, in accord with the diversity of HCM mutations, are also diverse. The mutations induce a set of initial changes, such as altered transcription rate and translation efficiency, changes in structure of the affected sarcomere protein, and sarcomere functions. These changes might be considered to be the inciting molecular events as they are the direct effects of the mutations.

    Effects of Mutations on Transcription and Translation

    In autosomal dominant HCM, a small fraction of the HCM mutations result in premature truncation of the encoded proteins because of a gain of a stop codon (such as the p.Gln425X mutation in the MYBPC3 gene11) or because of a frameshift (such as the c.2864-2865delCT in the MYBPC3 gene).75 A number of transcriptional surveillance and quality control mechanisms target the transcripts containing premature termination codon. Mechanisms such as the nonsense-mediated decay pathway identify the premature termination codon, releases the elongation factors (a set of proteins involved in adding amino acids during protein synthesis) from the template, and recruits the decay-inducing complex (a complex of proteins involved in mRNA degradation).76,77 The net effect is degradation of such transcripts and reduced protein levels. In the case of transcripts lacking a naturally occurring stop codon, the mRNA surveillance mechanism referred to as nonstop decay pathway identifies and releases the transcripts from ribosomes to prevent their translation.78,79 This is followed by degradation of the released transcripts by the exosome complex. Another mechanism is activation of the No-Go mRNA decay pathway, a mechanism that stalls progression of ribosomes during translation and results in endonucleolytic cleavage of the mRNA transcript near the stalled site.77 Collectively, these quality control mechanisms prevent synthesis of the truncated proteins.

    Gain- or loss-of-stop-codon mutations or frameshift mutations commonly abolish protein expression. In the heterozygous state, only the wild-type allele is expressed. The net effect, therefore, is haploinsufficiency, which is an uncommon mechanism in HCM, as the majority of the HCM mutations are missense mutations. Mutations in the MYBPC3 gene, however, are predominantly stop-codon and frameshift mutations.26,80,81 Thus, compared with other HCM-causing genes, haploinsufficiency is a more common mechanism in HCM caused by the MYBPC3 mutations.23,25,26,75,8284

    Transcripts containing premature termination codon that escape the nonsense-mediated decay are expected to be expressed as truncated proteins. However, premature truncated proteins are expressed at low levels and are not commonly detectable by antibody-based techniques.75,83,85 This suggests that such proteins are likely to be unstable, misfolded, targeted by unfolded protein responses, and degraded by the ubiquitin-proteasome system. Experimental data in mouse models implicate this system in degradation of truncated MYBPC3 protein.86,87

    In principle, both alleles of the causal gene carrying the heterozygous missense mutation are transcribed and translated into the corresponding wild-type and mutant proteins. However, efficiency of transcription and translation of the wild-type and mutant alleles may vary, generally being less for the mutant allele, because of suboptimal codon usage compared with the wild-type allele.8890 Such allelic imbalance is partially corrected by compensation from the wild-type allele, presumably to maintain the fixed stoichiometry of the sarcomere proteins. However, allelic compensation is often less than complete. Therefore, the mutation might reduce expression level of the corresponding protein and result in a mild deficiency of the involved protein.88,89 Considerable myocyte-to-myocyte variability in the transcript levels of mutant alleles in HCM has been detected, which may explain variability in the myocyte function.89 Overall, variability in the transcription efficiency of the 2 alleles is in accord with the complexity of transcriptional regulation, which depends not only on the composition of the nucleotides but also on the sequence of adjacent regulatory elements.

    Incorporation of the Mutant Protein Into Sarcomere

    Missense mutations that impart structural changes in the encoded protein may reduce efficiency of sarcomere assembly.91 Efficiency of incorporation of the mutant proteins containing missense mutations into sarcomeres has not been adequately determined. Analysis of the ratio of mutant to wild-type MYBPC3 protein in the myofilament, as a surrogate for efficiency of incorporation, show considerable variability, with the fraction of mutant protein comprising 30% to 80% of the total MYBPC3 protein in the myofilaments.90 Variability in the expression levels and incorporation of the mutant proteins into sarcomeres and myofilaments is expected to exert variable functional defects, differing among various mutations and individual myocytes carrying the same genetic mutation. Such heterogeneity in the expression and incorporation of the mutant proteins at the myocyte level might in part explain the variable phenotypic expression of HCM.

    Effects on Myofilament Function

    On incorporation into sarcomeres, the mutant proteins exert a diverse array of functional effects. Mutations could affect various components of the actomyosin cross-bridge cycling, ranging from Ca++ sensitivity of the troponin complex to the generation of power stroke. The latter is defined as bending of the myosin globular head at the hinge region and the ensuing displacement of the actin filament, which results in the force of contraction. Consequently, mutations could affect generation of force by a diverse array of functional defects, including altered Ca++ sensitivity and ATPase activity, which are interdependent.

    Given the limitations of the in vitro studies to properly recapitulate the in vivo conditions, transgenic, knockout, and knockin mouse models have been generated to delineate the mechanism(s) involved in the pathogenesis of HCM.9299 From the outset, it became clear that functional studies of HCM mutations in these models were compounded by the differences in sarcomere protein compositions between them and humans. As noted earlier, HCM mutations affect MYH7, which is the predominant myosin heavy chain isoform (>90%) in the human heart.100102 This contrasts with the murine heart, in which the predominant myosin isoform is MYH6 or α-myosin heavy chain. Consequently, to study the human MYH7 mutation, it had to be transposed into MYH6 in the mouse models. The problem with this approach is the presence of major differences between MYH6 and MYH7 proteins in their ATPase activities and actomyosin kinetics.103 The ATPase activity of the MYH6 protein and the velocity of actin displacement are several folds higher than those of MYH7 protein.100,104,105 The differences in these key biological aspects confound extrapolating the findings in model organisms that predominantly express MYH6 to human HCM.

    Considering the above differences in the compositions of sarcomere proteins, studies performed in hearts that predominantly express MYH7 rather than MYH6, particularly the human cardiac muscle bundles, offer findings that are more relevant to the functional consequences of HCM mutations. These studies suggest that HCM mutations reduce sensitivity of the actinmyosin complex dissociation in response to ATP. Therefore, at any given moment, more myosin and actin molecules are in the bound than in the dissociated state. Experimental data show less maximal tension development per unit of ATP hydrolyzed and, hence, reduced efficiency of force generation.106109 There is, however, considerable variability not only among mutations in different causal genes but also among mutations in the same gene.106111 For example, mutations in MYH7 and MYBPC3 negatively affect force generation but those in MYH7 reduce myofibrillar ATPase activity more than those in MYBPC3.106,107 Accordingly, the energy cost of tension generation is significantly higher for MYH7 than MYBPC3 mutations.107 These functional alterations seem to be the direct effects of the mutations, as myocardial efficiency is also reduced in individuals who carry the causal mutations and do not show cardiac hypertrophy.107 Similarly, experimental data in transgenic rabbits, which predominantly express MYH7, as the human heart, show that expression of the mutant MYH7 p.Arg403Gln protein reduces Ca++ sensitivity of the myofibrillar ATPase activity, which occurs early and precedes the development of cardiac hypertrophy.109 At the clinical level, the increased cost of tension generation correlates with reduced ratios of cardiac phosphocreatine to ATP in the heart in HCM.112

    Findings in murine models also show reduced Ca++ activated myofibrillar force generation and increased cost of tension generation.94,106,110,111,113 As observed in human cardiac tissues, studies in the murine models show significant variability in the functional defects.99,109,114 The reduction of Ca++ sensitivity is not uniform, and the magnitude of myofibrillar efficiency of force generation varies among causal genes and mutations. Single cell studies also point to the presence of considerable myocyte-to-myocyte variability not only at the level of the mutant transcripts but also in the Ca++ sensitivity, influencing maximal force generation.89 There is also considerable regional variability in gene expression and function, suggesting a mosaic molecular and functional phenotype in HCM.115

    Thin filament mutations.

    In contrast to HCM mutations in the thick filaments, mutations in the thin filaments enhance Ca++ sensitivity of myofibrillar ATPase activity and hence of force generation.110,111,114,116 These changes seem to be the direct effects of the mutations, as they precede the development of cardiac hypertrophy, at least in the murine models.98 Changes occurring after the development of cardiac hypertrophy, whether in the human or murine heart, are subject to secondary modifications of the sarcomere proteins, including phosphorylation of TNNI3 and MYBPC3, post-translational modifications that are known to affect sarcomere functions.116,117

    Secondary (Intermediary) Molecular Events

    As discussed above, the initial defects, imparted by the various causal mutations on sarcomere structure and functions, instigate a cascade of secondary (intermediary) molecular events. These include activation of the Ca++-sensitive and stress-responsive molecular pathways that collectively mediate programming of cardiac hypertrophy and induce the morphological and histological (tertiary) phenotypes that are recognized as HCM. In general, the intermediary molecular events involve the pathways that are also activated in other forms of cardiac hypertrophic responses, such as pressure overload–induced cardiac hypertrophy. Accordingly, a multitude of pathways are perturbed, including expression and activation of trophic and mitotic factors, such as calcineurin, mitogen-activated protein kinases, and transforming growth factor β pathways, and noncoding RNA and epigenetic factors.40,118127 The characteristics of the molecular responses, although generally similar, vary according to the biological functions of genes carrying the causal mutations.125,128,129 Nevertheless, cardiac hypertrophy, interstitial fibrosis, and myocyte disarray are considered to be consequences to activation of the intermediary molecules and pathways.130,131

    Tertiary (Histological) and Quaternary (Clinical) Phenotypes

    Histological and morphological phenotypes of HCM, including myocyte and cardiac hypertrophy, disarray, and interstitial fibrosis, among others, are the consequence of intermediary molecular changes in the heart. Histological and morphological phenotypes, discussed under cardiac pathology, produce the clinical manifestations of HCM, including heart failure and cardiac arrhythmias, which are also discussed later.

    Determinants of the HCM Phenotypes

    The morphological, histological, and clinical phenotypes of HCM are the consequence of complex interactions among a large number of determinants, ranging from the causal genetic mutation to environmental factors. The causal mutation is the prerequisite and a major determinant of the phenotype. In addition, the histological and clinical phenotypes is also influenced by genetic backgrounds, which include the presence of additional pathogenic variants in pathways implicated in cardiac hypertrophy, epigenetic factors including noncoding RNAs, post-translational protein modifications, and environmental factors (Figure 3). Thus, the tertiary and quaternary phenotypes of HCM are complex traits, influenced by a large number of determinants, each exerting a small effect, with the causal mutations imparting the largest effects.

    Figure 3.

    Figure 3. Determinants of phenotype in hypertrophic cardiomyopathy (HCM). Selected factors contributing to expression of cardiac phenotype in HCM are shown. The causal mutation imparts the main effect and several others, such as other pathogenic genetic variants (modifiers), genomics (such as noncoding RNAs), proteomics (such as post-translational modifications), and environmental factors (such as isometric exercises) contributing to expression of the phenotype.

    Initial genotype–phenotype correlation studies pointed to differences in severity and prognosis of patients with HCM caused by different mutations.132134 Considering multiplicity of determinants variability in phenotypic expression of in HCM is to be expected. Such multiplicity also limits the impact of each determinant in accurately predicting the severity of the disease or clinical outcomes in a given individual. This variability is not restricted to phenotypic expression of HCM among patients with different causal mutations, but it also extends to family members carrying the same mutation (Figure 4).134136 Overall, clinical phenotypes do not seem to differ significantly between the 2 most common HCM genes, that is, MYH7 and MYBPC3.133,135 Compared with HCM caused by mutations in the protein components of the thick myofilaments, such as MYH7 and MYBPC3, HCM caused by mutations in the thin myofilament proteins, such as TNNT2 (cardiac troponin T), exhibits a milder cardiac hypertrophy and an increased risk of systolic dysfunction.132,137 However, the risk of serious cardiac arrhythmias and sudden cardiac death (SCD) does not seem to differ between the 2 groups.137

    Figure 4.

    Figure 4. Variability in the phenotypic expression of hypertrophic cardiomyopathy (HCM). A, A truncated pedigree depicting dizygotic twins with HCM caused by the p.Ser48Pro mutation in the MYOZ2 gene. Despite sharing the same causal mutation, one expresses mild and the other severe cardiac hypertrophy, as reflected in the electrocardiograms (B) and echocardiographic images (C).

    The variability in phenotypic expression of HCM is in part because of the effects of modifier genetic variants and environmental factors. Each human nuclear genome contains an average of ≈11 000 nonsynonymous variants, 160 premature protein truncation variants, and 500 000 variants in the known regulatory regions.52,53 Pathogenic variants in genes and pathways implicated in regulating cardiac hypertrophy and fibrosis could influence expression of the HCM phenotype and, hence, function as modifier genetic variants. The modifier variants, unlike the causal mutations, are neither necessary nor sufficient to cause HCM. They simply influence expression of HCM, each exerting a modest effect. In accord with the diversity of the human genome, the modifier variants are also expected to differ among individuals and hence, in part, explain interindividual variability in the phenotypic expression of HCM.

    Although the majority of modifier genes and their variants in HCM are largely unknown, several modifier loci containing several candidate genes have been implicated in influencing expression of cardiac hypertrophy.138 For example, an insertion/deletion variant in the ACE gene (angiotensin-1 converting enzyme), which is associated with variation in the plasma levels of ACE,139 has been shown to modify, albeit modestly, expression of cardiac hypertrophy and the risk of SCD in HCM.140142 Several other genes, identified primarily by the candidate gene approach, are also implicated as modifiers of HCM.142144 Likewise, experimental data in mouse models have implicated the Fhl1 gene, encoding FHL1 (four-and-a-half LIM domain protein 1), as a possible modifier of cardiac hypertrophy and function.128 Overall, data on the modifier genes in human HCM are preliminary.

    Phenotypic Variability Because of Multiple Pathogenic Variants in the Causal Genes

    A subset of patients with HCM carries ≥2 mutations in sarcomere proteins.6870,73,74 The data, albeit scant, suggest that the presence of multiple mutations is associated with more severe hypertrophy in HCM.6870,74

    Phenotypic Variability Because of Loading Conditions

    Considering that cardiac hypertrophy is a consequence of the initial defects caused by the mutations, changes in loading conditions, such as systemic arterial hypertension, would also be expected to enhance penetrance of the sarcomere protein mutations and severity of hypertrophy. In accord with the above notion, one might surmise that heavy physical activity, particularly isometric exercises to accelerate and exaggerate cardiac hypertrophy in the presence of mutations in genes encoding sarcomere proteins.

    Pleiotropic (Multiple) Phenotypes of Mutations

    An intriguing aspect of sarcomere protein mutations is pleiotropy. Accordingly, mutations in the same gene could manifest as HCM, dilated cardiomyopathy, restrictive cardiomyopathy, and even left ventricular noncompaction syndrome.145148 The pleiotropic effects primarily apply to the effects of different mutations in a given genes, such as TNNT2 or MYH7, causing the differing phenotypes of HCM and dilated cardiomyopathy.22,149 It may reflect location of the mutations in different domains of the protein, resulting in differential interactions of the mutant proteins with the other protein constituents of sarcomeres and activating different sets of intermediary molecular events.99,110 Likewise, differential effects of the causal mutations on Ca++ sensitivity of ATPase activity and force generation could explain, in part, the ensuing pleiotropic phenotypes.99,110,150154 Accordingly, mutations in thin filament proteins, such as cardiac troponin T that cause HCM typically enhance Ca+2 sensitivity of myofibrillar force generation and ATPase activity, whereas those leading to dilated cardiomyopathy suppress these functional effects.99,110,150155 Moreover, differences in myofilament tension development between HCM- and dilated cardiomyopathy–causing mutations might explain the contrasting phenotypes resulting from sarcomere protein mutations.99,110,150156

    Phenotypic Characteristics

    Patients with HCM exhibit a variable phenotype with ventricular hypertrophy being the cardinal manifestation, myocyte hypertrophy, and disarray as well as interstitial fibrosis as key pathological hallmarks and impaired ventricular filling and dynamic left ventricular outflow tract (LVOT) obstruction as important pathophysiologic features.

    Pathology

    Cardiac hypertrophy is the keystone to clinical diagnosis, which is typically based on detection by cardiac imaging157 (Figure 5). The hypertrophy is frequently asymmetrical, and predominantly involves the basal interventricular septum, just below the aortic valve but usually involves the free wall of the left ventricle as well.158 In asymmetrical septal hypertrophy (an earlier name for HCM159), the ratio of the thickness of the septum to the free wall of the ventricle ≥1.3/1.0; this applies to many but not all patients, and it is not specific. The cardiac apex is occasionally the predominant site of involvement, and this subset is referred to as apical HCM. Rarely, the posterior or lateral walls are the sites of predominant hypertrophy.

    Figure 5.

    Figure 5. Echocardiographic phenotypes of hypertrophic cardiomyopathy (HCM). A, A parasternal view of the ventricles showing septal hypertrophy. B, A parasternal short-axis view showing concentric cardiac hypertrophy. C, Systolic anterior motion (SAM) of mitral leaflets, contributing to left ventricular outflow tract (LVOT) obstruction. D, Doppler velocities recorded at the LVOT showing ≈100 mm Hg gradient. E, Tissue Doppler recording of the interventricular septum showing reduced velocities. F, Color Doppler M-mode imaging of the left ventricle showing velocity of flow progression in the left ventricle during a cardiac cycle, which is used, along with other indices, to assess diastolic function.

    Other frequent pathological features include elongation of the anterior or both leaflets of the mitral valve, as well as abnormal insertion of the associated papillary muscle.160 The right ventricle is infrequently involved by hypertrophy and only rarely manifests outflow tract obstruction.158,161

    Microscopic Anatomy

    The cardiac myocytes are enlarged with bizarre shapes and pleiotropic nuclei. They are in disarray, with loss of normal parallel alignment because of haphazard orientation of hypertrophic myocytes, which give a swirling appearance to the myocardial architecture (Figure 6).162,163 This disarray typically involves >10% of the myocardium in HCM, is widely distributed, with a predilection for the hypertrophied interventricular septum.163,164 Extensive, severe myocyte disarray has been observed at autopsy in patients in whom SCD has occurred.165 Noninvasive assessment of myocyte disarray is challenging but might be accomplished using diffusion tensor imaging, which assesses myofibrillar orientation.166

    Figure 6.

    Figure 6. Histological phenotypes. A, A normal thin myocardial section stained with hematoxylin and eosin (H&E). B, A low-magnification (×4) H&E-stained thin myocardial section from a patient heart with hypertrophic cardiomyopathy showing disorganized myocardial architecture. C, A higher magnification (×20) H&E-stained myocardial section showing myocyte disarray (×). D, A thin myocardial section (×20) stained with Masson trichrome in blue showing areas of interstitial fibrosis.

    Increased interstitial fibrosis is a common feature of HCM.167169 Cardiac magnetic resonance imaging has emerged as a useful tool for providing precise measurements of cardiac structure and to quantify late gadolinium enhancement, a marker of interstitial fibrosis.157 Late gadolinium enhancement is frequently detectable in patients with HCM, and its extent is associated with adverse clinical outcomes, including heart failure SCD and nonfatal cardiac arrhythmias.168171 The intramural coronary arteries exhibit wall thickening and narrow lumina.172 These vessels are imbedded in the interstitial fibrous tissue and can contribute to myocardial ischemia. Cleavage products of collagen synthesis and degradation have been used as biomarkers for interstitial fibrosis.173175 Likewise, elevated levels of cytokines, cardiac troponin T, and markers of myocardial inflammation have been associated with and reflect myocardial necrosis and fibrosis in patients with HCM.176178 Moreover, circulating levels of several microRNAs (especially miR29a) are elevated in HCM and may serve as markers for cardiac hypertrophy and interstitial fibrosis.179181 However, the clinical utilities of plasma levels of these markers in evaluation of cardiac hypertrophy and myocardial fibrosis in HCM remain to be established.

    Hemodynamics

    The left ventricle in HCM usually has a normal end-diastolic volume, a high-normal (65% to 70%) or elevated (>70%) ejection fraction, and a reduced end-systolic volume. The LVOT is encroached on by the hypertrophied septum. In the presence of hyperdynamic ejection, there is systolic anterior motion of the anterior leaflet of the mitral valve, which abuts the hypertrophied interventricular system during systole, causing obstruction to outflow, reflected in a systolic pressure gradient between the left ventricular cavity and the aorta that is present at rest in approximately one third of patients with HCM (Figure 5).158,182184 The obstruction is dynamic in nature, a unique feature of HCM. The severity of the LVOT obstruction is influenced by the ventricular volume, which in turn is a function of the interactions between myocardial contractility, ventricular preload, and afterload. An increased contractility or a reduction in preload or afterload, by reducing ventricular volume, could cause or intensify obstruction; the opposite occurs with reductions in contractility and increases in preload and afterload.185188 Thus, muscular exercise, infusion of isoproterenol, inhalation of amyl nitrite, sublingual nitroglycerin, and the strain phase of the Valsalva maneuver can provoke LVOT obstruction in HCM patients without a resting gradient.

    Diastolic dysfunction is frequent in HCM.189 It is caused by the increased interstitial fibrosis and the slowed relaxation and increased stiffness of the thickened ventricular wall. Left atrial volume, an indirect indicator of left ventricular diastolic pressure, is often increased in patients with HCM and is a predictor of the development of atrial fibrillation and heart failure.190 Regional systolic myocardial function is also commonly impaired, as detected by pulsed and tissue Doppler imaging, 3-dimensional speckle-tracking echocardiography, and magnetic resonance imaging.191193 In ≈5% of patients with HCM, usually elderly patients with extensive, long-standing LVOT obstruction and severe interstitial fibrosis, systolic function declines, the left ventricular wall thins, the cavity enlarges, and heart failure with reduced ejection fraction, sometimes referred to as burnt out HCM, develops.

    Clinical Manifestations

    Despite the presence of cardiac hypertrophy, patients with HCM are commonly asymptomatic or minimally symptomatic. The most frequent symptoms result from 4 major pathophysiologic conditions: diastolic ventricular dysfunction, obstruction to LVOT, imbalance between myocardial oxygen supply and demand, and cardiac arrhythmias.

    Diastolic Dysfunction

    As a consequence of this common pathophysiologic manifestation in HCM (see above),189 the left ventricular end diastolic pressure is elevated,194 which in turn raises left atrial, pulmonary venous, and pulmonary capillary pressures.195 The left ventricular diastolic pressure increases markedly on exertion, causing exertional dyspnea, exercise intolerance, orthopnea, peripheral edema, and heart failure with preserved ejection fraction.

    LVOT Obstruction

    Approximately one third of patients with HCM have LVOT obstruction at rest, which is intensified with exercise. One third have provokable obstruction (see below), and the remaining third have left ventricular hypertrophy without obstruction at rest and is not provokable.196198 Patients with severe obstruction usually have elevated ventricular diastolic pressure and exertional dyspnea; some develop frank heart failure. Exertional or immediately postexertional syncope may be caused by severe obstruction, with or without ventricular arrhythmia.

    Chest Pain

    Patients with HCM often experience ischemic chest pain, which may or may not have the typical features of angina pectoris. This symptom occurs because of imbalance between myocardial oxygen supply and demand. There is myocardial hypoperfusion secondary to reduced blood flow through the aforementioned thick-walled, intramural coronary arteries with luminal narrowing and the increased oxygen demand of the hypertrophied myocardium.172

    Arrhythmias

    Palpitations, presyncope, and syncope, often caused by recurrent nonsustained ventricular tachycardia, are among the cardinal clinical manifestations. Supraventricular and ventricular ectopic beats are common and nonsustained ventricular tachycardia is detected in 20% to 30% of patients.199,200 Nonsustained ventricular tachycardia is a major risk factor for SCD, as such episodes may lead to ventricular fibrillation, the usual cause of SCD.199,201,202 Syncope may also be caused by severe LVOT obstruction.201204 The underlying mechanisms of ventricular arrhythmias in HCM are largely unknown. Potential mechanisms include ventricular remodeling that is associated with cardiac hypertrophy, interstitial fibrosis, myocardial ischemia, and myocyte disarray.

    Atrial fibrillation occurs in about a quarter of patients with HCM and LVOT obstruction and has an annual incidence of ≈ 2% to 3%.205208 This arrhythmia is poorly tolerated because the combination of the loss of the atrial contribution to ventricular filling and the rapid ventricular rate results in further elevations of left ventricular diastolic pressure and symptoms of heart failure.205,208,209 It is also a major risk factor for thromboembolic stroke.205,207,208 Left atrial size and function as well as LVOT obstruction are major risk factors for atrial fibrillation.207 The underpinning mechanism(s) of atrial fibrillation in HCM has not been delineated. Potential mechanisms include atrial enlargement and stretch because of diastolic dysfunction, atrial fibrosis, expression of the mutant protein, and altered gene expression.

    Physical Examination

    On palpation, the precordial impulse is usually forceful, and displaced leftward and the peripheral arterial pulses are brisk. In patients with marked ventricular hypertrophy, atrial contraction is often especially vigorous, reflected in a prominent atrial contraction (a) wave in the jugular venous pulse, a presystolic apical lift, and, importantly, a prominent fourth heart sound (S4).158

    In patients with LVOT obstruction, a harsh, midsystolic, grade 3 to 4/6 murmur loudest between the apex and the left sternal border is usually audible. This murmur typically varies with interventions that alter ventricular loading and contractility (see above). Thus, the murmur increases in intensity when left ventricular volume declines during the strain of the Valsalva maneuver, when assuming the erect position, and during and immediately after exercise. In contrast, it diminishes during squatting, isometric handgrip, and immediately after release of the Valsalva maneuver. Mitral regurgitation, which is frequently present, is usually accompanied by a prominent, high-pitched blowing holosystolic murmur loudest at the apex.158

    ECG

    The ECG is abnormal in most patients with HCM, including those with no or only mild LVOT obstruction. The most common abnormalities are voltage changes of left ventricular hypertrophy, ST-T wave changes, and deep Q waves probably caused by depolarization of a hypertrophied interventricular septum. ECG evidence of left atrial enlargement may also be evident. About 2% to 5% of patients with HCM exhibit ECG findings of pre-excitation and may present with atrioventricular nodal supraventricular reciprocating arrhythmias, the Wolff–Parkinson–White syndrome.

    Diagnosis

    The clinical diagnosis of HCM is based on the presence of left ventricular hypertrophy, typically defined by an end-diastolic ventricular septal thickness in adults ≥13 mm, occurring in the absence of abnormal loading conditions or other secondary causes, such as hypertension, aortic stenosis, the physiological hypertrophy of athletes (see below), or phenocopied conditions. The cut point of 13 mm in adults offers high sensitivity in detecting HCM but has the risk of overdiagnosis, particularly in the presence of concomitant diseases. A cut point of 15 mm has been recommended by the European Society of Cardiology working group.5 It offers a greater diagnostic specificity for HCM but a lower sensitivity. Expression of cardiac hypertrophy in HCM is age dependent. It is infrequent in childhood and most commonly develops during adolescence and seldom after the fifth decade.6 In children, a Z score, reflecting deviation from an age- and sex-matched population, is often used to define cardiac hypertrophy.210 Also of value in diagnosis are genetic testing (see below), electrocardiography, and cardiac magnetic resonance imaging.

    Hypertension or valvular aortic stenosis can develop in patients with HCM, and it can be challenging to clarify the coexistence of these conditions. Features characteristic of HCM such as the distribution pattern of cardiac hypertrophy, its severity, a hyperdynamic left ventricle with a small cavity and the presence of LVOT obstruction, and genetic testing all can aid in the recognition of HCM in the presence of secondary hypertrophy.

    Adolescents and adults who are active in competitive sports often exhibit significant physiological cardiac hypertrophy with left ventricular wall thickness between 13 and 18 mm.211 Given that HCM is the most common cause of SCD in young athletes, distinction between physiological hypertrophy of athletes and pathological hypertrophy of HCM is of considerable importance.212,213 An important distinguishing feature is the size of the left ventricular cavity, which is not enlarged in HCM but is typically enlarged in the physiological hypertrophy of the athlete’s heart.211,214 In addition, the distribution pattern of cardiac hypertrophy with asymmetrical septal hypertrophy strongly favors HCM.211,214216 Electrocardiographic findings of abnormal Q waves, left atrial enlargement, and prominent repolarization abnormalities are features of HCM and are uncommon in physiological cardiac hypertrophy.214,217 Genetic testing (see below) could also be helpful.218

    Myocardial hypertrophy may also occur in patients with so-called phenocopy conditions, which may mimic HCM. These include Fabry disease, the glycogen storage diseases, lysosomal storage diseases, mitochondrial diseases, triplet repeat syndromes, and others (Table 2). The possibility of a phenocopy condition may be suspected in patients with exceptionally severe ventricular hypertrophy with very-high-voltage QRS complexes and a delta wave on a 12-lead ECG and the presence of concomitant noncardiac phenotypes, such as skeletal myopathy. Genetic testing (see below), when positive, is helpful in the distinction between HCM and a phenocopy condition. Endomyocardial biopsy and specific histological examination may identify a phenocopy condition, while the presence of myocyte disarray supports the diagnosis of HCM.

    Table 2. Phenocopy Conditions for Hypertrophic Cardiomyopathy

    Phenotype Gene Protein Phenotypic Clue
    AMPK-mediated glycogen storage PRKAG2 Protein kinase A, γ subunit Normal or reduced left ventricular systolic function, pre-excitation pattern
    Pompe disease GAA α-1,4-glucosidase (acid maltase) Autosomal recessive, multiorgan disease, pre-excitation pattern
    Anderson–Fabry disease GLA α-galactosidase A X-linked, multisystem also involving skin, kidney, and peripheral nerves
    Danon disease LAMP2 Lysosome-associated membrane protein 2 X-linked dominant, proximal muscle weakness, intellectual disability, short PR on ECG, elevated CK levels
    Amyloidosis TTR Transthyretin Low QRS voltage, other organ involvement, subendothelial LGE
    Kearns–Sayre syndrome mtDNA Mitochondrial protein Multisystem disease
    Friedreich ataxia FRDA Frataxin Autosomal recessive, neurodegeneration
    Myotonic dystrophy DMPK Myotonin protein kinase Myotonia, muscular dystrophy, cataract, and frontal baldness
    ZNF9 Zinc finger factor 9
    Noonan/LEOPARD syndromes (rasopathies) PTPN11 Protein tyrosine phosphatase, nonreceptor type 11 Congenital heart defects, lentigines, Café-au-lait spots
    SOS1 and SOS2 Son of sevenless
    RAF1 Murine leukemia viral oncogene homolog 1
    KRAS Kirsten rat sarcoma virus homolog
    Others (A2ML1, BRAF, CBL, MAP2K1, MAP2K2, NRAS, RIT1, RRAS, and SHOC2)
    Neimann–Pick disease NPC1 Neimann–Pick Autosomal recessive neurodegenerative disease
    Refsum disease PAHX (PHYH) Phytanoyl-CoA hydroxylase Retinitis pigmentosa, peripheral neuropathy, and ataxia
    Deafness MYO6 Unconventional myosin 6 Autosomal dominant deafness

    CK indicatescreatine kinase; and LGE, late gadolinium enhancement.

    Prognosis

    HCM is the most common cause of SCD in adolescents and young adults, particularly in competitive athletes.202,203,212,213,219 The risk factors for SCD are shown in Table 3 and discussed below.

    Despite the occurrence of SCD and heart failure in a minority of patients, as Maron et al220,221 has pointed out, HCM is a relatively benign disease, with approximately two thirds of patients with HCM experiencing a normal life span without significant morbidity.209,220222 This is particularly the case for patients with HCM without significant LVOT obstruction who may be totally asymptomatic or experience mild or moderate exertional dyspnea secondary to diastolic dysfunction.196198,209 Although the prognosis is guarded in patients deemed to be at high risk of SCD and in those with heart failure secondary to severe LVOT obstruction, fortunately, therapies for both of these major complications are now available (see below).

    Genetic Testing

    The combination of an increased understanding of the molecular genetic basis of HCM and a variety of technical innovations for detecting mutations in causal genes has ushered in the era of genetic testing. Patients in whom the diagnosis of HCM has been established, or appears likely, should undergo such testing that is now available in both academic and certified commercial laboratories.56,223 Currently available techniques, primarily based on whole-exome sequencing followed by analysis of a panel of ≈100 candidate genes implicated in cardiomyopathies, will identify such causal variants in ≈30% to 50% of probands with HCM.222224 The main challenge is in interpretation of the findings, particularly in a single affected individual. This is because of the plethora of the genetic variants in each gene in the human genome and the uncertain evidence of causality for many genes implicated in HCM.32,33 Consequently, unambiguous ascertainment of causality in a single case or small families is almost impossible. To avoid overinterpretation of the genetic testing findings, some have recommended restricting the testing to the well-established causal gene in HCM.33

    Genetic testing, if positive, will support the diagnosis in a proband, but if it is negative will not exclude it. If a proband with a positive test is identified, cascade screening, that is, testing for the presence of the variant in family members, should be undertaken. A positive screen, that is, mutation carrier relative of a patient with HCM, should lead to a detailed examination for the presence of the HCM phenotype. If the phenotype is expressed, the relative may be considered to have HCM and should be managed as described below. Relatives who are mutation carriers but phenotype negative should be followed with clinical evaluation (history, physical examination, ECG, and echocardiogram) at yearly intervals or more frequently if symptoms develop. Detailed examination of the mutation carriers might detect clinical and laboratory abnormalities, such as reduced tissue Doppler velocities, and elevated serum biomarkers.191,225,226 Given the age-related penetrance of the mutation, many mutation carriers/phenotype-negative relatives, especially those <25 to 30 years will eventually express the phenotype. However, as such individuals age, the likelihood of their conversion to phenotype positivity diminishes progressively, and longer intervals between follow-up examinations (eg, every 3–5 years) are appropriate. Because of incomplete penetrance, some mutation carriers remain free of the clinical phenotype for their entire lives, but they should be informed that they can pass the gene on to their offspring. Family members who do not carry the causal mutation are unlikely to develop HCM and should be counseled appropriately; they may, of course, participate in competitive sports.

    An important contribution of genetic testing is that it allows distinction between HCM and the phenocopy conditions. (Table 2) Genetic testing for HCM detects pathogenic variants in genes known to cause phenocopy conditions in ≈3% of the tested individuals.56 Identification of the latter has important implications, because their natural history and treatment differ from HCM and from one another. For example, specific enzyme replacement therapy has become available for selected phenocopy conditions, such as Fabry disease.223

    A shortcoming of current genetic testing for HCM is its failure to identify ≈50% to 60% of patients with HCM.224 This is primarily because of the so-called missing causal genes (see above). In large families in which the causal gene and its pathogenic mutation have not been identified, linkage analysis may be useful. In small families or sporadic cases, either whole-exome sequencing or the candidate gene approach may be required. Another limitation of genetic testing results from the great genetic heterogeneity of HCM (>1500 individual mutations have been described), which has prevented the genetic analysis from predicting the severity of hypertrophy or the risk of complications including SCD and heart failure. However, as already mentioned, the rare detection of multiple mutations (compound heterozygosity) by genetic testing has been associated with particularly severe hypertrophy.57,6871

    Management

    Clinical Assessment

    Evaluation of patients with HCM includes a detailed personal and family history for HCM, SCD, or heart failure, physical examination, 12-lead ECG, and complete echocardiographic assessment. Cardiac rhythm monitoring, cardiac magnetic resonance imaging, and cardiopulmonary exercise testing are optional, the latter being useful in patients suspected of having reduced exercise tolerance. Periodic re-evaluation is recommended, including in asymptomatic patients. Because the majority of patients with HCM are asymptomatic or minimally symptomatic, they do not require pharmacological treatment. However, all should be counseled to the genetic nature of the disease, the risk of passing it on to offspring, and the need to avoid participation in competitive sports or intensive exercise. However, most patients with HCM may participate in low-intensity sports such as golf or bowling.

    Pharmacotherapy

    β-Adrenergic receptor blockers, without intrinsic sympathetic activity, were first used in the treatment of HCM in the 1960s227229 and have, since then, remained the cornerstone of pharmacological treatment of symptomatic patients. They are effective in the relief of ischemic chest discomfort and may attenuate exercise-induced LVOT obstruction and resulting dyspnea as well.230 Disopyramide, a negative inotropic agent, when added to a β-blocker may reduce symptoms further in patients with LVOT obstruction.231 L-type calcium channel blockers, such as verapamil or diltiazem, may be beneficial in patients who do not tolerate or respond to β-blockers.232 Diuretics may be used in patients with HCM, pulmonary congestion, and frank heart failure, but minimal effective doses and careful observation are required to avoid hypovolemia, hypotension, and intensification or provocation of LVOT obstruction. New-onset atrial fibrillation is best treated with cardioversion. Because persistent and paroxysmal atrial fibrillation are risk factors for thromboembolism, long-term anticoagulation is necessary.208

    Management of Patients at Risk of SCD

    The risk of SCD ranges from 0.5% to 2% per year in adults with HCM, which is the most common cause of SCD in adolescents. Competitive athletes are at particular risk.201,233,234 The major cause is ventricular fibrillation, which cannot be prevented reliably by pharmacological intervention. However, in most instances, it can be treated effectively with an implanted cardioverter/defibrillator (ICD), which as Maron et al235238 have pointed out, is recommended in patients at a high risk of SCD. At highest risk are patients who have survived a cardiac arrest caused by ventricular fibrillation or sustained ventricular tachycardia, in whom implantation of an ICD is strongly indicated for secondary prevention.201,233,234 For primary prevention, high-risk features include unexplained syncope, an abnormal blood pressure response to exercise (hypotension), massive (≥30 mm) thickness of the interventricular septum or ventricular wall, a family history of HCM with SCD, multiple episodes of documented nonsustained ventricular tachycardia, which may be identified by an extended (30-day) period of cardiac rhythm monitoring, and extensive (≥15% of the left ventricular mass) late gadolinium enhancement determined by cardiac magnetic resonance imaging. ICD implantation is typically considered in patients who have one (or more) of these primary risk factors (Table 3). The rate of appropriate ICD discharge in HCM patient ranges from 3% to 15% per year depending on the specific indications, and it is higher in those with a previous arrhythmic event and in children.235238 The rate of ICD complications is <5% per year.236,238

    Table 3. Risk Factors for Sudden Cardiac Death in Patients With HCM

    Major risk factors
     Previous episode of cardiac arrest (aborted SCD)
     Family history of SCD (more than one family member)
     Reflective of the causal mutations, including double mutations, and the modifier genes
     History of recurrent syncope (caused by arrhythmias)
     Sustained and repetitive nonsustained ventricular tachycardia
    Intermediary risk factors
     Severe cardiac hypertrophy
     Extensive late gadolinium enhancement on cardiac MRI
    Questionable risk factors
     Severe LVOT obstruction (LVOT gradient >80 mm Hg)
     Abnormal blood pressure response to exercise
     Severe myocyte disarray
     Early onset of clinical manifestations (young age)
     Presence of myocardial ischemia

    HCM indicates hypertrophic cardiomyopathy; LVOT, left ventricular outflow tract; MRI, magnetic resonance imaging; and SCD, sudden cardiac death.

    Patients With LVOT Obstruction and Heart Failure

    Patients with obstruction (systolic pressure gradient ≥50 mm Hg at rest or with provocation) who are symptomatic despite pharmacological treatment should be considered for septal reduction therapy, either surgical septal myectomy or alcoholic septal ablation.239 Advanced stages of heart failure with preserved or reduced ejection fraction who have failed pharmacotherapy and, when indicated, septal reduction therapy, may require implantation of a left ventricular device or cardiac transplantation.145

    Experimental Therapies

    Current pharmacological or interventional treatment of patients with HCM, while often effective in relieving or preventing symptoms, do not target either the underlying genetic defect or the key intermediary pathways involved in the pathogenesis of the phenotype. Hence, they are not effective in prevention or induction of regression of cardiac hypertrophy and fibrosis. Elucidation of the molecular genetics and pathogenesis of HCM is stimulating development and testing of many pharmacological interventions (Table 4).

    Preliminary studies in animal models of HCM suggested possible benefits of angiotensin II receptor blockers, statins, mineralocorticoid receptor blockers, and antioxidant N-acetylcysteine.122124,240242 Despite the beneficial effects of some of these approaches in the models, preliminary studies in humans have been largely disappointing.243,244

    Delivery of Mybpc-3 by means of adeno-associated virus encoding cardiac myosin-binding protein C prevented development of HCM in a mouse model.245 HCM-mutant mRNA encoding myosin-binding protein C has also been repaired by 5′-trans splicing, which prevented development of HCM in neonatal mice with frameshift mutations.246 Gedicke–Hornung et al247 produced a modified mRNA and protein encoded by the Mybpc-3 gene, which reduced a lethal mutation in a neonatal mouse and prevented the development of HCM. An RNAi approach delivered by an adeno-associated virus that selectively targets the mutant Mhy6 allele that encodes β myosin heavy chain has also been shown to delay expression of cardiac hypertrophy and fibrosis in this model.248

    The calcium channel blocker diltiazem has been shown to prevent the development of HCM in a mouse model and in a small randomized human pilot trial in MYBP-3 carriers.249 The VANISH trial has randomized 150 patients who are sarcomere mutation carriers with no or minimal symptoms to the angiotensin receptor blocker valsartan or placebo (NCT01912534). Results are expected in 2019.210

    MYK-461 is an orally administered small molecule that allosterically inhibits myosin ATPase activity, diminishes myocyte force production, and has been shown to suppress development of cardiac hypertrophy, myocyte disarray, and fibrosis in a mouse model of HCM.250 After successful completion of 3 phase I clinical trials with this compound, it is now being evaluated in a phase 2 trial in patients with HCM and LVOT obstruction [NCT02842242].

    A partial list of ongoing clinical trials in human patients with HCM is provided in Table 4.

    Table 4. Selected Ongoing Trials in HCM

    Study Medication Sample Size Primary End Point
    HALT-HCM NAC 42 Feasibility/LVM
    LIBERTY-HCM GS-6615 160 VO2 max
    INHERIT Losartan 130 LVM
    NCT01150461 Losartan 20 LVM
    NCT00319982 Diltiazem 39 Diastolic function
    METAL-HCM Perhexiline 44 VO2 max
    NCT00011076 Pirfenidne 50 Diastolic function
    RHYME Ranolazine 14 Safety
    NCT00001534 Enalapril 112 Symptoms
    NCT00001965 Cyclosporine 32 Safety
    VANISH Valsartan 150 Combined end point
    NCT01696370 Trimetazidine 90 VO2 max
    Light-CARMIDO BX1514M 40 6 MWT

    HCM indicates hypertrophic cardiomyopathy; and MWT, minute walk time.

    Future Directions

    Since its modern characterization more than a half century ago, progress in the diagnosis and management of patients with HCM has paralleled technological advances in genetic testing, cardiac imaging, prevention of serious arrhythmias, cardiac surgery, and interventional cardiology. Enhanced annotation of the human genetic variants and their variable relation to clinical expression is likely to facilitate identification of people who carry the pathogenic variants. It will provide much needed information on the prognosis of the growing number of such people, many of whom are relatives of patients with HCM and who harbor the same mutation but have no observable manifestations of HCM. Such individuals might, in the future, become subjects for treatments designed to slow or prevent conversion to phenotypic positivity. These may include pharmacological treatments and other interventions, now still in the experimental stage. Also, much more needs to be learned about nonmutation carriers who are phenotype positive, including sporadic cases of HCM, to identify their missing causal genes (if any) and their natural history.

    It is anticipated that in the future the field will shift from targeting phenotypes such as myocyte hypertrophy, fibrosis, arrhythmias, and LVOT obstruction, toward correcting the underlying genetic disorders. It is hoped that the focus will shift from population- to individual-based approaches, using genetic and phenotypic characterization of each patient or potential patient. This was elegantly expressed a century ago by Sir William Osler: “The good physician treats the disease; the great physician treats the patient who has the disease” (http://www.osler.org.uk/osleriana-2/oslers-aphorisms/, accessed 3/9/2017).

    Circulation Research Compendium on Cardiomyopathies

    Cardiomyopathies: An Overview

    Classification, Epidemiology, and Global Burden of Cardiomyopathies

    Dilated Cardiomyopathy: Genetic Determinants and Mechanisms

    Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy

    Hypertrophic Obstructive Cardiomyopathy: Surgical Myectomy and Septal Ablation

    Arrhythmogenic Cardiomyopathy

    Inflammatory Cardiomyopathic Syndromes

    Restrictive Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy

    Cardiomyopathies Due to Left Ventricular Noncompaction, Mitochondrial and Storage Diseases, and Inborn Errors of Metabolism

    Pediatric Cardiomyopathies

    Modern Imaging Techniques in Cardiomyopathies

    Editors: Eugene Braunwald and Ali J. Marian

    Nonstandard Abbreviations and Acronyms

    HCM

    hypertrophic cardiomyopathy

    ICD

    implanted cardioverter/defibrillator

    LoF

    loss of function

    LVOT

    left ventricular outflow tract

    SCD

    sudden cardiac death

    Footnotes

    Correspondence to Ali J. Marian, MD, Center for Cardiovascular Genetics, 6770 Bertner St, Suite C900A, Houston, TX 77030. E-mail

    References

    • 1. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE . Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary Artery Risk Development in (Young) Adults.Circulation. 1995; 92:785–789.LinkGoogle Scholar
    • 2. Maron BJ, Mathenge R, Casey SA, Poliac LC, Longe TF . Clinical profile of hypertrophic cardiomyopathy identified de novo in rural communities.J Am Coll Cardiol. 1999; 33:1590–1595.CrossrefMedlineGoogle Scholar
    • 3. Hada Y, Sakamoto T, Amano K, Yamaguchi T, Takenaka K, Takahashi H, Takikawa R, Hasegawa I, Takahashi T, Suzuki J . Prevalence of hypertrophic cardiomyopathy in a population of adult Japanese workers as detected by echocardiographic screening.Am J Cardiol. 1987; 59:183–184.CrossrefMedlineGoogle Scholar
    • 4. Zou Y, Song L, Wang Z, Ma A, Liu T, Gu H, Lu S, Wu P, Zhang dagger Y, Shen dagger L, Cai Y, Zhen double dagger Y, Liu Y, Hui R . Prevalence of idiopathic hypertrophic cardiomyopathy in China: a population-based echocardiographic analysis of 8080 adults.Am J Med. 2004; 116:14–18.CrossrefMedlineGoogle Scholar
    • 5. Authors/Task Force Members, Elliott PM, Anastasakis A, Borger MA, Borggrefe M, Cecchi F, Charron P, Hagege AA, Lafont A, Limongelli G, Mahrholdt H, McKenna WJ, Mogensen J, Nihoyannopoulos P, Nistri S, Pieper PG, Pieske B, Rapezzi C, Rutten FH, Tillmanns C, Watkins H . 2014 ESC guidelines on diagnosis and management of hypertrophic cardiomyopathy: the task force for the diagnosis and management of hypertrophic cardiomyopathy of the european society of cardiology (ESC).Eur Heart J. 2014; 35:2733–2779. doi: 10.1093/eurheartj/ehu284.CrossrefMedlineGoogle Scholar
    • 6. Charron P, Dubourg O, Desnos M, Isnard R, Hagege A, Millaire A, Carrier L, Bonne G, Tesson F, Richard P, Bouhour JB, Schwartz K, Komajda M . Diagnostic value of electrocardiography and echocardiography for familial hypertrophic cardiomyopathy in a genotyped adult population.Circulation. 1997; 96:214–219.LinkGoogle Scholar
    • 7. Niimura H, Bachinski LL, Sangwatanaroj S, Watkins H, Chudley AE, McKenna W, Kristinsson A, Roberts R, Sole M, Maron BJ, Seidman JG, Seidman CE . Mutations in the gene for cardiac myosin-binding protein C and late-onset familial hypertrophic cardiomyopathy.N Engl J Med. 1998; 338:1248–1257. doi: 10.1056/NEJM199804303381802.CrossrefMedlineGoogle Scholar
    • 8. Bick AG, Flannick J, Ito K, et al . Burden of rare sarcomere gene variants in the Framingham and Jackson Heart Study cohorts.Am J Hum Genet. 2012; 91:513–519. doi: 10.1016/j.ajhg.2012.07.017.CrossrefMedlineGoogle Scholar
    • 9. Maron BJ, Peterson EE, Maron MS, Peterson JE . Prevalence of hypertrophic cardiomyopathy in an outpatient population referred for echocardiographic study.Am J Cardiol. 1994; 73:577–580.CrossrefMedlineGoogle Scholar
    • 10. Semsarian C, Ingles J, Maron MS, Maron BJ . New perspectives on the prevalence of hypertrophic cardiomyopathy.J Am Coll Cardiol. 2015; 65:1249–1254. doi: 10.1016/j.jacc.2015.01.019.CrossrefMedlineGoogle Scholar
    • 11. Niimura H, Patton KK, McKenna WJ, Soults J, Maron BJ, Seidman JG, Seidman CE . Sarcomere protein gene mutations in hypertrophic cardiomyopathy of the elderly.Circulation. 2002; 105:446–451.LinkGoogle Scholar
    • 12. Sachdev B, Takenaka T, Teraguchi H, Tei C, Lee P, McKenna WJ, Elliott PM . Prevalence of Anderson-Fabry disease in male patients with late onset hypertrophic cardiomyopathy.Circulation. 2002; 105:1407–1411.LinkGoogle Scholar
    • 13. Seo J, Kim M, Hong GR, Kim DS, Son JW, Cho IJ, Shim CY, Chang HJ, Ha JW, Chung N . Fabry disease in patients with hypertrophic cardiomyopathy: a practical approach to diagnosis.J Hum Genet. 2016; 61:775–780. doi: 10.1038/jhg.2016.52.CrossrefMedlineGoogle Scholar
    • 14. Chimenti C, Pieroni M, Morgante E, Antuzzi D, Russo A, Russo MA, Maseri A, Frustaci A . Prevalence of Fabry disease in female patients with late-onset hypertrophic cardiomyopathy.Circulation. 2004; 110:1047–1053. doi: 10.1161/01.CIR.0000139847.74101.03.LinkGoogle Scholar
    • 15. Greaves SC, Roche AH, Neutze JM, Whitlock RM, Veale AM . Inheritance of hypertrophic cardiomyopathy: a cross sectional and M mode echocardiographic study of 50 families.Br Heart J. 1987; 58:259–266.CrossrefMedlineGoogle Scholar
    • 16. Branzi A, Romeo G, Specchia S, Lolli C, Binetti G, Devoto M, Bacchi M, Magnani B . Genetic heterogeneity of hypertrophic cardiomyopathy.Int J Cardiol. 1985; 7:129–138.CrossrefMedlineGoogle Scholar
    • 17. Hartmannova H, Kubanek M, Sramko M, Piherova L, Noskova L, Hodanova K, Stranecky V, Pristoupilova A, Sovova J, Marek T, Maluskova J, Ridzon P, Kautzner J, Hulkova H, Kmoch S . Isolated X-linked hypertrophic cardiomyopathy caused by a novel mutation of the four-and-a-half LIM domain 1 gene.Circ Cardiovasc Genet. 2013; 6:543–551. doi: 10.1161/CIRCGENETICS.113.000245.LinkGoogle Scholar
    • 18. Marian AJ . Challenges in the diagnosis of Anderson-Fabry disease: a deceptively simple and yet complicated genetic disease.J Am Coll Cardiol. 2016; 68:1051–1053. doi: 10.1016/j.jacc.2016.06.026.CrossrefMedlineGoogle Scholar
    • 19. Gelb BD, Roberts AE, Tartaglia M . Cardiomyopathies in Noonan syndrome and the other RASopathies.Prog Pediatr Cardiol. 2015; 39:13–19. doi: 10.1016/j.ppedcard.2015.01.002.CrossrefMedlineGoogle Scholar
    • 20. Elliott P, Baker R, Pasquale F, Quarta G, Ebrahim H, Mehta AB, Hughes DA ; ACES study group. Prevalence of Anderson-Fabry disease in patients with hypertrophic cardiomyopathy: the European Anderson-Fabry disease survey.Heart. 2011; 97:1957–1960. doi: 10.1136/heartjnl-2011-300364.CrossrefMedlineGoogle Scholar
    • 21. Pare JA, Fraser RG, Pirozynski WJ, Shanks JA, Stubington D . Hereditary cardiovascular dysplasia. A form of familial cardiomyopathy.Am J Med. 1961; 31:37–62.CrossrefMedlineGoogle Scholar
    • 22. Geisterfer-Lowrance AA, Kass S, Tanigawa G, Vosberg HP, McKenna W, Seidman CE, Seidman JG . A molecular basis for familial hypertrophic cardiomyopathy: a beta cardiac myosin heavy chain gene missense mutation.Cell. 1990; 62:999–1006.CrossrefMedlineGoogle Scholar
    • 23. Millat G, Bouvagnet P, Chevalier P, Dauphin C, Jouk PS, Da Costa A, Prieur F, Bresson JL, Faivre L, Eicher JC, Chassaing N, Crehalet H, Porcher R, Rodriguez-Lafrasse C, Rousson R . Prevalence and spectrum of mutations in a cohort of 192 unrelated patients with hypertrophic cardiomyopathy.Eur J Med Genet. 2010; 53:261–267. doi: 10.1016/j.ejmg.2010.07.007.CrossrefMedlineGoogle Scholar
    • 24. Kaski JP, Syrris P, Esteban MT, Jenkins S, Pantazis A, Deanfield JE, McKenna WJ, Elliott PM . Prevalence of sarcomere protein gene mutations in preadolescent children with hypertrophic cardiomyopathy.Circ Cardiovasc Genet. 2009; 2:436–441. doi: 10.1161/CIRCGENETICS.108.821314.LinkGoogle Scholar
    • 25. Erdmann J, Daehmlow S, Wischke S, Senyuva M, Werner U, Raible J, Tanis N, Dyachenko S, Hummel M, Hetzer R, Regitz-Zagrosek V . Mutation spectrum in a large cohort of unrelated consecutive patients with hypertrophic cardiomyopathy.Clin Genet. 2003; 64:339–349.CrossrefMedlineGoogle Scholar
    • 26. Richard P, Charron P, Carrier L, et al ; EUROGENE Heart Failure Project. Hypertrophic cardiomyopathy: distribution of disease genes, spectrum of mutations, and implications for a molecular diagnosis strategy.Circulation. 2003; 107:2227–2232. doi: 10.1161/01.CIR.0000066323.15244.54.LinkGoogle Scholar
    • 27. Thierfelder L, Watkins H, MacRae C, Lamas R, McKenna W, Vosberg HP, Seidman JG, Seidman CE . Alpha-tropomyosin and cardiac troponin T mutations cause familial hypertrophic cardiomyopathy: a disease of the sarcomere.Cell. 1994; 77:701–712.CrossrefMedlineGoogle Scholar
    • 28. 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. doi: 10.1038/ng0897-379.CrossrefMedlineGoogle Scholar
    • 29. Mogensen J, Klausen IC, Pedersen AK, Egeblad H, Bross P, Kruse TA, Gregersen N, Hansen PS, Baandrup U, Borglum AD . Alpha-cardiac actin is a novel disease gene in familial hypertrophic cardiomyopathy.J Clin Invest. 1999; 103:R39–R43. doi: 10.1172/JCI6460.CrossrefMedlineGoogle Scholar
    • 30. Poetter K, Jiang H, Hassanzadeh S, Master SR, Chang A, Dalakas MC, Rayment I, Sellers JR, Fananapazir L, Epstein ND . Mutations in either the essential or regulatory light chains of myosin are associated with a rare myopathy in human heart and skeletal muscle.Nat Genet. 1996; 13:63–69. doi: 10.1038/ng0596-63.CrossrefMedlineGoogle Scholar
    • 31. Geier C, Gehmlich K, Ehler E, et al . Beyond the sarcomere: CSRP3 mutations cause hypertrophic cardiomyopathy.Hum Mol Genet. 2008; 17:2753–2765. doi: 10.1093/hmg/ddn160.CrossrefMedlineGoogle Scholar
    • 32. Walsh R, Buchan R, Wilk A, John S, Felkin LE, Thomson KL, Chiaw TH, Loong CC, Pua CJ, Raphael C, Prasad S, Barton PJ, Funke B, Watkins H, Ware JS, Cook SA . Defining the genetic architecture of hypertrophic cardiomyopathy: re-evaluating the role of non-sarcomeric genes [published online ahead of print January 11, 2017].Eur Heart J. doi: 10.1093/eurheartj/ehw603.Google Scholar
    • 33. Walsh R, Thomson KL, Ware JS, et al . Reassessment of Mendelian gene pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference samples.Genet Med. 2017; 19:192–203. doi: 10.1038/gim.2016.90.CrossrefMedlineGoogle Scholar
    • 34. Satoh M, Takahashi M, Sakamoto T, Hiroe M, Marumo F, Kimura A . Structural analysis of the titin gene in hypertrophic cardiomyopathy: identification of a novel disease gene.Biochem Biophys Res Commun. 1999; 262:411–417. doi: 10.1006/bbrc.1999.1221.CrossrefMedlineGoogle Scholar
    • 35. Hayashi T, Arimura T, Itoh-Satoh M, et al . Tcap gene mutations in hypertrophic cardiomyopathy and dilated cardiomyopathy.J Am Coll Cardiol. 2004; 44:2192–2201. doi: 10.1016/j.jacc.2004.08.058.CrossrefMedlineGoogle Scholar
    • 36. Marian AJ . Hypertrophic cardiomyopathy: from genetics to treatment.Eur J Clin Invest. 2010; 40:360–369.CrossrefMedlineGoogle Scholar
    • 37. Kimura A . Molecular genetics and pathogenesis of cardiomyopathy.J Hum Genet. 2016; 61:41–50. doi: 10.1038/jhg.2015.83.CrossrefMedlineGoogle Scholar
    • 38. Ho CY, Charron P, Richard P, Girolami F, Van Spaendonck-Zwarts KY, Pinto Y . Genetic advances in sarcomeric cardiomyopathies: state of the art.Cardiovasc Res. 2015; 105:397–408. doi: 10.1093/cvr/cvv025.CrossrefMedlineGoogle Scholar
    • 39. Osio A, Tan L, Chen SN, Lombardi R, Nagueh SF, Shete S, Roberts R, Willerson JT, Marian AJ . Myozenin 2 is a novel gene for human hypertrophic cardiomyopathy.Circ Res. 2007; 100:766–768. doi: 10.1161/01.RES.0000263008.66799.aa.LinkGoogle Scholar
    • 40. Chen SN, Czernuszewicz G, Tan Y, Lombardi R, Jin J, Willerson JT, Marian AJ . Human molecular genetic and functional studies identify TRIM63, encoding Muscle RING Finger Protein 1, as a novel gene for human hypertrophic cardiomyopathy.Circ Res. 2012; 111:907–919. doi: 10.1161/CIRCRESAHA.112.270207.LinkGoogle Scholar
    • 41. Friedrich FW, Wilding BR, Reischmann S, et al . Evidence for FHL1 as a novel disease gene for isolated hypertrophic cardiomyopathy.Hum Mol Genet. 2012; 21:3237–3254. doi: 10.1093/hmg/dds157.CrossrefMedlineGoogle Scholar
    • 42. Landstrom AP, Parvatiyar MS, Pinto JR, Marquardt ML, Bos JM, Tester DJ, Ommen SR, Potter JD, Ackerman MJ . Molecular and functional characterization of novel hypertrophic cardiomyopathy susceptibility mutations in TNNC1-encoded troponin C.J Mol Cell Cardiol. 2008; 45:281–288. doi: 10.1016/j.yjmcc.2008.05.003.CrossrefMedlineGoogle Scholar
    • 43. Carniel E, Taylor MR, Sinagra G, et al . Alpha-myosin heavy chain: a sarcomeric gene associated with dilated and hypertrophic phenotypes of cardiomyopathy.Circulation. 2005; 112:54–59. doi: 10.1161/CIRCULATIONAHA.104.507699.LinkGoogle Scholar
    • 44. Landstrom AP, Adekola BA, Bos JM, Ommen SR, Ackerman MJ . PLN-encoded phospholamban mutation in a large cohort of hypertrophic cardiomyopathy cases: summary of the literature and implications for genetic testing.Am Heart J. 2011; 161:165–171. doi: 10.1016/j.ahj.2010.08.001.CrossrefMedlineGoogle Scholar
    • 45. Hayashi T, Arimura T, Ueda K, Shibata H, Hohda S, Takahashi M, Hori H, Koga Y, Oka N, Imaizumi T, Yasunami M, Kimura A . Identification and functional analysis of a caveolin-3 mutation associated with familial hypertrophic cardiomyopathy.Biochem Biophys Res Commun. 2004; 313:178–184.CrossrefMedlineGoogle Scholar
    • 46. Almomani R, Verhagen JM, Herkert JC, et al . Biallelic truncating mutations in ALPK3 cause severe pediatric cardiomyopathy.J Am Coll Cardiol. 2016; 67:515–525. doi: 10.1016/j.jacc.2015.10.093.CrossrefMedlineGoogle Scholar
    • 47. Landstrom AP, Weisleder N, Batalden KB, Bos JM, Tester DJ, Ommen SR, Wehrens XH, Claycomb WC, Ko JK, Hwang M, Pan Z, Ma J, Ackerman MJ . Mutations in JPH2-encoded junctophilin-2 associated with hypertrophic cardiomyopathy in humans.J Mol Cell Cardiol. 2007; 42:1026–1035. doi: 10.1016/j.yjmcc.2007.04.006.CrossrefMedlineGoogle Scholar
    • 48. Marian AJ . Nature’s genetic gradients and the clinical phenotype.Circ Cardiovasc Genet. 2009; 2:537–539. doi: 10.1161/CIRCGENETICS.109.921940.LinkGoogle Scholar
    • 49. Marian AJ . Causality in genetics: the gradient of genetic effects and back to Koch’s postulates of causality.Circ Res. 2014; 114:e18–e21. doi: 10.1161/CIRCRESAHA.114.302904.LinkGoogle Scholar
    • 50. Marian AJ . The case of “missing causal genes” and the practice of medicine: a Sherlock Holmes approach of deductive reasoning.Circ Res. 2016; 119:21–24. doi: 10.1161/CIRCRESAHA.116.308830.LinkGoogle Scholar
    • 51. MacArthur DG, Manolio TA, Dimmock DP, et al . Guidelines for investigating causality of sequence variants in human disease.Nature. 2014; 508:469–476. doi: 10.1038/nature13127.CrossrefMedlineGoogle Scholar
    • 52. Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA, Abecasis GR ; 1000 Genomes Project Consortium. A global reference for human genetic variation.Nature. 2015; 526:68–74. doi: 10.1038/nature15393.CrossrefMedlineGoogle Scholar
    • 53. Abecasis GR, Altshuler D, Auton A, Brooks LD, Durbin RM, Gibbs RA, Hurles ME, McVean GA ; 1000 Genomes Project Consortium. A map of human genome variation from population-scale sequencing.Nature. 2010; 467:1061–1073. doi: 10.1038/nature09534.CrossrefMedlineGoogle Scholar
    • 54. Saltzman AJ, Mancini-DiNardo D, Li C, et al . Short communication: the cardiac myosin binding protein C Arg502Trp mutation: a common cause of hypertrophic cardiomyopathy.Circ Res. 2010; 106:1549–1552. doi: 10.1161/CIRCRESAHA.109.216291.LinkGoogle Scholar
    • 55. Page SP, Kounas S, Syrris P, Christiansen M, Frank-Hansen R, Andersen PS, Elliott PM, McKenna WJ . Cardiac myosin binding protein-C mutations in families with hypertrophic cardiomyopathy: disease expression in relation to age, gender, and long term outcome.Circ Cardiovasc Genet. 2012; 5:156–166. doi: 10.1161/CIRCGENETICS.111.960831.LinkGoogle Scholar
    • 56. Alfares AA, Kelly MA, McDermott G, et al . Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: expanded panels offer limited additional sensitivity.Genet Med. 2015; 17:880–888. doi: 10.1038/gim.2014.205.CrossrefMedlineGoogle Scholar
    • 57. Hodatsu A, Konno T, Hayashi K, Funada A, Fujita T, Nagata Y, Fujino N, Kawashiri MA, Yamagishi M . Compound heterozygosity deteriorates phenotypes of hypertrophic cardiomyopathy with founder MYBPC3 mutation: evidence from patients and zebrafish models.Am J Physiol Heart Circ Physiol. 2014; 307:H1594–H1604. doi: 10.1152/ajpheart.00637.2013.CrossrefMedlineGoogle Scholar
    • 58. Dausse E, Komajda M, Fetler L, Dubourg O, Dufour C, Carrier L, Wisnewsky C, Bercovici J, Hengstenberg C, al-Mahdawi S . Familial hypertrophic cardiomyopathy. Microsatellite haplotyping and identification of a hot spot for mutations in the beta-myosin heavy chain gene.J Clin Invest. 1993; 92:2807–2813. doi: 10.1172/JCI116900.CrossrefMedlineGoogle Scholar
    • 59. Forissier JF, Carrier L, Farza H, et al . Codon 102 of the cardiac troponin T gene is a putative hot spot for mutations in familial hypertrophic cardiomyopathy.Circulation. 1996; 94:3069–3073.LinkGoogle Scholar
    • 60. Coviello DA, Maron BJ, Spirito P, Watkins H, Vosberg HP, Thierfelder L, Schoen FJ, Seidman JG, Seidman CE . Clinical features of hypertrophic cardiomyopathy caused by mutation of a “hot spot” in the alpha-tropomyosin gene.J Am Coll Cardiol. 1997; 29:635–640.CrossrefMedlineGoogle Scholar
    • 61. Marian AJ, Yu QT, Mares A, Hill R, Roberts R, Perryman MB . Detection of a new mutation in the beta-myosin heavy chain gene in an individual with hypertrophic cardiomyopathy.J Clin Invest. 1992; 90:2156–2165. doi: 10.1172/JCI116101.CrossrefMedlineGoogle Scholar
    • 62. Watkins H, Seidman CE, Seidman JG, Feng HS, Sweeney HL . Expression and functional assessment of a truncated cardiac troponin T that causes hypertrophic cardiomyopathy. Evidence for a dominant negative action.J Clin Invest. 1996; 98:2456–2461. doi: 10.1172/JCI119063.CrossrefMedlineGoogle Scholar
    • 63. Blair E, Redwood C, de Jesus Oliveira M, Moolman-Smook JC, Brink P, Corfield VA, Ostman-Smith I, Watkins H . Mutations of the light meromyosin domain of the beta-myosin heavy chain rod in hypertrophic cardiomyopathy.Circ Res. 2002; 90:263–269.LinkGoogle Scholar
    • 64. Homburger JR, Green EM, Caleshu C, Sunitha MS, Taylor RE, Ruppel KM, Metpally RP, Colan SD, Michels M, Day SM, Olivotto I, Bustamante CD, Dewey FE, Ho CY, Spudich JA, Ashley EA . Multidimensional structure-function relationships in human beta-cardiac myosin from population-scale genetic variation.Proc Natl Acad Sci USA. 2016; 113:6701–6706. doi: 10.1073/pnas.1606950113.CrossrefMedlineGoogle Scholar
    • 65. Towe EC, Bos JM, Ommen SR, Gersh BJ, Ackerman MJ . Genotype-phenotype correlations in apical variant hypertrophic cardiomyopathy.Congenit Heart Dis. 2015; 10:E139–E145. doi: 10.1111/chd.12242.CrossrefMedlineGoogle Scholar
    • 66. Chapman KB, Solomon SD, Boeke JD . SDH1, the gene encoding the succinate dehydrogenase flavoprotein subunit from Saccharomyces cerevisiae.Gene. 1992; 118:131–136.CrossrefMedlineGoogle Scholar
    • 67. Marian AJ, Belmont J . Strategic approaches to unraveling genetic causes of cardiovascular diseases.Circ Res. 2011; 108:1252–1269. doi: 10.1161/CIRCRESAHA.110.236067.LinkGoogle Scholar
    • 68. Blair E, Price SJ, Baty CJ, Ostman-Smith I, Watkins H . Mutations in cis can confound genotype-phenotype correlations in hypertrophic cardiomyopathy.J Med Genet. 2001; 38:385–388.CrossrefMedlineGoogle Scholar
    • 69. Maron BJ, Maron MS, Semsarian C . Double or compound sarcomere mutations in hypertrophic cardiomyopathy: a potential link to sudden death in the absence of conventional risk factors.Heart Rhythm. 2012; 9:57–63. doi: 10.1016/j.hrthm.2011.08.009.CrossrefMedlineGoogle Scholar
    • 70. Girolami F, Ho CY, Semsarian C, Baldi M, Will ML, Baldini K, Torricelli F, Yeates L, Cecchi F, Ackerman MJ, Olivotto I . Clinical features and outcome of hypertrophic cardiomyopathy associated with triple sarcomere protein gene mutations.J Am Coll Cardiol. 2010; 55:1444–1453. doi: 10.1016/j.jacc.2009.11.062.CrossrefMedlineGoogle Scholar
    • 71. Lekanne Deprez RH, Muurling-Vlietman JJ, Hruda J, Baars MJ, Wijnaendts LC, Stolte-Dijkstra I, Alders M, van Hagen JM . Two cases of severe neonatal hypertrophic cardiomyopathy caused by compound heterozygous mutations in the MYBPC3 gene.J Med Genet. 2006; 43:829–832. doi: 10.1136/jmg.2005.040329.CrossrefMedlineGoogle Scholar
    • 72. Van Driest SL, Vasile VC, Ommen SR, Will ML, Tajik AJ, Gersh BJ, Ackerman MJ . Myosin binding protein C mutations and compound heterozygosity in hypertrophic cardiomyopathy.J Am Coll Cardiol. 2004; 44:1903–1910. doi: 10.1016/j.jacc.2004.07.045.CrossrefMedlineGoogle Scholar
    • 73. Li L, Bainbridge MN, Tan Y, Willerson JT, Marian AJ . A potential oligogenic etiology of hypertrophic cardiomyopathy: a classic single-gene disorder.Circ Res. 2017; 120:1084–1090. doi: 10.1161/CIRCRESAHA.116.310559.LinkGoogle Scholar
    • 74. Ingles J, Doolan A, Chiu C, Seidman J, Seidman C, Semsarian C . Compound and double mutations in patients with hypertrophic cardiomyopathy: implications for genetic testing and counselling.J Med Genet. 2005; 42:e59. doi: 10.1136/jmg.2005.033886.CrossrefMedlineGoogle Scholar
    • 75. van Dijk SJ, Dooijes D, dos Remedios C, Michels M, Lamers JM, Winegrad S, Schlossarek S, Carrier L, ten Cate FJ, Stienen GJ, van der Velden J . Cardiac myosin-binding protein C mutations and hypertrophic cardiomyopathy: haploinsufficiency, deranged phosphorylation, and cardiomyocyte dysfunction.Circulation. 2009; 119:1473–1483. doi: 10.1161/CIRCULATIONAHA.108.838672.LinkGoogle Scholar
    • 76. Popp MW, Maquat LE . Leveraging rules of nonsense-mediated mRNA decay for genome engineering and personalized medicine.Cell. 2016; 165:1319–1322. doi: 10.1016/j.cell.2016.05.053.CrossrefMedlineGoogle Scholar
    • 77. Siwaszek A, Ukleja M, Dziembowski A . Proteins involved in the degradation of cytoplasmic mRNA in the major eukaryotic model systems.RNA Biol. 2014; 11:1122–1136. doi: 10.4161/rna.34406.CrossrefMedlineGoogle Scholar
    • 78. Vasudevan S, Peltz SW, Wilusz CJ . Non-stop decay–a new mRNA surveillance pathway.Bioessays. 2002; 24:785–788. doi: 10.1002/bies.10153.CrossrefMedlineGoogle Scholar
    • 79. Frischmeyer PA, van Hoof A, O’Donnell K, Guerrerio AL, Parker R, Dietz HC . An mRNA surveillance mechanism that eliminates transcripts lacking termination codons.Science. 2002; 295:2258–2261. doi: 10.1126/science.1067338.CrossrefMedlineGoogle Scholar
    • 80. Carrier L, Bonne G, Bährend E, et al . Organization and sequence of human cardiac myosin binding protein C gene (MYBPC3) and identification of mutations predicted to produce truncated proteins in familial hypertrophic cardiomyopathy.Circ Res. 1997; 80:427–434.LinkGoogle Scholar
    • 81. Schlossarek S, Mearini G, Carrier L . Cardiac myosin-binding protein C in hypertrophic cardiomyopathy: mechanisms and therapeutic opportunities.J Mol Cell Cardiol. 2011; 50:613–620. doi: 10.1016/j.yjmcc.2011.01.014.CrossrefMedlineGoogle Scholar
    • 82. Marston S, Copeland O, Jacques A, Livesey K, Tsang V, McKenna WJ, Jalilzadeh S, Carballo S, Redwood C, Watkins H . Evidence from human myectomy samples that MYBPC3 mutations cause hypertrophic cardiomyopathy through haploinsufficiency.Circ Res. 2009; 105:219–222. doi: 10.1161/CIRCRESAHA.109.202440.LinkGoogle Scholar
    • 83. Rottbauer W, Gautel M, Zehelein J, Labeit S, Franz WM, Fischer C, Vollrath B, Mall G, Dietz R, Kübler W, Katus HA . Novel splice donor site mutation in the cardiac myosin-binding protein-C gene in familial hypertrophic cardiomyopathy. Characterization of cardiac transcript and protein.J Clin Invest. 1997; 100:475–482. doi: 10.1172/JCI119555.CrossrefMedlineGoogle Scholar
    • 84. Marston S, Copeland O, Gehmlich K, Schlossarek S, Carrier L, Carrrier L . How do MYBPC3 mutations cause hypertrophic cardiomyopathy?J Muscle Res Cell Motil. 2012; 33:75–80. doi: 10.1007/s10974-011-9268-3.CrossrefMedlineGoogle Scholar
    • 85. Moolman JA, Reith S, Uhl K, Bailey S, Gautel M, Jeschke B, Fischer C, Ochs J, McKenna WJ, Klues H, Vosberg HP . A newly created splice donor site in exon 25 of the MyBP-C gene is responsible for inherited hypertrophic cardiomyopathy with incomplete disease penetrance.Circulation. 2000; 101:1396–1402.LinkGoogle Scholar
    • 86. Sarikas A, Carrier L, Schenke C, Doll D, Flavigny J, Lindenberg KS, Eschenhagen T, Zolk O . Impairment of the ubiquitin-proteasome system by truncated cardiac myosin binding protein C mutants.Cardiovasc Res. 2005; 66:33–44. doi: 10.1016/j.cardiores.2005.01.004.CrossrefMedlineGoogle Scholar
    • 87. Barefield D, Kumar M, Gorham J, Seidman JG, Seidman CE, de Tombe PP, Sadayappan S . Haploinsufficiency of MYBPC3 exacerbates the development of hypertrophic cardiomyopathy in heterozygous mice.J Mol Cell Cardiol. 2015; 79:234–243. doi: 10.1016/j.yjmcc.2014.11.018.CrossrefMedlineGoogle Scholar
    • 88. Tripathi S, Schultz I, Becker E, Montag J, Borchert B, Francino A, Navarro-Lopez F, Perrot A, Özcelik C, Osterziel KJ, McKenna WJ, Brenner B, Kraft T . Unequal allelic expression of wild-type and mutated β-myosin in familial hypertrophic cardiomyopathy.Basic Res Cardiol. 2011; 106:1041–1055. doi: 10.1007/s00395-011-0205-9.CrossrefMedlineGoogle Scholar
    • 89. Kraft T, Montag J, Radocaj A, Brenner B . Hypertrophic cardiomyopathy: cell-to-cell imbalance in gene expression and contraction force as trigger for disease phenotype development.Circ Res. 2016; 119:992–995. doi: 10.1161/CIRCRESAHA.116.309804.LinkGoogle Scholar
    • 90. Helms AS, Davis FM, Coleman D, Bartolone SN, Glazier AA, Pagani F, Yob JM, Sadayappan S, Pedersen E, Lyons R, Westfall MV, Jones R, Russell MW, Day SM . Sarcomere mutation-specific expression patterns in human hypertrophic cardiomyopathy.Circ Cardiovasc Genet. 2014; 7:434–443. doi: 10.1161/CIRCGENETICS.113.000448.LinkGoogle Scholar
    • 91. Wolny M, Colegrave M, Colman L, White E, Knight PJ, Peckham M . Cardiomyopathy mutations in the tail of β-cardiac myosin modify the coiled-coil structure and affect integration into thick filaments in muscle sarcomeres in adult cardiomyocytes.J Biol Chem. 2013; 288:31952–31962. doi: 10.1074/jbc.M113.513291.CrossrefMedlineGoogle Scholar
    • 92. Geisterfer-Lowrance AA, Christe M, Conner DA, Ingwall JS, Schoen FJ, Seidman CE, Seidman JG . A mouse model of familial hypertrophic cardiomyopathy.Science. 1996; 272:731–734.CrossrefMedlineGoogle Scholar
    • 93. Tardiff JC, Factor SM, Tompkins BD, Hewett TE, Palmer BM, Moore RL, Schwartz S, Robbins J, Leinwand LA . A truncated cardiac troponin T molecule in transgenic mice suggests multiple cellular mechanisms for familial hypertrophic cardiomyopathy.J Clin Invest. 1998; 101:2800–2811. doi: 10.1172/JCI2389.CrossrefMedlineGoogle Scholar
    • 94. Frey N, Brixius K, Schwinger RH, Benis T, Karpowski A, Lorenzen HP, Luedde M, Katus HA, Franz WM . Alterations of tension-dependent ATP utilization in a transgenic rat model of hypertrophic cardiomyopathy.J Biol Chem. 2006; 281:29575–29582. doi: 10.1074/jbc.M507740200.CrossrefMedlineGoogle Scholar
    • 95. Oberst L, Zhao G, Park JT, Brugada R, Michael LH, Entman ML, Roberts R, Marian AJ . Dominant-negative effect of a mutant cardiac troponin T on cardiac structure and function in transgenic mice.J Clin Invest. 1998; 102:1498–1505. doi: 10.1172/JCI4088.CrossrefMedlineGoogle Scholar
    • 96. Marian AJ, Wu Y, Lim DS, McCluggage M, Youker K, Yu QT, Brugada R, DeMayo F, Quinones M, Roberts R . A transgenic rabbit model for human hypertrophic cardiomyopathy.J Clin Invest. 1999; 104:1683–1692. doi: 10.1172/JCI7956.CrossrefMedlineGoogle Scholar
    • 97. Lim DS, Oberst L, McCluggage M, Youker K, Lacy J, DeMayo F, Entman ML, Roberts R, Michael LH, Marian AJ . Decreased left ventricular ejection fraction in transgenic mice expressing mutant cardiac troponin T-Q(92), responsible for human hypertrophic cardiomyopathy.J Mol Cell Cardiol. 2000; 32:365–374. doi: 10.1006/jmcc.1999.1081.CrossrefMedlineGoogle Scholar
    • 98. Fraysse B, Weinberger F, Bardswell SC, Cuello F, Vignier N, Geertz B, Starbatty J, Krämer E, Coirault C, Eschenhagen T, Kentish JC, Avkiran M, Carrier L . Increased myofilament Ca2+ sensitivity and diastolic dysfunction as early consequences of Mybpc3 mutation in heterozygous knock-in mice.J Mol Cell Cardiol. 2012; 52:1299–1307. doi: 10.1016/j.yjmcc.2012.03.009.CrossrefMedlineGoogle Scholar
    • 99. Debold EP, Schmitt JP, Patlak JB, Beck SE, Moore JR, Seidman JG, Seidman C, Warshaw DM . Hypertrophic and dilated cardiomyopathy mutations differentially affect the molecular force generation of mouse alpha-cardiac myosin in the laser trap assay.Am J Physiol Heart Circ Physiol. 2007; 293:H284–H291. doi: 10.1152/ajpheart.00128.2007.CrossrefMedlineGoogle Scholar
    • 100. Lompré AM, Nadal-Ginard B, Mahdavi V . Expression of the cardiac ventricular alpha- and beta-myosin heavy chain genes is developmentally and hormonally regulated.J Biol Chem. 1984; 259:6437–6446.CrossrefMedlineGoogle Scholar
    • 101. Miyata S, Minobe W, Bristow MR, Leinwand LA . Myosin heavy chain isoform expression in the failing and nonfailing human heart.Circ Res. 2000; 86:386–390.LinkGoogle Scholar
    • 102. Reiser PJ, Portman MA, Ning XH, Schomisch Moravec C . Human cardiac myosin heavy chain isoforms in fetal and failing adult atria and ventricles.Am J Physiol Heart Circ Physiol. 2001; 280:H1814–H1820.CrossrefMedlineGoogle Scholar
    • 103. Walklate J, Ujfalusi Z, Geeves MA . Myosin isoforms and the mechanochemical cross-bridge cycle.J Exp Biol. 2016; 219:168–174. doi: 10.1242/jeb.124594.CrossrefMedlineGoogle Scholar
    • 104. Schwartz K, Lecarpentier Y, Martin JL, Lompré AM, Mercadier JJ, Swynghedauw B . Myosin isoenzymic distribution correlates with speed of myocardial contraction.J Mol Cell Cardiol. 1981; 13:1071–1075.CrossrefMedlineGoogle Scholar
    • 105. Litten RZ, Martin BJ, Low RB, Alpert NR . Altered myosin isozyme patterns from pressure-overloaded and thyrotoxic hypertrophied rabbit hearts.Circ Res. 1982; 50:856–864.LinkGoogle Scholar
    • 106. Witjas-Paalberends ER, Piroddi N, Stam K, et al . Mutations in MYH7 reduce the force generating capacity of sarcomeres in human familial hypertrophic cardiomyopathy.Cardiovasc Res. 2013; 99:432–441. doi: 10.1093/cvr/cvt119.CrossrefMedlineGoogle Scholar
    • 107. Witjas-Paalberends ER, Güçlü A, Germans T, et al . Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations.Cardiovasc Res. 2014; 103:248–257. doi: 10.1093/cvr/cvu127.CrossrefMedlineGoogle Scholar
    • 108. Bloemink M, Deacon J, Langer S, Vera C, Combs A, Leinwand L, Geeves MA . The hypertrophic cardiomyopathy myosin mutation R453C alters ATP binding and hydrolysis of human cardiac β-myosin.J Biol Chem. 2014; 289:5158–5167. doi: 10.1074/jbc.M113.511204.CrossrefMedlineGoogle Scholar
    • 109. Nagueh SF, Chen S, Patel R, Tsybouleva N, Lutucuta S, Kopelen HA, Zoghbi WA, Quiñones MA, Roberts R, Marian AJ . Evolution of expression of cardiac phenotypes over a 4-year period in the beta-myosin heavy chain-Q403 transgenic rabbit model of human hypertrophic cardiomyopathy.J Mol Cell Cardiol. 2004; 36:663–673. doi: 10.1016/j.yjmcc.2004.02.010.CrossrefMedlineGoogle Scholar
    • 110. Lombardi R, Bell A, Senthil V, Sidhu J, Noseda M, Roberts R, Marian AJ . Differential interactions of thin filament proteins in two cardiac troponin T mouse models of hypertrophic and dilated cardiomyopathies.Cardiovasc Res. 2008; 79:109–117. doi: 10.1093/cvr/cvn078.CrossrefMedlineGoogle Scholar
    • 111. Solaro RJ, Varghese J, Marian AJ, Chandra M . Molecular mechanisms of cardiac myofilament activation: modulation by pH and a troponin T mutant R92Q.Basic Res Cardiol. 2002; 97(suppl 1):I102–I110.CrossrefMedlineGoogle Scholar
    • 112. Crilley JG, Boehm EA, Blair E, Rajagopalan B, Blamire AM, Styles P, McKenna WJ, Ostman-Smith I, Clarke K, Watkins H . Hypertrophic cardiomyopathy due to sarcomeric gene mutations is characterized by impaired energy metabolism irrespective of the degree of hypertrophy.J Am Coll Cardiol. 2003; 41:1776–1782.CrossrefMedlineGoogle Scholar
    • 113. Hoskins AC, Jacques A, Bardswell SC, McKenna WJ, Tsang V, dos Remedios CG, Ehler E, Adams K, Jalilzadeh S, Avkiran M, Watkins H, Redwood C, Marston SB, Kentish JC . Normal passive viscoelasticity but abnormal myofibrillar force generation in human hypertrophic cardiomyopathy.J Mol Cell Cardiol. 2010; 49:737–745. doi: 10.1016/j.yjmcc.2010.06.006.CrossrefMedlineGoogle Scholar
    • 114. Gupte TM, Haque F, Gangadharan B, Sunitha MS, Mukherjee S, Anandhan S, Rani DS, Mukundan N, Jambekar A, Thangaraj K, Sowdhamini R, Sommese RF, Nag S, Spudich JA, Mercer JA . Mechanistic heterogeneity in contractile properties of α-tropomyosin (TPM1) mutants associated with inherited cardiomyopathies.J Biol Chem. 2015; 290:7003–7015. doi: 10.1074/jbc.M114.596676.CrossrefMedlineGoogle Scholar
    • 115. Cordero-Reyes AM, Youker K, Hamilton DJ, Torre-Amione G, Marian AJ, Nagueh SF . Molecular, cellular, and functional characterization of myocardial regions in hypertrophic cardiomyopathy.Circ Cardiovasc Imaging. 2012; 5:419–422. doi: 10.1161/CIRCIMAGING.112.972802.LinkGoogle Scholar
    • 116. Kraft T, Witjas-Paalberends ER, Boontje NM, Tripathi S, Brandis A, Montag J, Hodgkinson JL, Francino A, Navarro-Lopez F, Brenner B, Stienen GJ, van der Velden J . Familial hypertrophic cardiomyopathy: functional effects of myosin mutation R723G in cardiomyocytes.J Mol Cell Cardiol. 2013; 57:13–22. doi: 10.1016/j.yjmcc.2013.01.001.CrossrefMedlineGoogle Scholar
    • 117. Sequeira V, Wijnker PJ, Nijenkamp LL, et al . Perturbed length-dependent activation in human hypertrophic cardiomyopathy with missense sarcomeric gene mutations.Circ Res. 2013; 112:1491–1505. doi: 10.1161/CIRCRESAHA.111.300436.LinkGoogle Scholar
    • 118. Li RK, Li G, Mickle DA, Weisel RD, Merante F, Luss H, Rao V, Christakis GT, Williams WG . Overexpression of transforming growth factor-beta1 and insulin-like growth factor-I in patients with idiopathic hypertrophic cardiomyopathy.Circulation. 1997; 96:874–881.LinkGoogle Scholar
    • 119. Helms AS, Alvarado FJ, Yob J, Tang VT, Pagani F, Russell MW, Valdivia HH, Day SM . Genotype-dependent and -independent calcium signaling dysregulation in human hypertrophic cardiomyopathy.Circulation. 2016; 134:1738–1748. doi: 10.1161/CIRCULATIONAHA.115.020086.LinkGoogle Scholar
    • 120. Ruggiero A, Chen SN, Lombardi R, Rodriguez G, Marian AJ . Pathogenesis of hypertrophic cardiomyopathy caused by myozenin 2 mutations is independent of calcineurin activity.Cardiovasc Res. 2013; 97:44–54. doi: 10.1093/cvr/cvs294.CrossrefMedlineGoogle Scholar
    • 121. Fatkin D, McConnell BK, Mudd JO, Semsarian C, Moskowitz IG, Schoen FJ, Giewat M, Seidman CE, Seidman JG . An abnormal Ca(2+) response in mutant sarcomere protein-mediated familial hypertrophic cardiomyopathy.J Clin Invest. 2000; 106:1351–1359. doi: 10.1172/JCI11093.CrossrefMedlineGoogle Scholar
    • 122. Teekakirikul P, Eminaga S, Toka O, et al . Cardiac fibrosis in mice with hypertrophic cardiomyopathy is mediated by non-myocyte proliferation and requires Tgf-β.J Clin Invest. 2010; 120:3520–3529. doi: 10.1172/JCI42028.CrossrefMedlineGoogle Scholar
    • 123. Senthil V, Chen SN, Tsybouleva N, Halder T, Nagueh SF, Willerson JT, Roberts R, Marian AJ . Prevention of cardiac hypertrophy by atorvastatin in a transgenic rabbit model of human hypertrophic cardiomyopathy.Circ Res. 2005; 97:285–292. doi: 10.1161/01.RES.0000177090.07296.ac.LinkGoogle Scholar
    • 124. Patel R, Nagueh SF, Tsybouleva N, Abdellatif M, Lutucuta S, Kopelen HA, Quinones MA, Zoghbi WA, Entman ML, Roberts R, Marian AJ . Simvastatin induces regression of cardiac hypertrophy and fibrosis and improves cardiac function in a transgenic rabbit model of human hypertrophic cardiomyopathy.Circulation. 2001; 104:317–324.LinkGoogle Scholar
    • 125. Kuster DW, Mulders J, Ten Cate FJ, Michels M, Dos Remedios CG, da Costa Martins PA, van der Velden J, Oudejans CB . MicroRNA transcriptome profiling in cardiac tissue of hypertrophic cardiomyopathy patients with MYBPC3 mutations.J Mol Cell Cardiol. 2013; 65:59–66. doi: 10.1016/j.yjmcc.2013.09.012.CrossrefMedlineGoogle Scholar
    • 126. Leptidis S, El Azzouzi H, Lok SI, de Weger R, Olieslagers S, Olieslagers S, Kisters N, Silva GJ, Heymans S, Cuppen E, Berezikov E, De Windt LJ, da Costa Martins P . A deep sequencing approach to uncover the miRNOME in the human heart.PLoS One. 2013; 8:e57800. doi: 10.1371/journal.pone.0057800.CrossrefMedlineGoogle Scholar
    • 127. Yang W, Li Y, He F, Wu H . Microarray profiling of long non-coding RNA (lncRNA) associated with hypertrophic cardiomyopathy.BMC Cardiovasc Disord. 2015; 15:62. doi: 10.1186/s12872-015-0056-7.CrossrefMedlineGoogle Scholar
    • 128. Christodoulou DC, Wakimoto H, Onoue K, et al . 5’RNA-Seq identifies Fhl1 as a genetic modifier in cardiomyopathy.J Clin Invest. 2014; 124:1364–1370. doi: 10.1172/JCI70108.CrossrefMedlineGoogle Scholar
    • 129. Sasagawa S, Nishimura Y, Okabe S, Murakami S, Ashikawa Y, Yuge M, Kawaguchi K, Kawase R, Okamoto R, Ito M, Tanaka T . Downregulation of GSTK1 is a common mechanism underlying hypertrophic cardiomyopathy.Front Pharmacol. 2016; 7:162. doi: 10.3389/fphar.2016.00162.CrossrefMedlineGoogle Scholar
    • 130. Marian AJ . Pathogenesis of diverse clinical and pathological phenotypes in hypertrophic cardiomyopathy.Lancet. 2000; 355:58–60.CrossrefMedlineGoogle Scholar
    • 131. Lan F, Lee AS, Liang P, et al . Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells.Cell Stem Cell. 2013; 12:101–113. doi: 10.1016/j.stem.2012.10.010.CrossrefMedlineGoogle Scholar
    • 132. Watkins H, McKenna WJ, Thierfelder L, Suk HJ, Anan R, O’Donoghue A, Spirito P, Matsumori A, Moravec CS, Seidman JG . Mutations in the genes for cardiac troponin T and alpha-tropomyosin in hypertrophic cardiomyopathy.N Engl J Med. 1995; 332:1058–1064. doi: 10.1056/NEJM199504203321603.CrossrefMedlineGoogle Scholar
    • 133. Watkins H, Rosenzweig A, Hwang DS, Levi T, McKenna W, Seidman CE, Seidman JG . Characteristics and prognostic implications of myosin missense mutations in familial hypertrophic cardiomyopathy.N Engl J Med. 1992; 326:1108–1114. doi: 10.1056/NEJM199204233261703.CrossrefMedlineGoogle Scholar
    • 134. Marian AJ, Mares A, Kelly DP, Yu QT, Abchee AB, Hill R, Roberts R . Sudden cardiac death in hypertrophic cardiomyopathy. Variability in phenotypic expression of beta-myosin heavy chain mutations.Eur Heart J. 1995; 16:368–376.CrossrefMedlineGoogle Scholar
    • 135. Lopes LR, Rahman MS, Elliott PM . A systematic review and meta-analysis of genotype-phenotype associations in patients with hypertrophic cardiomyopathy caused by sarcomeric protein mutations.Heart. 2013; 99:1800–1811. doi: 10.1136/heartjnl-2013-303939.CrossrefMedlineGoogle Scholar
    • 136. Marian AJ . On genetic and phenotypic variability of hypertrophic cardiomyopathy: nature versus nurture.J Am Coll Cardiol. 2001; 38:331–334.CrossrefMedlineGoogle Scholar
    • 137. Coppini R, Ho CY, Ashley E, Day S, Ferrantini C, Girolami F, Tomberli B, Bardi S, Torricelli F, Cecchi F, Mugelli A, Poggesi C, Tardiff J, Olivotto I . Clinical phenotype and outcome of hypertrophic cardiomyopathy associated with thin-filament gene mutations.J Am Coll Cardiol. 2014; 64:2589–2600. doi: 10.1016/j.jacc.2014.09.059.CrossrefMedlineGoogle Scholar
    • 138. Daw EW, Chen SN, Czernuszewicz G, Lombardi R, Lu Y, Ma J, Roberts R, Shete S, Marian AJ . Genome-wide mapping of modifier chromosomal loci for human hypertrophic cardiomyopathy.Hum Mol Genet. 2007; 16:2463–2471. doi: 10.1093/hmg/ddm202.CrossrefMedlineGoogle Scholar
    • 139. Tiret L, Rigat B, Visvikis S, Breda C, Corvol P, Cambien F, Soubrier F . Evidence, from combined segregation and linkage analysis, that a variant of the angiotensin I-converting enzyme (ACE) gene controls plasma ACE levels.Am J Hum Genet. 1992; 51:197–205.MedlineGoogle Scholar
    • 140. Lechin M, Quiñones MA, Omran A, Hill R, Yu QT, Rakowski H, Wigle D, Liew CC, Sole M, Roberts R . Angiotensin-I converting enzyme genotypes and left ventricular hypertrophy in patients with hypertrophic cardiomyopathy.Circulation. 1995; 92:1808–1812.LinkGoogle Scholar
    • 141. Marian AJ, Yu QT, Workman R, Greve G, Roberts R . Angiotensin-converting enzyme polymorphism in hypertrophic cardiomyopathy and sudden cardiac death.Lancet. 1993; 342:1085–1086.CrossrefMedlineGoogle Scholar
    • 142. Kaufman BD, Auerbach S, Reddy S, Manlhiot C, Deng L, Prakash A, Printz BF, Gruber D, Papavassiliou DP, Hsu DT, Sehnert AJ, Chung WK, Mital S . RAAS gene polymorphisms influence progression of pediatric hypertrophic cardiomyopathy.Hum Genet. 2007; 122:515–523. doi: 10.1007/s00439-007-0429-9.CrossrefMedlineGoogle Scholar
    • 143. Mouton JM, van der Merwe L, Goosen A, Revera M, Brink PA, Moolman-Smook JC, Kinnear C . MYBPH acts as modifier of cardiac hypertrophy in hypertrophic cardiomyopathy (HCM) patients.Hum Genet. 2016; 135:477–483. doi: 10.1007/s00439-016-1649-7.CrossrefMedlineGoogle Scholar
    • 144. Brugada R, Kelsey W, Lechin M, Zhao G, Yu QT, Zoghbi W, Quinones M, Elstein E, Omran A, Rakowski H, Wigle D, Liew CC, Sole M, Roberts R, Marian AJ . Role of candidate modifier genes on the phenotypic expression of hypertrophy in patients with hypertrophic cardiomyopathy.J Investig Med. 1997; 45:542–551.MedlineGoogle Scholar
    • 145. Fujino N, Shimizu M, Ino H, Yamaguchi M, Yasuda T, Nagata M, Konno T, Mabuchi H . A novel mutation Lys273Glu in the cardiac troponin T gene shows high degree of penetrance and transition from hypertrophic to dilated cardiomyopathy.Am J Cardiol. 2002; 89:29–33.CrossrefMedlineGoogle Scholar
    • 146. Mogensen J, Kubo T, Duque M, Uribe W, Shaw A, Murphy R, Gimeno JR, Elliott P, McKenna WJ . Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin I mutations.J Clin Invest. 2003; 111:209–216. doi: 10.1172/JCI16336.CrossrefMedlineGoogle Scholar
    • 147. Hoedemaekers YM, Caliskan K, Majoor-Krakauer D, van de Laar I, Michels M, Witsenburg M, ten Cate FJ, Simoons ML, Dooijes D . Cardiac beta-myosin heavy chain defects in two families with non-compaction cardiomyopathy: linking non-compaction to hypertrophic, restrictive, and dilated cardiomyopathies.Eur Heart J. 2007; 28:2732–2737. doi: 10.1093/eurheartj/ehm429.CrossrefMedlineGoogle Scholar
    • 148. Dellefave LM, Pytel P, Mewborn S, Mora B, Guris DL, Fedson S, Waggoner D, Moskowitz I, McNally EM . Sarcomere mutations in cardiomyopathy with left ventricular hypertrabeculation.Circ Cardiovasc Genet. 2009; 2:442–449. doi: 10.1161/CIRCGENETICS.109.861955.LinkGoogle Scholar
    • 149. Kamisago M, Sharma SD, DePalma SR, Solomon S, Sharma P, McDonough B, Smoot L, Mullen MP, Woolf PK, Wigle ED, Seidman JG, Seidman CE . Mutations in sarcomere protein genes as a cause of dilated cardiomyopathy.New Eng J Med. 2000; 343:1688–1696.CrossrefMedlineGoogle Scholar
    • 150. Sommese RF, Nag S, Sutton S, Miller SM, Spudich JA, Ruppel KM . Effects of troponin T cardiomyopathy mutations on the calcium sensitivity of the regulated thin filament and the actomyosin cross-bridge kinetics of human β-cardiac myosin.PLoS One. 2013; 8:e83403. doi: 10.1371/journal.pone.0083403.CrossrefMedlineGoogle Scholar
    • 151. Hershberger RE, Pinto JR, Parks SB, Kushner JD, Li D, Ludwigsen S, Cowan J, Morales A, Parvatiyar MS, Potter JD . Clinical and functional characterization of TNNT2 mutations identified in patients with dilated cardiomyopathy.Circ Cardiovasc Genet. 2009; 2:306–313. doi: 10.1161/CIRCGENETICS.108.846733.LinkGoogle Scholar
    • 152. Jagatheesan G, Rajan S, Schulz EM, Ahmed RP, Petrashevskaya N, Schwartz A, Boivin GP, Arteaga GM, Wang T, Wang YG, Ashraf M, Liggett SB, Lorenz J, Solaro RJ, Wieczorek DF . An internal domain of beta-tropomyosin increases myofilament Ca(2+) sensitivity.Am J Physiol Heart Circ Physiol. 2009; 297:H181–H190. doi: 10.1152/ajpheart.00329.2008.CrossrefMedlineGoogle Scholar
    • 153. Harada K, Potter JD . Familial hypertrophic cardiomyopathy mutations from different functional regions of troponin T result in different effects on the pH and Ca2+ sensitivity of cardiac muscle contraction.J Biol Chem. 2004; 279:14488–14495. doi: 10.1074/jbc.M309355200.CrossrefMedlineGoogle Scholar
    • 154. Revera M, van der Merwe L, Heradien M, Goosen A, Corfield VA, Brink PA, Moolman-Smook JC . Troponin T and beta-myosin mutations have distinct cardiac functional effects in hypertrophic cardiomyopathy patients without hypertrophy.Cardiovasc Res. 2008; 77:687–694. doi: 10.1093/cvr/cvm075.CrossrefMedlineGoogle Scholar
    • 155. Moore JR, Leinwand L, Warshaw DM . Understanding cardiomyopathy phenotypes based on the functional impact of mutations in the myosin motor.Circ Res. 2012; 111:375–385. doi: 10.1161/CIRCRESAHA.110.223842.LinkGoogle Scholar
    • 156. Davis J, Davis LC, Correll RN, et al . A tension-based model distinguishes hypertrophic versus dilated cardiomyopathy.Cell. 2016; 165:1147–1159. doi: 10.1016/j.cell.2016.04.002.CrossrefMedlineGoogle Scholar
    • 157. Jan MF, Tajik AJ . Modern imaging techniques in cardiomyopathies.Circ Res. 2017; 121:874–891. doi: 10.1161/CIRCRESAHA.117.309600.LinkGoogle Scholar
    • 158. Braunwald E, Lambrew CT, Rockoff SD, Ross J, Morrow AG . Idiopathic hypertrophic subaortic stenosis. I. A description of the disease based upon an analysis of 64 patients.Circulation. 1964; 30(suppl 4):3–119.Google Scholar
    • 159. Henry WL, Clark CE, Epstein SE . Asymmetric septal hypertrophy. Echocardiographic identification of the pathognomonic anatomic abnormality of IHSS.Circulation. 1973; 47:225–233.LinkGoogle Scholar
    • 160. Maron MS, Olivotto I, Harrigan C, Appelbaum E, Gibson CM, Lesser JR, Haas TS, Udelson JE, Manning WJ, Maron BJ . Mitral valve abnormalities identified by cardiovascular magnetic resonance represent a primary phenotypic expression of hypertrophic cardiomyopathy.Circulation. 2011; 124:40–47. doi: 10.1161/CIRCULATIONAHA.110.985812.LinkGoogle Scholar
    • 161. Malik R, Maron MS, Rastegar H, Pandian NG . Hypertrophic cardiomyopathy with right ventricular outflow tract and left ventricular intracavitary obstruction.Echocardiography. 2014; 31:682–685. doi: 10.1111/echo.12543.CrossrefMedlineGoogle Scholar
    • 162. Teare D . Asymmetrical hypertrophy of the heart in young adults.Br Heart J. 1958; 20:1–8.CrossrefMedlineGoogle Scholar
    • 163. Davies MJ, McKenna WJ . Hypertrophic cardiomyopathy–pathology and pathogenesis.Histopathology. 1995; 26:493–500.CrossrefMedlineGoogle Scholar
    • 164. Davies MJ . The current status of myocardial disarray in hypertrophic cardiomyopathy.Br Heart J. 1984; 51:361–363.CrossrefMedlineGoogle Scholar
    • 165. Varnava AM, Elliott PM, Baboonian C, Davison F, Davies MJ, McKenna WJ . Hypertrophic cardiomyopathy: histopathological features of sudden death in cardiac troponin T disease.Circulation. 2001; 104:1380–1384.LinkGoogle Scholar
    • 166. McGill LA, Ismail TF, Nielles-Vallespin S, et al . Reproducibility of in-vivo diffusion tensor cardiovascular magnetic resonance in hypertrophic cardiomyopathy.J Cardiovasc Magn Reson. 2012; 14:86. doi: 10.1186/1532-429X-14-86.CrossrefMedlineGoogle Scholar
    • 167. Almaas VM, Haugaa KH, Strøm EH, Scott H, Dahl CP, Leren TP, Geiran OR, Endresen K, Edvardsen T, Aakhus S, Amlie JP . Increased amount of interstitial fibrosis predicts ventricular arrhythmias, and is associated with reduced myocardial septal function in patients with obstructive hypertrophic cardiomyopathy.Europace. 2013; 15:1319–1327. doi: 10.1093/europace/eut028.CrossrefMedlineGoogle Scholar
    • 168. McLELLAN AJ, Ellims AH, Prabhu S, Voskoboinik A, Iles LM, Hare JL, Kaye DM, Macciocca I, Mariani JA, Kalman JM, Taylor AJ, Kistler PM . Diffuse ventricular fibrosis on cardiac magnetic resonance imaging associates with ventricular tachycardia in patients with hypertrophic cardiomyopathy.J Cardiovasc Electrophysiol. 2016; 27:571–580. doi: 10.1111/jce.12948.CrossrefMedlineGoogle Scholar
    • 169. Briasoulis A, Mallikethi-Reddy S, Palla M, Alesh I, Afonso L . Myocardial fibrosis on cardiac magnetic resonance and cardiac outcomes in hypertrophic cardiomyopathy: a meta-analysis.Heart. 2015; 101:1406–1411. doi: 10.1136/heartjnl-2015-307682.CrossrefMedlineGoogle Scholar
    • 170. O’Hanlon R, Grasso A, Roughton M, et al . Prognostic significance of myocardial fibrosis in hypertrophic cardiomyopathy.J Am Coll Cardiol. 2010; 56:867–874. doi: 10.1016/j.jacc.2010.05.010.CrossrefMedlineGoogle Scholar
    • 171. Schelbert EB, Piehler KM, Zareba KM, Moon JC, Ugander M, Messroghli DR, Valeti US, Chang CC, Shroff SG, Diez J, Miller CA, Schmitt M, Kellman P, Butler J, Gheorghiade M, Wong TC . Myocardial fibrosis quantified by extracellular volume is associated with subsequent hospitalization for heart failure, death, or both across the spectrum of ejection fraction and heart failure stage.J Am Heart Assoc. 2015; 4.LinkGoogle Scholar
    • 172. Maron BJ, Wolfson JK, Epstein SE, Roberts WC . Intramural (“small vessel”) coronary artery disease in hypertrophic cardiomyopathy.J Am Coll Cardiol. 1986; 8:545–557.CrossrefMedlineGoogle Scholar
    • 173. Lombardi R, Betocchi S, Losi MA, Tocchetti CG, Aversa M, Miranda M, D’Alessandro G, Cacace A, Ciampi Q, Chiariello M . Myocardial collagen turnover in hypertrophic cardiomyopathy.Circulation. 2003; 108:1455–1460. doi: 10.1161/01.CIR.0000090687.97972.10.LinkGoogle Scholar
    • 174. Ho CY, López B, Coelho-Filho OR, Lakdawala NK, Cirino AL, Jarolim P, Kwong R, González A, Colan SD, Seidman JG, Díez J, Seidman CE . Myocardial fibrosis as an early manifestation of hypertrophic cardiomyopathy.N Engl J Med. 2010; 363:552–563. doi: 10.1056/NEJMoa1002659.CrossrefMedlineGoogle Scholar
    • 175. Ellims AH, Taylor AJ, Mariani JA, Ling LH, Iles LM, Maeder MT, Kaye DM . Evaluating the utility of circulating biomarkers of collagen synthesis in hypertrophic cardiomyopathy.Circ Heart Fail. 2014; 7:271–278. doi: 10.1161/CIRCHEARTFAILURE.113.000665.LinkGoogle Scholar
    • 176. Kuusisto J, Kärjä V, Sipola P, Kholová I, Peuhkurinen K, Jääskeläinen P, Naukkarinen A, Ylä-Herttuala S, Punnonen K, Laakso M . Low-grade inflammation and the phenotypic expression of myocardial fibrosis in hypertrophic cardiomyopathy.Heart. 2012; 98:1007–1013. doi: 10.1136/heartjnl-2011-300960.CrossrefMedlineGoogle Scholar
    • 177. Zen K, Irie H, Doue T, Takamiya M, Yamano T, Sawada T, Azuma A, Matsubara H . Analysis of circulating apoptosis mediators and proinflammatory cytokines in patients with idiopathic hypertrophic cardiomyopathy: comparison between nonobstructive and dilated-phase hypertrophic cardiomyopathy.Int Heart J. 2005; 46:231–244.CrossrefMedlineGoogle Scholar
    • 178. Moreno V, Hernández-Romero D, Vilchez JA, García-Honrubia A, Cambronero F, Casas T, González J, Martínez P, Climent V, de la Morena G, Valdés M, Marín F . Serum levels of high-sensitivity troponin T: a novel marker for cardiac remodeling in hypertrophic cardiomyopathy.J Card Fail. 2010; 16:950–956. doi: 10.1016/j.cardfail.2010.07.245.CrossrefMedlineGoogle Scholar
    • 179. Fang L, Ellims AH, Moore XL, White DA, Taylor AJ, Chin-Dusting J, Dart AM . Circulating microRNAs as biomarkers for diffuse myocardial fibrosis in patients with hypertrophic cardiomyopathy.J Transl Med. 2015; 13:314. doi: 10.1186/s12967-015-0672-0.CrossrefMedlineGoogle Scholar
    • 180. Derda AA, Thum S, Lorenzen JM, Bavendiek U, Heineke J, Keyser B, Stuhrmann M, Givens RC, Kennel PJ, Schulze PC, Widder JD, Bauersachs J, Thum T . Blood-based microRNA signatures differentiate various forms of cardiac hypertrophy.Int J Cardiol. 2015; 196:115–122. doi: 10.1016/j.ijcard.2015.05.185.CrossrefMedlineGoogle Scholar
    • 181. Roncarati R, Viviani Anselmi C, Losi MA, et al . Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy.J Am Coll Cardiol. 2014; 63:920–927. doi: 10.1016/j.jacc.2013.09.041.CrossrefMedlineGoogle Scholar
    • 182. Brock R . Functional obstruction of the left ventricle; acquired aortic subvalvar stenosis.Guys Hosp Rep. 1957; 106:221–238.MedlineGoogle Scholar
    • 183. BROCK R . Functional obstruction of the left ventricle (acquired aortic subvalvar stenosis).Guys Hosp Rep. 1959; 108:126–143.MedlineGoogle Scholar
    • 184. Morrow AG, Braunwald E . Functional aortic stenosis; a malformation characterized by resistance to left ventricular outflow without anatomic obstruction.Circulation. 1959; 20:181–189.LinkGoogle Scholar
    • 185. Maron MS, Olivotto I, Zenovich AG, Link MS, Pandian NG, Kuvin JT, Nistri S, Cecchi F, Udelson JE, Maron BJ . Hypertrophic cardiomyopathy is predominantly a disease of left ventricular outflow tract obstruction.Circulation. 2006; 114:2232–2239. doi: 10.1161/CIRCULATIONAHA.106.644682.LinkGoogle Scholar
    • 186. Shah JS, Esteban MT, Thaman R, Sharma R, Mist B, Pantazis A, Ward D, Kohli SK, Page SP, Demetrescu C, Sevdalis E, Keren A, Pellerin D, McKenna WJ, Elliott PM . Prevalence of exercise-induced left ventricular outflow tract obstruction in symptomatic patients with non-obstructive hypertrophic cardiomyopathy.Heart. 2008; 94:1288–1294. doi: 10.1136/hrt.2007.126003.CrossrefMedlineGoogle Scholar
    • 187. Braunwald E, Oldham HN, Ross J, Linhart JW, Mason DT, Fort L The circulatory response of patients with idiopathic hypertrophic subaortic stenosis to nitroglycerin and to the valsalva maneuver.Circulation. 1964; 29:422–431.LinkGoogle Scholar
    • 188. Braunwald E, Ebert PA . Hemogynamic alterations in idiopathic hypertrophic subaortic stenosis induced by sympathomimetic drugs.Am J Cardiol. 1962; 10:489–495.CrossrefMedlineGoogle Scholar
    • 189. Stewart S, Mason DT, Braunwald E . Impaired rate of left ventricular filling in idiopathic hypertrophic subaortic stenosis and valvular aortic stenosis.Circulation. 1968; 37:8–14.LinkGoogle Scholar
    • 190. Guttmann OP, Pavlou M, O’Mahony C, Monserrat L, Anastasakis A, Rapezzi C, Biagini E, Gimeno JR, Limongelli G, Garcia-Pavia P, McKenna WJ, Omar RZ, Elliott PM ; Hypertrophic Cardiomyopathy Outcomes Investigators. Prediction of thrombo-embolic risk in patients with hypertrophic cardiomyopathy (HCM Risk-CVA).Eur J Heart Fail. 2015; 17:837–845. doi: 10.1002/ejhf.316.CrossrefMedlineGoogle Scholar
    • 191. Nagueh SF, McFalls J, Meyer D, Hill R, Zoghbi WA, Tam JW, Quiñones MA, Roberts R, Marian AJ . Tissue Doppler imaging predicts the development of hypertrophic cardiomyopathy in subjects with subclinical disease.Circulation. 2003; 108:395–398. doi: 10.1161/01.CIR.0000084500.72232.8D.LinkGoogle Scholar
    • 192. Kramer CM, Reichek N, Ferrari VA, Theobald T, Dawson J, Axel L . Regional heterogeneity of function in hypertrophic cardiomyopathy.Circulation. 1994; 90:186–194.LinkGoogle Scholar
    • 193. Urbano-Moral JA, Rowin EJ, Maron MS, Crean A, Pandian NG . Investigation of global and regional myocardial mechanics with 3-dimensional speckle tracking echocardiography and relations to hypertrophy and fibrosis in hypertrophic cardiomyopathy.Circ Cardiovasc Imaging. 2014; 7:11–19. doi: 10.1161/CIRCIMAGING.113.000842.LinkGoogle Scholar
    • 194. Sen-Chowdhry S, Jacoby D, Moon JC, McKenna WJ . Update on hypertrophic cardiomyopathy and a guide to the guidelines.Nat Rev Cardiol. 2016; 13:651–675. doi: 10.1038/nrcardio.2016.140.CrossrefMedlineGoogle Scholar
    • 195. Biagini E, Spirito P, Rocchi G, Ferlito M, Rosmini S, Lai F, Lorenzini M, Terzi F, Bacchi-Reggiani L, Boriani G, Branzi A, Boni L, Rapezzi C . Prognostic implications of the Doppler restrictive filling pattern in hypertrophic cardiomyopathy.Am J Cardiol. 2009; 104:1727–1731. doi: 10.1016/j.amjcard.2009.07.057.CrossrefMedlineGoogle Scholar
    • 196. Braunwald E, Aygen MM . Idiopathic myocardial hypertrophy without congestive heart failure or obstruction to blood flow. Clinical, hemodynamic and angiocardiographic studies in fourteen patients.Am J Med. 1963; 35:7–19.CrossrefMedlineGoogle Scholar
    • 197. Maron BJ, Rowin EJ, Maron MS, Braunwald E . Nonobstructive hypertrophic cardiomyopathy out of the shadows: known from the beginning but largely ignored … until now.Am J Med. 2017; 130:119–123. doi: 10.1016/j.amjmed.2016.09.015.CrossrefMedlineGoogle Scholar
    • 198. Maron MS, Rowin EJ, Olivotto I, Casey SA, Arretini A, Tomberli B, Garberich RF, Link MS, Chan RH, Lesser JR, Maron BJ . Contemporary natural history and management of nonobstructive hypertrophic cardiomyopathy.J Am Coll Cardiol. 2016; 67:1399–1409. doi: 10.1016/j.jacc.2016.01.023.CrossrefMedlineGoogle Scholar
    • 199. Monserrat L, Elliott PM, Gimeno JR, Sharma S, Penas-Lado M, McKenna WJ . Non-sustained ventricular tachycardia in hypertrophic cardiomyopathy: an independent marker of sudden death risk in young patients.J Am Coll Cardiol. 2003; 42:873–879.CrossrefMedlineGoogle Scholar
    • 200. Adabag AS, Casey SA, Kuskowski MA, Zenovich AG, Maron BJ . Spectrum and prognostic significance of arrhythmias on ambulatory Holter electrocardiogram in hypertrophic cardiomyopathy.J Am Coll Cardiol. 2005; 45:697–704. doi: 10.1016/j.jacc.2004.11.043.CrossrefMedlineGoogle Scholar
    • 201. Elliott PM, Poloniecki J, Dickie S, Sharma S, Monserrat L, Varnava A, Mahon NG, McKenna WJ . Sudden death in hypertrophic cardiomyopathy: identification of high risk patients.J Am Coll Cardiol. 2000; 36:2212–2218.CrossrefMedlineGoogle Scholar
    • 202. Christiaans I, van Engelen K, van Langen IM, Birnie E, Bonsel GJ, Elliott PM, Wilde AA . Risk stratification for sudden cardiac death in hypertrophic cardiomyopathy: systematic review of clinical risk markers.Europace. 2010; 12:313–321. doi: 10.1093/europace/eup431.CrossrefMedlineGoogle Scholar
    • 203. O’Mahony C, Jichi F, Pavlou M, Monserrat L, Anastasakis A, Rapezzi C, Biagini E, Gimeno JR, Limongelli G, McKenna WJ, Omar RZ, Elliott PM ; Hypertrophic Cardiomyopathy Outcomes Investigators. A novel clinical risk prediction model for sudden cardiac death in hypertrophic cardiomyopathy (HCM risk-SCD).Eur Heart J. 2014; 35:2010–2020. doi: 10.1093/eurheartj/eht439.CrossrefMedlineGoogle Scholar
    • 204. Nienaber CA, Hiller S, Spielmann RP, Geiger M, Kuck KH . Syncope in hypertrophic cardiomyopathy: multivariate analysis of prognostic determinants.J Am Coll Cardiol. 1990; 15:948–955.CrossrefMedlineGoogle Scholar
    • 205. Olivotto I, Cecchi F, Casey SA, Dolara A, Traverse JH, Maron BJ . Impact of atrial fibrillation on the clinical course of hypertrophic cardiomyopathy.Circulation. 2001; 104:2517–2524.LinkGoogle Scholar
    • 206. Siontis KC, Geske JB, Ong K, Nishimura RA, Ommen SR, Gersh BJ . Atrial fibrillation in hypertrophic cardiomyopathy: prevalence, clinical correlations, and mortality in a large high-risk population.J Am Heart Assoc. 2014; 3:e001002. doi: 10.1161/JAHA.114.001002.LinkGoogle Scholar
    • 207. Debonnaire P, Joyce E, Hiemstra Y, Mertens BJ, Atsma DE, Schalij MJ, Bax JJ, Delgado V, Marsan NA . Left atrial size and function in hypertrophic cardiomyopathy patients and risk of new-onset atrial fibrillation.Circ Arrhythm Electrophysiol. 2017; 10.LinkGoogle Scholar
    • 208. Guttmann OP, Rahman MS, O’Mahony C, Anastasakis A, Elliott PM . Atrial fibrillation and thromboembolism in patients with hypertrophic cardiomyopathy: systematic review.Heart. 2014; 100:465–472. doi: 10.1136/heartjnl-2013-304276.CrossrefMedlineGoogle Scholar
    • 209. Eriksson MJ, Sonnenberg B, Woo A, Rakowski P, Parker TG, Wigle ED, Rakowski H . Long-term outcome in patients with apical hypertrophic cardiomyopathy.J Am Coll Cardiol. 2002; 39:638–645.CrossrefMedlineGoogle Scholar
    • 210. Ho CY, McMurray JJV, Cirino AL, Colan SD, Day SM, Desai AS, Lipshultz SE, MacRae CA, Shi L, Solomon SD, Orav EJ, Braunwald E ; for the VANISH trial investigators and executive committee. Valsartan for attenuating disease evolution in early sarcomeric hypertrophic cardiomyopathy: the design of the VANISH trial.Am Heart J. 2017; 187:145–155. doi: 10.1016/j.ahj.2017.02.008.CrossrefMedlineGoogle Scholar
    • 211. Grazioli G, Usín D, Trucco E, Sanz M, Montserrat S, Vidal B, Gutierrez J, Canal R, Brugada J, Mont L, Sitges M . Differentiating hypertrophic cardiomyopathy from athlete’s heart: an electrocardiographic and echocardiographic approach.J Electrocardiol. 2016; 49:539–544. doi: 10.1016/j.jelectrocard.2016.03.005.CrossrefMedlineGoogle Scholar
    • 212. Maron BJ, Doerer JJ, Haas TS, Tierney DM, Mueller FO . Sudden deaths in young competitive athletes: analysis of 1866 deaths in the United States, 1980-2006.Circulation. 2009; 119:1085–1092. doi: 10.1161/CIRCULATIONAHA.108.804617.LinkGoogle Scholar
    • 213. Maron BJ, Shirani J, Poliac LC, Mathenge R, Roberts WC, Mueller FO . Sudden death in young competitive athletes. Clinical, demographic, and pathological profiles.JAMA. 1996; 276:199–204.CrossrefMedlineGoogle Scholar
    • 214. Caselli S, Maron MS, Urbano-Moral JA, Pandian NG, Maron BJ, Pelliccia A . Differentiating left ventricular hypertrophy in athletes from that in patients with hypertrophic cardiomyopathy.Am J Cardiol. 2014; 114:1383–1389. doi: 10.1016/j.amjcard.2014.07.070.CrossrefMedlineGoogle Scholar
    • 215. Butz T, van Buuren F, Mellwig KP, Langer C, Plehn G, Meissner A, Trappe HJ, Horstkotte D, Faber L . Two-dimensional strain analysis of the global and regional myocardial function for the differentiation of pathologic and physiologic left ventricular hypertrophy: a study in athletes and in patients with hypertrophic cardiomyopathy.Int J Cardiovasc Imaging. 2011; 27:91–100. doi: 10.1007/s10554-010-9665-5.CrossrefMedlineGoogle Scholar
    • 216. Petersen SE, Selvanayagam JB, Francis JM, Myerson SG, Wiesmann F, Robson MD, Ostman-Smith I, Casadei B, Watkins H, Neubauer S . Differentiation of athlete’s heart from pathological forms of cardiac hypertrophy by means of geometric indices derived from cardiovascular magnetic resonance.J Cardiovasc Magn Reson. 2005; 7:551–558.CrossrefMedlineGoogle Scholar
    • 217. Sheikh N, Papadakis M, Ghani S, Zaidi A, Gati S, Adami PE, Carré F, Schnell F, Wilson M, Avila P, McKenna W, Sharma S . Comparison of electrocardiographic criteria for the detection of cardiac abnormalities in elite black and white athletes.Circulation. 2014; 129:1637–1649. doi: 10.1161/CIRCULATIONAHA.113.006179.LinkGoogle Scholar
    • 218. Kadota C, Arimura T, Hayashi T, Naruse TK, Kawai S, Kimura A . Screening of sarcomere gene mutations in young athletes with abnormal findings in electrocardiography: identification of a MYH7 mutation and MYBPC3 mutations.J Hum Genet. 2015; 60:641–645. doi: 10.1038/jhg.2015.81.CrossrefMedlineGoogle Scholar
    • 219. Bagnall RD, Weintraub RG, Ingles J, et al . A prospective study of sudden cardiac death among children and young adults.N Engl J Med. 2016; 374:2441–2452. doi: 10.1056/NEJMoa1510687.CrossrefMedlineGoogle Scholar
    • 220. Maron BJ, Rowin EJ, Casey SA, Link MS, Lesser JR, Chan RH, Garberich RF, Udelson JE, Maron MS . Hypertrophic cardiomyopathy in adulthood associated with low cardiovascular mortality with contemporary management strategies.J Am Coll Cardiol. 2015; 65:1915–1928. doi: 10.1016/j.jacc.2015.02.061.CrossrefMedlineGoogle Scholar
    • 221. Maron BJ, Ommen SR, Semsarian C, Spirito P, Olivotto I, Maron MS . Hypertrophic cardiomyopathy: present and future, with translation into contemporary cardiovascular medicine.J Am Coll Cardiol. 2014; 64:83–99. doi: 10.1016/j.jacc.2014.05.003.CrossrefMedlineGoogle Scholar
    • 222. Klarich KW, Attenhofer Jost CH, Binder J, Connolly HM, Scott CG, Freeman WK, Ackerman MJ, Nishimura RA, Tajik AJ, Ommen SR . Risk of death in long-term follow-up of patients with apical hypertrophic cardiomyopathy.Am J Cardiol. 2013; 111:1784–1791. doi: 10.1016/j.amjcard.2013.02.040.CrossrefMedlineGoogle Scholar
    • 223. Wilcox WR, Banikazemi M, Guffon N, Waldek S, Lee P, Linthorst GE, Desnick RJ, Germain DP ; International Fabry Disease Study Group. Long-term safety and efficacy of enzyme replacement therapy for Fabry disease.Am J Hum Genet. 2004; 75:65–74. doi: 10.1086/422366.CrossrefMedlineGoogle Scholar
    • 224. Gruner C, Ivanov J, Care M, Williams L, Moravsky G, Yang H, Laczay B, Siminovitch K, Woo A, Rakowski H . Toronto hypertrophic cardiomyopathy genotype score for prediction of a positive genotype in hypertrophic cardiomyopathy.Circ Cardiovasc Genet. 2013; 6:19–26. doi: 10.1161/CIRCGENETICS.112.963363.LinkGoogle Scholar
    • 225. Ho CY, Day SM, Colan SD, et al ; HCMNet Investigators. The burden of early phenotypes and the influence of wall thickness in hypertrophic cardiomyopathy mutation carriers: findings from the HCMNet study.JAMA Cardiol. 2017; 2:419–428. doi: 10.1001/jamacardio.2016.5670.CrossrefMedlineGoogle Scholar
    • 226. Nagueh SF, Bachinski LL, Meyer D, Hill R, Zoghbi WA, Tam JW, Quiñones MA, Roberts R, Marian AJ . Tissue Doppler imaging consistently detects myocardial abnormalities in patients with hypertrophic cardiomyopathy and provides a novel means for an early diagnosis before and independently of hypertrophy.Circulation. 2001; 104:128–130.LinkGoogle Scholar
    • 227. Harrison DC, Braunwald E, Glick G, Mason DT, Chidsey CA, Ross J Effects of beta adrenergic blockade on the circulation with particular reference to observations in patients with hypertrophic subaortic stenosis.Circulation. 1964; 29:84–98.LinkGoogle Scholar
    • 228. Cohen LS, Braunwald E . Chronic beta adrenergic receptor blockade in the treatment of idiopathic hypertrophic subaortic stenosis.Prog Cardiovasc Dis. 1968; 11:211–221.CrossrefMedlineGoogle Scholar
    • 229. Nistri S, Olivotto I, Maron MS, Ferrantini C, Coppini R, Grifoni C, Baldini K, Sgalambro A, Cecchi F, Maron BJ . β Blockers for prevention of exercise-induced left ventricular outflow tract obstruction in patients with hypertrophic cardiomyopathy.Am J Cardiol. 2012; 110:715–719. doi: 10.1016/j.amjcard.2012.04.051.CrossrefMedlineGoogle Scholar
    • 230. Flamm MD, Harrison DC, Hancock EW . Muscular subaortic stenosis. Prevention of outflow obstruction with propranolol.Circulation. 1968; 38:846–858.LinkGoogle Scholar
    • 231. Sherrid MV, Barac I, McKenna WJ, Elliott PM, Dickie S, Chojnowska L, Casey S, Maron BJ . Multicenter study of the efficacy and safety of disopyramide in obstructive hypertrophic cardiomyopathy.J Am Coll Cardiol. 2005; 45:1251–1258. doi: 10.1016/j.jacc.2005.01.012.CrossrefMedlineGoogle Scholar
    • 232. Gilligan DM, Chan WL, Joshi J, Clarke P, Fletcher A, Krikler S, Oakley CM . A double-blind, placebo-controlled crossover trial of nadolol and verapamil in mild and moderately symptomatic hypertrophic cardiomyopathy.J Am Coll Cardiol. 1993; 21:1672–1679.CrossrefMedlineGoogle Scholar
    • 233. Spirito P, Autore C, Formisano F, et al . Risk of sudden death and outcome in patients with hypertrophic cardiomyopathy with benign presentation and without risk factors.Am J Cardiol. 2014; 113:1550–1555. doi: 10.1016/j.amjcard.2014.01.435.CrossrefMedlineGoogle Scholar
    • 234. Maron BJ, Rowin EJ, Casey SA, Lesser JR, Garberich RF, McGriff DM, Maron MS . Hypertrophic cardiomyopathy in children, adolescents, and young adults associated with low cardiovascular mortality with contemporary management strategies.Circulation. 2016; 133:62–73. doi: 10.1161/CIRCULATIONAHA.115.017633.LinkGoogle Scholar
    • 235. Maron BJ, Shen WK, Link MS, Epstein AE, Almquist AK, Daubert JP, Bardy GH, Favale S, Rea RF, Boriani G, Estes NA, Spirito P . Efficacy of implantable cardioverter-defibrillators for the prevention of sudden death in patients with hypertrophic cardiomyopathy.N Engl J Med. 2000; 342:365–373. doi: 10.1056/NEJM200002103420601.CrossrefMedlineGoogle Scholar
    • 236. Maron BJ, Spirito P, Ackerman MJ, et al . Prevention of sudden cardiac death with implantable cardioverter-defibrillators in children and adolescents with hypertrophic cardiomyopathy.J Am Coll Cardiol. 2013; 61:1527–1535. doi: 10.1016/j.jacc.2013.01.037.CrossrefMedlineGoogle Scholar
    • 237. Maron BJ, Spirito P, Shen WK, et al . Implantable cardioverter-defibrillators and prevention of sudden cardiac death in hypertrophic cardiomyopathy.JAMA. 2007; 298:405–412. doi: 10.1001/jama.298.4.405.CrossrefMedlineGoogle Scholar
    • 238. Vriesendorp PA, Schinkel AF, Van Cleemput J, Willems R, Jordaens LJ, Theuns DA, van Slegtenhorst MA, de Ravel TJ, ten Cate FJ, Michels M . Implantable cardioverter-defibrillators in hypertrophic cardiomyopathy: patient outcomes, rate of appropriate and inappropriate interventions, and complications.Am Heart J. 2013; 166:496–502. doi: 10.1016/j.ahj.2013.06.009.CrossrefMedlineGoogle Scholar
    • 239. Nishimura RA, Seggewiss H, Schaff HV . Hypertrophic obstructive cardiomyopathy: surgical myectomy and septal ablation.Circ Res. 2017; 121:771–783. doi: 10.1161/CIRCRESAHA.116.309348.LinkGoogle Scholar
    • 240. Lim DS, Lutucuta S, Bachireddy P, Youker K, Evans A, Entman M, Roberts R, Marian AJ . Angiotensin II blockade reverses myocardial fibrosis in a transgenic mouse model of human hypertrophic cardiomyopathy.Circulation. 2001; 103:789–791.LinkGoogle Scholar
    • 241. Tsybouleva N, Zhang L, Chen S, Patel R, Lutucuta S, Nemoto S, DeFreitas G, Entman M, Carabello BA, Roberts R, Marian AJ . Aldosterone, through novel signaling proteins, is a fundamental molecular bridge between the genetic defect and the cardiac phenotype of hypertrophic cardiomyopathy.Circulation. 2004; 109:1284–1291. doi: 10.1161/01.CIR.0000121426.43044.2B.LinkGoogle Scholar
    • 242. Marian AJ, Senthil V, Chen SN, Lombardi R . Antifibrotic effects of antioxidant N-acetylcysteine in a mouse model of human hypertrophic cardiomyopathy mutation.J Am Coll Cardiol. 2006; 47:827–834. doi: 10.1016/j.jacc.2005.10.041.CrossrefMedlineGoogle Scholar
    • 243. Shimada YJ, Passeri JJ, Baggish AL, O’Callaghan C, Lowry PA, Yannekis G, Abbara S, Ghoshhajra BB, Rothman RD, Ho CY, Januzzi JL, Seidman CE, Fifer MA . Effects of losartan on left ventricular hypertrophy and fibrosis in patients with nonobstructive hypertrophic cardiomyopathy.JACC Heart Fail. 2013; 1:480–487. doi: 10.1016/j.jchf.2013.09.001.CrossrefMedlineGoogle Scholar
    • 244. Nagueh SF, Lombardi R, Tan Y, Wang J, Willerson JT, Marian AJ . Atorvastatin and cardiac hypertrophy and function in hypertrophic cardiomyopathy: a pilot study.Eur J Clin Invest. 2010; 40:976–983. doi: 10.1111/j.1365-2362.2010.02349.x.CrossrefMedlineGoogle Scholar
    • 245. Mearini G, Stimpel D, Geertz B, et al . Mybpc3 gene therapy for neonatal cardiomyopathy enables long-term disease prevention in mice.Nat Commun. 2014; 5:5515. doi: 10.1038/ncomms6515.CrossrefMedlineGoogle Scholar
    • 246. Mearini G, Stimpel D, Krämer E, Geertz B, Braren I, Gedicke-Hornung C, Précigout G, Müller OJ, Katus HA, Eschenhagen T, Voit T, Garcia L, Lorain S, Carrier L . Repair of Mybpc3 mRNA by 5’-trans-splicing in a mouse model of hypertrophic cardiomyopathy.Mol Ther Nucleic Acids. 2013; 2:e102. doi: 10.1038/mtna.2013.31.CrossrefMedlineGoogle Scholar
    • 247. Gedicke-Hornung C, Behrens-Gawlik V, Reischmann S, et al . Rescue of cardiomyopathy through U7snRNA-mediated exon skipping in Mybpc3-targeted knock-in mice.EMBO Mol Med. 2013; 5:1128–1145. doi: 10.1002/emmm.201202168.CrossrefMedlineGoogle Scholar
    • 248. Jiang J, Wakimoto H, Seidman JG, Seidman CE . Allele-specific silencing of mutant Myh6 transcripts in mice suppresses hypertrophic cardiomyopathy.Science. 2013; 342:111–114. doi: 10.1126/science.1236921.CrossrefMedlineGoogle Scholar
    • 249. Ho CY, Lakdawala NK, Cirino AL, Lipshultz SE, Sparks E, Abbasi SA, Kwong RY, Antman EM, Semsarian C, González A, López B, Diez J, Orav EJ, Colan SD, Seidman CE . Diltiazem treatment for pre-clinical hypertrophic cardiomyopathy sarcomere mutation carriers: a pilot randomized trial to modify disease expression.JACC Heart Fail. 2015; 3:180–188. doi: 10.1016/j.jchf.2014.08.003.CrossrefMedlineGoogle Scholar
    • 250. Green EM, Wakimoto H, Anderson RL, et al . A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice.Science. 2016; 351:617–621. doi: 10.1126/science.aad3456.CrossrefMedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.