Next Article in Journal
Integrated Analysis of Metabolome and Transcriptome Reveals the Effect of Burdock Fructooligosaccharide on the Quality of Chinese Cabbage (Brassica rapa L. ssp. Pekinensis)
Previous Article in Journal
Sodium-Glucose Cotransporter 2 Inhibitor Therapy in Different Scenarios of Heart Failure: An Overview of the Current Literature
Previous Article in Special Issue
Analysis of the Association between Copy Number Variation and Ventricular Fibrillation in ST-Elevation Acute Myocardial Infarction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dilated Cardiomyopathy: A Genetic Journey from Past to Future

by
Noah A. Newman
1 and
Michael A. Burke
2,*
1
Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
2
Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(21), 11460; https://doi.org/10.3390/ijms252111460
Submission received: 20 September 2024 / Revised: 21 October 2024 / Accepted: 23 October 2024 / Published: 25 October 2024
(This article belongs to the Special Issue Genetic Insights into Cardiovascular Diseases)

Abstract

:
Dilated cardiomyopathy (DCM) is characterized by reduced systolic function and cardiac dilation. Cases without an identified secondary cause are classified as idiopathic dilated cardiomyopathy (IDC). Over the last 35 years, many cases of IDC have increasingly been recognized to be genetic in etiology with a core set of definitively causal genes in up to 40% of cases. While over 200 genes have been associated with DCM, the evidence supporting pathogenicity for most remains limited. Further, rapid advances in sequencing and bioinformatics have recently revealed a complex genetic spectrum ranging from monogenic to polygenic in DCM. These advances have also led to the discovery of causal and modifier genetic variants in secondary forms of DCM (e.g., alcohol-induced cardiomyopathy). Current guidelines recommend genetic counseling and screening, as well as endorsing a handful of genotype-specific therapies (e.g., device placement in LMNA cardiomyopathy). The future of genetics in DCM will likely involve polygenic risk scores, direct-to-consumer testing, and pharmacogenetics, requiring providers to have a thorough understanding of this rapidly developing field. Herein we outline three decades of genetics in DCM, summarize recent advances, and project possible future avenues for the field.

1. Introduction

The cardiomyopathies are a set of diseases with primary abnormalities in cardiac structure and function [1]. These disorders, which are typically grouped into morphologic subtypes, were associated with apparent familial transmission in many cases. This led to the hypothesis that the cardiomyopathies could have underlying genetic etiologies. Investigations over the last 35 years have confirmed a monogenic basis for these disorders. The first identified was hypertrophic cardiomyopathy (HCM), [2,3] which is now known to be a disease of the sarcomere, with >1500 variants identified in eight definitive sarcomere genes [4]. Similarly, causative variants for arrhythmogenic right ventricular cardiomyopathy (ARVC) have established ARVC as a disease of the desmosome [4], while channelopathies are predominantly caused by mutations in potassium and sodium channel genes [5]. Recently, additional genes have been identified that may contribute to these cardiomyopathies [6]. However, the evidence supporting their pathogenicity is less robust, frequently leading to diagnostic uncertainty in clinical testing [7,8,9].
Unlike these other cardiomyopathies, dilated cardiomyopathy (DCM) is unique in that it is genetically heterogeneous with a variety of culprit gene ontologies that lead to the common phenotype of a dilated and hypocontractile ventricle [1]. Further, the breathtaking pace of genetics discoveries, driven by disruptive technological advances in DNA sequencing and computing power, has led to a near constant evolution in our understanding and use of genetic information in patients with DCM. This review will discuss both the evolving genetic basis of DCM over the last three decades and the current and future impact of clinical genetic testing on the management of patients with DCM.

2. Epidemiology

DCM is a broad term that reflects the dilation and decreased systolic function of either or both ventricles [10]. Individuals with DCM typically develop heart failure (HF), and DCM is the most frequent underlying reason for heart transplantation. DCM is categorized into ischemic and non-ischemic etiologies, with nearly equal prevalence based on recent clinical trials in patients with reduced systolic function [11,12]. Many secondary causes of DCM exist including infections, drugs, toxins, nutrient deficiencies, and autoimmune diseases [13,14]. Historically, when a secondary cause could not be identified, DCM was classified as “idiopathic” (IDC). With progress in genetics, a substantial proportion of what used to be labeled IDC can now be called genetic. For the purposes of this review, we will focus on genetic causes of DCM.
Early and still widely cited epidemiologic data on DCM reported an incidence of ~6–8 per 100,000 population year [15,16,17] and a prevalence of ~1:2500 [17]. However, this is a substantial underestimate. When accounting for the prevalence of DCM among clinical trials and population cohorts it has been estimated that the true prevalence of DCM is closer to 1:250, or 0.4% [18]. This estimate has recently been confirmed using >39,000 patients imaged with cardiac MRI (CMR) in the UK Biobank. In this cohort, the population prevalence of DCM was found to be 1 in 220 (0.45%, 95% CI 0.39–0.53%), which encompasses the 1:250 estimate [19].
DCM carries a poor prognosis. In long-term studies of contemporary patient populations with non-ischemic DCM, roughly 50% experienced a composite of non-fatal life-threatening arrhythmia, unplanned cardiovascular hospitalization, or cardiovascular death over 12 years [20], and 17% experienced death, transplant, or ventricular assist device (VAD) implant over 8 years [21]. Prognosis has been noted to be worse in patients with pathogenic/likely pathogenic (P/LP) variants compared to those without an identified genetic contribution [22,23,24]. Notably, prognosis has improved when comparing patients diagnosed in the 1970s to those diagnosed in the 2010s [21], which may be in part attributed to increased utilization in guideline-directed medical therapy (GDMT), device therapies such as implantable cardioverter-defibrillators (ICD), and earlier patient identification [25].

3. Familial Clustering in Idiopathic DCM

Early studies suggested that 10–20% of patients with IDC have a family history [26]. However, a patient-reported family history is an insensitive marker for genetic disease. Among 109 probands with IDC, a reported family history was present in only 11%, while clinical screening with echocardiography identified DCM in the family members of 32% of probands, a nearly 3-fold difference [27]. Further, 44% of probands were found to have a disease-causing DCM variant, yet a family history was present in only 36% of these individuals. In a separate cohort of genotype-positive patients with DCM who were referred for VT ablation, a family history of DCM was present in only 27% and a family history of sudden death in 35% [28]. Indeed, in most large genetic studies of IDC, the prevalence of a reported family history is typically less than 50% even in those with putatively causal DCM gene mutations (i.e., in those with a clear heritable cause).
There are many reasons that explain the limited identification of familial disease in clinical practice (Table 1). An obvious reason is unrecognized disease in family members, a major problem in DCM as unsuspected but affected family members sometimes present with sudden death as the initial manifestation of disease [29,30]. This under-recognition of family members is partly due to the fact that virtually all causal DCM genetic mutations have (1) incomplete penetrance, meaning a percentage of individuals with a putatively causal DCM variant will never develop the phenotype; and (2) variable expressivity, meaning individuals with the same exact mutation will manifest different aspects of the disease phenotype and can have a vastly different disease severity. Additionally, the mutation could be a de novo variant in the proband, meaning there will be no history in siblings or older generations. This is referred to as sporadic DCM, another form of genetic DCM. Overall, while a family history increases the likelihood of finding a causal variant on genetic testing, the absence of a family history does not exclude the possibility of genetic disease. Further, a family history can be present in the absence of a pathogenic mutation because our understanding of DCM genetics remains incomplete.
The majority of genetic DCM occurs in adults, with a very variable age of onset, even within families. Consequently, the absence of the phenotype in a genotype-positive individual does not absolve them of future risk and underscores the need for lifelong screening as per guidelines [4]. Perhaps not surprisingly, when screening family members with cardiac imaging, the prevalence of familial DCM is higher due to identification of subclinical disease. The recent DCM precision medicine study estimated 30% of IDC to be familial. But, when utilizing an expanded definition that included either left ventricle (LV) chamber dilation or reduced LV ejection fraction (LVEF), which portend a 10-fold increased risk for progression to DCM [31], the prevalence of familial DCM was greater than 50% [32]. At the time of initial evaluation in seemingly asymptomatic relatives, up to 20% will be found to have overt DCM, with an age-dependent increase in prevalence [27,29,33]. Importantly, these individuals warrant proper treatment as per guidelines [34]. These facts underscore the guideline recommendations for both clinical and genetic screening in IDC [4,34].

4. Gene Ontologies in DCM

Since the late 1990s, there have been tremendous advances in genetic sequencing allowing for even more nuanced appreciation of the influence of genetic variants on cardiomyopathy. DCM can result from a multiplicity of variants implicated in many different cellular processes. This includes genes involved in force generation, signaling, protein trafficking, electrolyte homeostasis, cellular architecture, energy regulation, and gene expression (Figure 1A–E and Table 2). Currently, mutations in over 250 genes have been associated with DCM. However, strong genetic evidence for the vast majority is lacking [7,8]. To define variant pathogenicity, the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) published guidelines based on a diverse set of criteria. Scores are used to classify variants according to the following qualifiers: pathogenic (P), likely pathogenic (LP), variant of uncertain significance (VUS), likely benign (LB), or benign (B) [35].

4.1. Sarcomere Mutations in DCM

Similar to HCM, mutations in sarcomere genes are causal in DCM [37]. Definitive disease-causing variants in both disorders have been identified in cardiac actin (ACTC1), β-myosin heavy chain (MYH7), the troponins (TNNT2, TNNI3), and α-tropomyosin (TPM1), with putative DCM mutations also identified in cardiac troponin-c (TNNC1) and myosin binding protein C (MYBPC3) [1]. Sarcomere gene mutations underlie 30–40% of HCM and constitute nearly all P/LP variants on clinical genetic testing [127,128]. By contrast, sarcomere gene mutations are present in <10% of IDC cases [37,129,130,131,132].
Notably, detailed genetic and molecular analyses have shown that causal DCM and HCM variants in a given gene have opposing pathophysiologic effects. In a genome-wide association study (GWAS) of 1733 unrelated patients with HCM and 5521 patients with DCM, six loci were associated with HCM and three with DCM [133]. GWAS was also performed on LV phenotypic traits (e.g., LVEF, LV wall thickness, LV strain) in 19,260 participants who underwent CMR in the UK Biobank. This demonstrated strong associations between LV phenotypic traits, with opposing genetic effects in HCM and DCM. For instance, a positive genetic correlation was identified between HCM and LV wall thickness, while a positive genetic correlation was found between DCM and LV end diastolic volume, both disease hallmarks. Strikingly, all four measures of LV contractility assessed were positively correlated with HCM and negatively correlated with DCM. To identify additional genetic loci associated with HCM and DCM, the authors performed multi-trait analysis of GWAS (MTAG). MTAG improves the power of GWAS by identifying genetic loci that are associated with two correlated phenotypes (e.g., HCM and LVEF). This identified 10 additional loci for each HCM and DCM. Remarkably, 72.4% of these loci were significantly associated with both HCM and DCM, and of these, all but one were associated with an opposite direction of effect (i.e., if positively correlated with HCM, the risk allele was negatively associated with DCM and vice versa) [133].
This translates to in vivo models of sarcomere gene mutations. Distinct mutations in the TNNC1 gene alter the calcium binding affinity of cardiac troponin-c [134], which in turn alters myofilament tension. Variants that increase calcium binding prolonged tension, producing hypercontractility, while those with lower calcium binding reduced myofilament tension and thereby contractility. This difference leads to differential activation of the nodal extracellular signal regulated kinase (ERK1/2) pathway. High myofilament tension increased ERK1/2 pathway signaling, while reduced tension blocks this activation. The result is concentric (HCM) or eccentric (DCM) cardiac hypertrophy, respectively. Thus, distinct cardiomyopathy mutations in the same gene can alter myofilament tension in very different ways, leading to marked differences in myocardial remodeling and, consequently, phenotype [134]. Collectively, these data prove that shared genetic pathways in HCM and DCM result in opposing pathophysiologic effects and demonstrate how genetics can provide a critical window into mechanistic understanding.

4.2. Nuclear and Cytoskeletal Architecture Mutations in DCM

Mutations in several structural genes also cause DCM, though the exact mechanisms remain unclear. These structural genes encode for key proteins in cytoskeletal and nuclear architecture. The most common of these structural genes, accounting for approximately 6% of all DCM cases, is LMNA, which encodes lamin A/C, a critical component of the nuclear lamina [135]. In humans, patients with LMNA variants have high disease penetrance (>90%) and a very high burden of electrophysiologic abnormalities including atrioventricular (AV) block, atrial fibrillation, and malignant ventricular arrhythmias (VAs) [136]. One third of cases of DCM with AV block are due to LMNA mutations [137], and nearly two thirds of individuals will have some degree of AV block within 7 years of a diagnosis of LMNA cardiomyopathy [138]. Risk factors that predict malignant VAs include (1) male sex; (2) non-missense mutation; (3) documented history of non-sustained VAs; and (4) LVEF < 45%. Defibrillator placement is reasonable in the presence of two or more of these risk factors [139]. These electrophysiologic complications are both progressive and highly prevalent in LMNA cardiomyopathy [138]. Consequently, when a cardiac implantable electronic device (CIED) is required for either a pacing or arrhythmia indication, placement of a dual chamber defibrillator is reasonable [139,140]. Hence, LMNA cardiomyopathy constitutes one of the few genotype-specific evidenced-based guideline recommendations in the cardiomyopathies.
A recently identified DCM gene, filamin C (FLNC) [74], binds to actin and is crucial for anchoring the cytoskeleton to the sarcomere and intercalated discs. It is required for sarcomere thin filament assembly [141,142,143]. Mutations in FLNC cause accumulation of Z-disc proteins, activate aberrant intracellular signaling, and alter cardiomyocyte calcium handling, which ultimately leads to reduced contractility [142,144]. FLNC mutations account for approximately 3% of DCM cases [74,131,145]. FLNC variants have also been associated with ACM [74] and HCM [77]. Interestingly, the type of variant seems to influence the phenotype: truncating variants almost exclusively cause DCM while missense variants predominate in those with HCM.
Structural genes implicated in DCM are often associated with syndromic forms of disease, most commonly with muscular dystrophy, secondary to their expression in both cardiac and skeletal muscle tissue. The archetypal syndromic cardiomyopathy is Duchenne muscular dystrophy, caused by mutations in the dystrophin (DMD) gene [81]. DCM is typically present in patients with Duchenne by their early 20 s. Mutations in DMD also cause Becker muscular dystrophy and X-linked DCM, a rare cardiac-only form of the disease [79]. Variants in FLNC have long been associated with myofibrillar myopathy, with cardiomyopathy only being identified recently. LMNA cardiomyopathy is associated with Emery-Dreifuss muscular dystrophy (EDMD) type 2, limb girdle muscular dystrophy, and congenital muscular dystrophy. Similarly, the inner nuclear envelope protein emerin, encoded by the EMD gene, is the cause of EDMD type 1, which is associated with cardiomyopathy in a majority of cases [146]. Mutations in the cytoskeletal protein desmin, encoded by the DES gene, cause DCM that frequently coexists with skeletal myopathy [82]. Skeletal muscle involvement has recently been found in the giant protein titin, though the clinical impact of these findings was unclear [147]. The presence of skeletal myopathy in other putative DCM genes as well as DCM being identified in other neuromuscular disorders pervade the literature. Importantly, when DCM is accompanied by neuromuscular disease, a P/LP variant is identified in >60% of cases on clinical genetic testing [129].
Mitochondrial genes also cause congenital forms of syndromic DCM. For example, Barth syndrome is a consequence of a rare X-linked mutation in the TAZ gene that encodes for tafazzin, which is responsible for altering mitochondrial membrane lipids [148]. This disease can be fatal in childhood and is further characterized by short stature and neutropenia [91]. Additionally, DCM with ataxia syndrome is an autosomal recessive disorder of the DNAJC19 gene that encodes for the DnaJ heat shock protein family (Hsp40) member C19, an inner mitochondrial membrane protein. Predominantly occurring in a Hutterite population in Alberta, Canada, these patients are characterized by DCM and developmental delay with early mortality in childhood [149].

4.3. Protein Trafficking

The Bcl2-associated athanogene 3 (BAG3) gene encodes a multi-functional protein that is a structural component of the sarcomere and also functions to maintain normal cell function by regulating protein folding, apoptosis, autophagy, and mitochondrial function [150]. It is a very ancient protein found throughout the animal kingdom and has a close homolog in plants, suggesting it arose early in the development of multicellular life. Mutations have been identified in several DCM cohorts, with high penetrance [94,151,152]. Pathogenic mutations seem to reduce cellular BAG3 levels. Interestingly, BAG3 levels also decline in advanced HF, suggesting that reduced BAG3 expression is a common pathway in both genetic and non-genetic forms of DCM, marking BAG3 as a particularly intriguing therapeutic target [151].

4.4. Titin

Despite the many disease-causing genes identified in the first two decades of the genetics era, the yield of clinical genetic testing for DCM remained frustratingly low (~10%). With the advent of massively parallel—so called “next generation”—sequencing (NGS) technology in the first decade of the new millennium, this would soon change with the identification of titin (TTN) as the commonest DCM-causing gene. Titin is, by far, the largest protein encoded by the human genome. A single titin protein spans half the length of the sarcomere to connect the Z-disc to the M-line [153]. It is essential for proper sarcomere assembly [154,155], and provides most of the passive tensile strength of the sarcomere [156,157]. Its incredibly large size made it cost-prohibitive to fully sequence in a large cohort of patients until the advent of NGS technology. Using NGS in a large, multi-center cohort of patients with DCM, researchers identified TTN mutations in over 21% of patients, doubling the yield of clinical genetic testing across other established DCM genes [56].
In this landmark study, TTN cosegregated in DCM families with a combined lod score of 11.1, unequivocally confirming the pathogenicity of TTN in DCM. However, putatively causal TTN variants were also identified in 3% of well-phenotyped controls without cardiac structural abnormalities, a prevalence roughly 20-fold greater than the estimated population prevalence of DCM. This created a vexing problem in establishing pathogenicity of novel variants on clinical genetic testing. An elegant molecular genetic analysis demonstrated that the majority of these pathogenic variants were located in exons that were not expressed in the final protein (e.g., the exon was spliced out of the transcript, yielding a normal TTN protein) [158]. In the remaining ~1% of ostensibly normal individuals harboring potentially pathogenic TTN variants in constitutively expressed exons, CMR demonstrated mild eccentric remodeling and mildly reduced contractility (i.e., subclinical disease) in most, revealing high penetrance but very variable expressivity for TTN in DCM [159]. Consequently, clinical genetic testing assesses not only the variant but whether that variant is in an exon that is part of the final mRNA transcript.
Importantly, the variants identified to be causal in TTN are truncating variants (TTNtv) that drastically alter titin structure by the removal of a large portion of the protein that variably contributes to distensibility, stretch-sensed signaling, and attachment to the M-line. When analyzed across a range of cohorts, TTNtv were identified in 11–15% of adult cases of sporadic DCM and 23–27% of familial DCM [56,158,159,160,161,162,163]. Assuming that up to 40% of DCM has a monogenic basis, then the approximate prevalence of currently actionable TTNtv in adult DCM is around 17–18%.
Non-truncating variants in TTN can also cause disease. A recent analysis identified non-canonical splice site variants as pathologic in an additional 1–2% of DCM cases [164], bringing the yield for TTN close to 20% in an unselected DCM cohort. Missense variants (those that cause a single amino acid change, leaving the protein intact) in TTN can also cause DCM as evidenced by several detailed familial studies [55,165,166]. Human induced pluripotent stem cells carrying putatively damaging TTN missense variants exhibit contractile dysfunction similar to TTNtv-expressing cells, further supporting causality of TTN missense variants [167]. Using in silico testing to predict pathogenicity suggests that TTN missense variants could be causal in up to an additional 5 to >20% of DCM cases [162,168,169]. However, due to the immense size of the TTN coding region, missense variants are extremely common, with an average of 23 per person [56,170]. When limiting to just rare variants with a minor allele frequency of at least <0.01, all individuals (i.e., not just those with DCM) carry ~1–2 rare TTN missense variants [56,168,170]. Importantly, in all but one study, TTN missense mutations are not enriched among DCM patients as compared to the general population [56,162,169,170]. Therefore, in the absence of detailed linkage studies in families of sufficient size, the majority of TTN missense variants currently remain uninterpretable (reported as VUS) on clinical genetic testing; this is a highly active area of research.

4.5. Gene Expression

The critical role played by TTN is supported by mutations in ribonucleic acid binding protein 20 (RBM20) gene [96], which encodes a nuclear protein that splices mRNA transcripts prior to their translation into protein. Interestingly, the molecular mechanism in the majority of RBM20 variants appears to be the inability of an otherwise functional RBM20 protein to be transported into the nucleus [171]. Among many other genes, RBM20 is necessary for the proper splicing of TTN mRNA [172]. In disease causing variants, a shift occurs in titin production from the N2B isoform to the larger and more compliant N2BA isoform, creating longer sarcomeres with altered Frank Sterling mechanics [173,174]. RBM20 also controls splicing of additional genes important in cardiomyocyte contraction, including those involved in Ca2+ regulation (CAMK2D and CACNA1C) [175], which may account for the notably high burden of VAs in these patients [176].
The T-box protein 20 (TBX20) gene, encoding a key developmental cardiac transcription factor, has long been associated with congenital heart defects [98]. While isolated cases in adult DCM have been reported, a recent analysis of nearly 7500 unrelated DCM probands identified substantial enrichment for TBX20 truncating variants, making up 0.3% of all DCM cases [99]. Subsequent cosegregation analyses confirmed linkage with a lod score of 4.5. These individuals have a high prevalence of LV non-compaction (LVNC), and 34% also had concomitant congenital heart defects. This is the largest study of its kind and suggests other congenital defects may also constitute a small proportion of DCM cases.

5. Overlapping Cardiomyopathic Syndromes

5.1. Arrhythmogenic Cardiomyopathy

Arrhythmogenic right ventricular cardiomyopathy (ARVC) has been identified for decades [177] and is characterized by fatty infiltration and fibrosis of the right ventricle in association with HF and a particularly high burden of malignant VAs [178]. Early genetic studies defined ARVC as a disease of the cardiac desmosome with the identification of mutations in desmosomal genes plakoglobin (JUP), desmoplakin (DSP), plakophilin (PKP2), desmoglein 2 (DSG2), and desmocollin 2 (DSC2), and the non-desmosomal transmembrane protein 43 (TMEM43) gene [1]. Mutations in one of these genes may be found in ~60% of cases of ARVC [179,180].
With larger studies and improved imaging, particularly via CMR, it has become apparent that ARVC is not limited to the right ventricle. LV involvement is identified in 66–84% of cases of classic ARVC [181,182,183]. Up to 25% will have outright LV systolic dysfunction, with the causal gene having a large influence on this phenotypic trait (e.g., 0% LV systolic dysfunction with JUP and TMEM43 mutations, but >50% with DSP) [182]. Conversely, classic ARVC has also been identified in several definitive DCM genes, namely phospholamban (~5% of those with a putative pathogenic mutation) [182], LMNA (~4%) [68], and FLNC (1–3%) [184,185].
Improvements in imaging and the phenotyping of larger cohorts with mutations in specific genes has recently led to the discovery of ARVC-like features but with predominantly LV involvement, so-called ALVC. Because of this phenotypic heterogeneity, the consensus term now utilized in guidelines is “arrhythmogenic cardiomyopathy” (ACM), which encompasses ARVC and ALVC, and includes numerous known DCM genes that feature a high burden of arrhythmias in addition to the classic ARVC genes [34,186] (Figure 2). These include LMNA, DES, FLNC, RBM20, BAG3, and TTN. Additionally, multiple mutations in phospholamban (PLN), the molecular regulator of sarcoplasmic reticulum calcium cycling, cause DCM with a high burden of arrhythmias [121,122,123]. The altered flux of calcium between the sarcoplasmic reticulum and cytosol alters myofilament tension, leading to both reduced contractility [134] and electrical instability [122]. Mutations in the voltage-gated sodium channel α-subunit (SCN5A) can also cause DCM with VAs. SCN5A is a particularly promiscuous gene, with mutations that cause ACM, long-QT syndrome (type 3), Brugada syndrome, conduction delay, ectopic Purkinje foci, atrial fibrillation, and sinus node dysfunction via the varying effects of gain- or loss-of-function mutations that alter sodium current and membrane potential [187].
The desmosomal gene DSP is associated with a particularly unique form of ACM. DCM-causing mutations in DSP were originally described as part of Carvajal syndrome, an extremely rare autosomal recessive disease with DCM, palmoplantar keratoderma, and wooly hair [104]. With the contemporaneous discovery of the desmosome as the genetic basis of ARVC, rare autosomal dominant ARVC-causing mutations in DSP were soon identified [105]. However, subsequent work revealed that dominant mutations in DSP could also cause ALVC [188]. In a large natural history study of 107 patients with DSP cardiomyopathy, primary LV dysfunction was nearly four times as prevalent as RV-predominant disease and was associated with particularly poor outcomes [189]. Penetrance was also found to be much higher (nearly 60% by age 40) than what is typically seen with other desmosomal gene mutations. Interestingly, the disease is punctuated by episodic myocardial injury with troponin elevation mimicking acute myocarditis, a unique feature heretofore not definitively observed with other cardiomyopathy genes. This finding suggests that at least a subset of DSP cardiomyopathy might be responsive to immunosuppression, marking an active area of research that as yet is supported only by case reports [190].

5.2. Left Ventricular Non-Compaction Cardiomyopathy

Formation of LV trabeculations is a normal part of early embryologic development, with myocardial compaction occurring at later fetal stages [191]. LVNC defines a state where ventricular trabeculation exceeds normal parameters. This need not be found with aberrant cardiac function. Among healthy individuals, LVNC is common [192] and the prevalence is substantially higher in those who engage in vigorous physical activity [193]. Indeed, non-compaction is consistently identified in 1.5–8% of athletes [194,195] and in ~8% of healthy pregnant women [196]. Notably, excess LV trabeculation is not a static phenotypic trait, with both appearance and resolution occurring in various physiologic states [196,197]. The reason for this transience remains elusive.
Confounding our understanding of LVNC is a progressive change in diagnostic criteria [198]. Also, the prevalence of LVNC varies drastically by the imaging modality used: criteria for LVNC are met in approximately 1.3% of cases by echo, whereas this increases to 14.8% (a more than 11-fold difference) with CMR [199]. Among individuals with cardiomyopathy, LVNC can be a coexistent phenotype, most commonly with DCM [200], and also with HCM [201,202]. However, coexistent hypertrabeculation does not impact outcomes [198].
The best evidence supporting a genetic basis for LVNC comes from rare mutations in the mindbomb homolog 1 (MIB1) gene, a key component to the NOTCH signaling pathway, which plays a critical role in normal cardiac embryologic development [203]. Two distinct loss-of-function mutations were identified in two families with LVNC. However, subsequent modeling of these heterozygous mutations in mice failed to reproduce the phenotype [204]. This prompted whole exome sequencing that revealed co-inheritance of multiple modifier mutations with the MIB1 variants in all affected individuals. These modifiers were required for development of LVNC in vivo, painting a complex genetic picture.
Other putative LVNC variants often display mixed phenotypes both within and across pedigrees. This variant-specific phenotypic heterogeneity most commonly coincides with DCM [165,205,206], but also occasionally with HCM, particularly apical HCM [207,208]. In large cohorts, the spectrum of putative pathogenic mutations largely resembles that of DCM, with variants in TTN being most common and multiple distinct gene ontologies identified, though HCM genes are also found [209,210,211,212]. Notably, in a cohort of 840 LVNC patients, a very small subset of mutations, particularly truncating variants in MYH7, were highly enriched in LVNC patients relative to controls or those with HCM and DCM, suggesting that a small subset of mutations within known cardiomyopathy genes either cause or act as modifiers to produce the LVNC phenotype [212].
These data demonstrate that the presence of LVNC is not necessarily a pathophysiologic state, and even when it is, there is substantial overlap with DCM, and to a lesser extent HCM, in the vast majority of cases. Importantly, LVEF is the primary driver of adverse events in LVNC and is a key metric to assess when considering therapy [213]. Because of this phenotypic pleiotropy, recent guidelines have dropped LVNC as a distinct cardiomyopathy, rather indicating that LVNC is a phenotypic variant of DCM and/or HCM, with the recommendation to manage patients according to guidelines specific for those disorders [214,215]. Whether a small subset of LVNC has a distinct underlying genetic architecture remains to be determined.

5.3. Restrictive Cardiomyopathy

Linkage analysis defined a mutation in TNNI3 as the cause of restrictive cardiomyopathy (RCM) in a large family [216]. However, multiple family members met clinical criteria for HCM, and histologic analysis showed myocyte hypertrophy, fibrosis, and myofibrillar disarray consistent with HCM, a finding that has been observed in other patients with RCM [216,217]. Similarly, a mutation in the myopalladin (MYPN) gene was identified in RCM [64]. However, histologic analysis showed evidence for myofibrillar disarray, and other MYPN mutations have been associated with HCM [218] and DCM [65]. Additional mutations in ACTC1 [217], TNNT2 [217,219], and MYL2 [220] have been associated with RCM, but phenotypic heterogeneity, including HCM and DCM, existed in affected first-degree relatives.
Restrictive physiology is an extreme phenotypic variant in HCM, being identified in 1.5–6% of cases, and portends a particularly poor prognosis [221,222,223]. While rare regardless of the subgroup, restrictive physiology appears to be more common with thin filament gene mutations in HCM [223]. Much like LVNC with DCM, these data suggest that RCM is indicative of a genetic predilection to restrictive physiology in the context of a different cardiomyopathy (HCM) rather than a distinct genetic entity.

6. Genetics in Secondary Forms of DCM

Approximately 25–30% of non-ischemic DCM is associated with an identifiable secondary cause [18]. Recent evidence has demonstrated that several of these secondary forms of DCM have at least a partial genetic basis. The commonest mutations identified in all populations are TTNtvs. Importantly, this suggests that these insults are non-genetic modifiers that may alter the penetrance and/or expressivity of TTNtvs and indicate that TTNtvs are a likely genetic predisposition to other secondary forms of non-ischemic DCM.

6.1. Peripartum Cardiomyopathy

Peripartum cardiomyopathy (PPCM) is defined as new onset DCM occurring between ~1 month prepartum and ~5 months postpartum, and has long been noted to have familial clustering in some cases [26,224]. Recent evidence from large multicenter cohorts has identified truncating variants in definitive DCM genes among 15% of unselected PPCM cases [225,226]. TTNtvs account for the majority, and constitute ~10% of all PPCM cases, a rate similar to that observed in sporadic DCM. Additional genes significantly enriched in PPCM include FLNC, DSP, and BAG3 [226]. This subset of genes mirrors DCM and suggests that other genetic factors are likely to play a role (e.g., other DCM genes, missense variants, genetic modifiers). However, since PPCM is a rare disease, the power to detect even rarer variants will be limited.

6.2. Cardiotoxins

Similar findings have been noted in patients with chemotherapy-induced cardiomyopathy [227] and alcohol-induced cardiomyopathy [228]. In a diverse cohort of adult patients with DCM attributed to cancer therapies (> 90% anthracyclines), 13.4% had potentially pathogenic variants in known DCM genes [227]. This was nearly triple the rate found in unselected cancer patients from the Cancer Genome Atlas. Again, TTNtvs were the most common, accounting for 60% of all variants identified in adults, and being found in 8.1% of all adult patients with chemotherapy-induced DCM. Notably, in pediatric cancer, TTNtvs were less common, accounting for 5% of all cases. This is concordant with pediatric DCM data, where TTNtvs consistently make up a significantly lower percent of P/LP variants, particularly in younger children (6% in children < 13 years old) [229].
Alcohol-induced cardiomyopathy is classically defined when DCM is present in the absence of another cause in an individual that is consuming ≥80 g/day (approximately six each of: 12 oz beers that are 5% alcohol; 5 oz glasses of wine that are 12% alcohol; 1.5 oz shots of liquor that is 80 proof) for more than 5 years, a hard metric to quantify definitively. However, using this strict definition, 141 patients were identified who met criteria [228]. In these individuals, 13.5% were found to carry putatively causal variants in known DCM genes, again with TTNtv constituting the majority and being found in 10% of all individuals with alcohol-induced cardiomyopathy. Notably, over 40% of these individuals were found to have a family history of DCM.
After establishing the contribution of genetics to alcohol-induced cardiomyopathy, these authors sought to assess the role of moderate alcohol intake on DCM. Using a large and unselected DCM referral cohort, 15.5% were found to have “excess alcohol consumption” (defined by the authors as >24 g/day, similar to the upper limit of societal and government health guidelines across multiple countries) [228]. In a multi-variable analysis, neither the presence of a TTNtv nor excess alcohol consumption alone were associated with LVEF. However, in individuals with both a TTNtv and excess alcohol consumption, the LVEF was significantly and substantially (~10%; p < 0.01) lower than the cohort mean. This clever analysis identifies even moderate alcohol intake as a phenotypic modifier in DCM.

6.3. Acute Myocarditis

Acute myocarditis has confounded physicians for decades as the etiology typically remains elusive, even in the setting of a biopsy-confirmed histopathologic subset. While viruses have long been implicated, proving causality is extremely hard [230]. Recent evidence has shown there is a strong genetic predilection in at least a subset of cases. As noted above, there is a clear inflammatory subset among patients with DSP cardiomyopathy, a form of ACM [189]. In several cohorts of patients with biopsy-proven myocarditis, putatively causal DCM and ACM mutations are found in 8–33%, with TTNtv typically the commonest gene identified [231,232,233,234,235]. Notably, DSP mutations were identified in all cohorts.
Acute myocarditis presents with a very broad phenotypic spectrum from mild cases with normal LVEF to fulminant cases with severely reduced LVEF and cardiogenic shock. DCM-associated genes are more likely to be identified in myocarditis with reduced LVEF and shock [231,234,235]. By contrast in less severe cases and those with normal LVEF, fewer potentially pathogenic variants are identified, and the majority are in genes that cause ACM, in particular DSP. In nearly six years of follow-up, those with acute myocarditis and a pathogenic mutation were 3.1-fold less likely to have recovery of LVEF. Thus, identifying these individuals via genetic testing provides important prognostic information and could identify a cohort requiring closer long-term monitoring for maintenance of standard HF GDMT, though data supporting that approach remain limited.

7. The Natural History of DCM

With the dawn of the genetics era came great hope that genotyping would allow for gene-specific therapies for HF (i.e., pharmacogenomics). However, with the growth in our knowledge of cardiomyopathy genetics came the understanding that incomplete penetrance and variable disease expressivity were the norm in nearly all forms of genetic DCM. Within families, those carrying the same mutation can have starkly different clinical trajectories. Remarkably, this variability is even true in monozygotic twins [236]. This, coupled with the high cost of sequencing large cohorts of patients limited the ability to define gene-specific phenotypes that could in turn result in therapy tailored to the underlying cause.
The advent of NGS has permitted sequencing of large cohorts of patients with DCM at exponentially lower cost than was possible in the first two decades of the genomics era. Recently, this has led to several natural history studies of genetic DCM. Typically patients with genetic DCM are younger [23,237], have higher overall event rates [238], and, importantly, have an increased risk of progression to end-stage HF requiring advanced HF therapies including transplant or mechanical circulatory support (MCS) [23,237]. In the presence of a genetic etiology, patients also had higher likelihood of malignant VAs and are less likely to have reverse remodeling with standard HF treatments [23,24,237,239].
As more patients with genetic DCM have been identified, the power to detect differences between carriers of variants in specific genes has also grown. Several large studies in carriers of TTNtv have identified features unique to titin cardiomyopathy. While some studies show lower survival than IDC [158], the balance of evidence holds that TTNtv carriers have outcomes (death, transplant, MCS) that are similar to non-genetic DCM [23,56,160,240,241]. TTNtv carriers also have a higher incidence of VAs than non-genetic DCM [56,158] and may have poorer right ventricular function [158]. However, TTNtv seems particularly amenable to reverse remodeling, supporting the importance of GDMT in these individuals [23,242].
LMNA mutations produce the most malignant genetic DCM [243]. A hallmark feature of LMNA cardiomyopathy is a high burden of conduction system disease, atrial arrhythmias, and malignant VAs [138,243]. These are inexorably progressive, eventually develop in the majority of individuals, and guide management when considering CIED placement. LMNA cardiomyopathy also shows low rates of reverse remodeling [23,242] and very high rates of progression to end-stage HF [23,138,243]. Finally, LMNA mutations have high penetrance [243,244], which influences genetic counseling and makes cascade genetic testing particularly important.
Several other DCM genes have also been found to have disease-specific features. These influence prognosis and genetic counseling for patients and family members. BAG3 mutations are highly penetrant (>80% by age 40), have high rates of malignant VAs, and a composite of death/transplant/MCS [152]. FLNC patients have a lower survival than those with TTNtv and have a high burden of malignant VAs [245,246]. Notably, this burden is identified even in those with only mildly reduced LVEF (36–49%) and the total burden is comparable to those with LMNA cardiomyopathy. This raises the consideration for defibrillator implantation with higher LVEF, though there is insufficient data for a specific recommendation. Sarcomere variants may have the highest penetrance among all DCM-causing genes [244], with penetrance of MYH7 reaching nearly 90% by age 60 [247]. Over 1/3 of patients with MYH7 variants will have LVNC, but notably, rates of malignant VAs are lower than most other forms of DCM and overall outcomes are comparable to TTNtvs [247]. Notably, the rate of adverse events is substantially higher in DCM-causing MYH7 variants than in those causing HCM [248]. Understanding the natural history of distinct genetic causes is crucial for the future development of targeted therapies and is analogous to understanding the differences between different forms of cancer.

8. DCM: Beyond the Monogenic Hypothesis

The last 30 years have clearly defined a causal role for rare monogenic forms of DCM. Yet, putative pathogenic variants are only identified in ~40% of cases on genetic testing in unselected DCM cohorts. As noted, penetrance and expressivity are highly variable in DCM, and are influenced by genetic modifier variants [249], the epigenome, which is highly dynamic in DCM [250], and environmental factors, including other disease states, which are often associated with a worse prognosis [239]. With the rise of NGS and the sequencing of large cohorts, it is now apparent that rare putative disease-causing variants have a population prevalence that is much greater than that of overt or even subclinical DCM [19,251,252,253]. For instance, the penetrance of TTNtvs in the population may be as low as 10% [19]. It is important to note, however, that the penetrance of a putatively pathogenic variant in a family with the disease phenotype is much higher than those in an unselected population, a clear example of selection bias. This must be considered when providing genetic counseling.
These data may cast doubt on whether such mutations are truly causal. However, the sequencing and aggregation of very large population cohorts (e.g., gnomAD: https://gnomad.broadinstitute.org/ accessed on 19 April 2024) also permits burden testing—an aggregate statistical method used to identify phenotypically significant rare variants based on the difference between the prevalence of putative disease-causing variants in disease cohorts vs. controls. Burden testing has demonstrated a strong enrichment of most major DCM genes over the general population [7,132]. This cements pathogenicity when coupled with the robust genetic and functional data already published in these genes. In addition, by virtue of sequencing thousands of DCM patients and hundreds of thousands of individuals in the general population, burden testing essentially rules out the possibility of another as-yet-discovered gene making a large contribution to the monogenic basis of DCM. Extremely rare causal variants in new genes will continue to be found, but they will contribute only a very small percent to the etiology of IDC.
So, what of the 60% or more who have DCM without a definitive monogenic predisposition? Three GWASs and one exome-wide association study specifically focusing on idiopathic or sporadic DCM have been performed, identifying a collective 11 risk alleles [95,254,255,256]. Approximately half of the loci contain known DCM-causing genes, thereby establishing a link between common genetic variants and DCM, and identifying other genetic loci that are also involved. Using a discovery cohort of nearly 7000 individuals and eight identified common risk alleles for DCM, a polygenic risk score (PRS) was generated. A PRS is a cumulative score of single-nucleotide polymorphisms associated with a disease that can be used to determine disease associations and aid in prognosis. This PRS found a ~3-fold increased risk of DCM in individuals with eight vs. five risk alleles (the median of the population); similarly, there was a ~3-fold decrease in the risk of DCM in those with only one or two risk alleles [256].
Structural changes identified by imaging are a defining feature of DCM, are often perceptible before overt DCM is diagnosed, and are another powerful way to identify potential genetic loci [257]. A GWAS in over 36,000 patients without HF, DCM, or coronary disease who underwent CMR in the UK Biobank, identified 57 loci associated with some parameter of LV structure or function (45 of which had not previously been associated with DCM or cardiac imaging) [258]. A PRS for LV systolic function (highly correlated to DCM in the discovery cohort) was performed in the remaining participants in the UK Biobank (nearly 360,000 individuals) and found a strong association with incident DCM over nine years of follow-up. The polygenic background also influences the phenotype in TTNtv carriers. Per one SD increase in this PRS, LV end systolic volume increased by 7.2 mL and LVEF decreased by 2.6% [258].
Collectively, these data demonstrate that genetic DCM is not simply a monogenic/Mendelian disorder, but rather a complex genetic disease that has both a monogenic and polygenic basis. The polygenic component of DCM also helps to explain the marked variance in penetrance and expressivity that is a hallmark of this disease (Figure 3). With continued refinement in larger and more diverse cohorts, PRS testing may become an important compliment to cascade genetic testing that can influence the potential penetrance and prognosis in family members with genetic DCM.

9. Using Genetics in the Management of DCM

The use of genetic testing is increasing rapidly in clinical practice. In addition, there has been a steady rise in direct-to-consumer genetic testing, with patients occasionally coming to clinic with genetic results in-hand. Consequently, understanding the utility and limitations of genetics and genetic testing is important for modern clinical practice.

9.1. Guideline-Based Recommendations

Numerous documents across multiple medical societies provide guidance on DCM (Figure 4). All guidelines recommend (1) collecting a three-generation family history [4,34,259]; (2) clinical screening for potential at-risk first-degree relatives; and (3) management at a center with expertise in genetic cardiomyopathies. The family history is often an afterthought in a busy clinical practice. A study comparing family histories taken by inpatient cardiology teams to those collected by genetic health care professionals demonstrated a 4-fold increase in the detection rate of a familial pattern by the latter group [260]. Further, reviewing ≥ three generations is useful in identifying the mode of inheritance and detecting variants with low penetrance [261]. These data support both the need for detailed family histories as well as the important role of the genetics counselor in cardiology practice.
There is also consensus across guidelines to offer cascade genetic screening and genetic counseling to first-degree relatives of patients with genetic DCM [4,34,259]. However, guidelines differ on which probands with IDC to refer. Guidelines and consensus documents endorsed by the ESC, HFSA, ACMG, HRS, and EHRA support genetic testing/counseling in all patients with IDC. By contrast, the recent AHA/ACC/HFSA HF guidelines only support testing of “select” patients with DCM without providing additional details. While controlled prospective studies have not shown a direct benefit of genetic testing, it informs prognosis, risk stratification and, in the case of LMNA cardiomyopathy, treatment. In a study of 4782 patients with suspected genetic cardiomyopathy or arrhythmia syndromes, genetic testing identified a putative pathogenic variant in 20% [262]. Of those gene-positive individuals, two thirds were variants that would alter prognosis and/or management. Importantly, had genetic testing only been offered to patients with a high suspicion of genetic disease, 14.4% of positive test results would have been missed [262]. Further, in first-degree relatives of probands with genetic DCM, cascade genetic testing was a more cost-effective approach to periodic clinical surveillance [263].

9.2. The Burden of VUSs

Commercial genetic testing panels include many genes with weak genetic evidence, and/or that have only been identified in a very small number of individuals. These weakly associated genes virtually always return VUSs because nearly all variants will be novel/private to that individual, and there simply is not enough data without detailed familial cosegregation studies to define pathogenicity [127,128,262,264]. For example, in 240 HCM patients who did not have a variant in one of the eight sarcomere genes, only one of 186 rare variants (0.5%) across 51 additional genes was reported as likely pathogenic, with 94.5% being VUSs [128]. Dealing with VUSs represents perhaps the single most challenging aspect of clinical genetics.
That said, using burden testing in DCM has shown that the prevalence of VUSs in many DCM genes far exceeds the population prevalence of rare variations in those genes, indicating that at least some of these VUSs are indeed pathogenic [7]. In silico tools are used to predict whether a particular variant will alter protein function. However, none are particularly sensitive. These in silico tools use machine learning and are trained using genome-wide data; consequently, they are not disease-specific, a major flaw that ignores disease-specific mechanisms or other evidence that may be known and aid in definitive variant prediction in a well characterized subset of disease-specific genes. With this in mind, a disease-specific prediction tool for inherited cardiomyopathies and arrhythmias has been developed (Cardioboost: https://www.cardiodb.org/cardioboost/ accessed on 1 June 2024) [265]. Cardioboost far outperforms other in silico tools. It reclassifies many variants that would otherwise be labeled as “indeterminate” and reduces the proportion of such variants by more than half as compared to genome-wide tools. We use Cardioboost to evaluate VUSs on clinical genetic testing in definitive DCM or arrhythmia genes. It is not meant as a stand-alone tool to change a VUS to a different classification but can aid in boosting a variant as part of the ACMG/AMP criteria (specifically as supporting evidence in criteria PP3 and BP4).

9.3. The Future of Genetic Testing

Genetics is one of the most rapidly progressing fields in all of cardiovascular medicine (Figure 5). The first 30 years can be considered the monogenic era: numerous genes were unequivocally defined to be causal in genetic DCM. This was greatly aided by the development of NGS which has both increased throughput and decreased costs by several orders of magnitude. While the future will likely bring more disease genes, burden testing strongly suggests that the majority of monogenic causes of genetic DCM have been identified, with the possible exception of the causal burden of TTN missense variants.
The field has now entered a new era of polygenic discovery. The future is sure to hold more sequencing of large cohorts with concomitant identification of more genetic loci. Hopefully, there will also be an increase in the diversity of population and disease cohorts, which remain dominated by those of European ancestry [280]. This will allow refinement of PRS testing, which will eventually aid in prognosis and genetic counseling. With continued decreases in sequencing costs, direct-to-consumer testing will also likely increase. This will pose challenges not only to patients struggling to interpret this information, but also to practitioners for how to counsel them. Investment is therefore needed in genetic counseling to address the growing proportion of individuals who will have access to genetic data.
Finally, we can anticipate the next era, which we hope will be dominated by therapeutic advances in DCM treatment guided by or generated as a result of genetic and molecular genetic discovery. Gene-specific medications are starting to become available [281], though none has yet proven effective. Gene editing therapies are also being investigated. For example, CRISPR-Cas9 technologies have been used to modify genetic DCM causing variants in mice and human stem cells with promising results [282,283]. In-human studies are far rarer—the first such clinical trial (NCT05836259) for genetic cardiomyopathy is currently ongoing that targets the MYBPC3 gene implicated in HCM, but is still in the 1b phase [284]. Additional research is developing new viral vectors, also referred to as myotropic adeno-associated viral vectors (MyoAAVs), that are more specific to and effective at delivering therapeutic genes to cardiomyocytes [285].
In conclusion, the role of genetics continues to expand in both IDC and non-ischemic secondary causes of DCM and now encompasses fundamental aspects of the diagnosis, management, and prognostication of patients with DCM.

Author Contributions

M.A.B. and N.A.N. contributed to the writing of the manuscript. N.A.N. illustrated the figures within. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Burke, M.A.; Cook, S.A.; Seidman, J.G.; Seidman, C.E. Clinical and Mechanistic Insights Into the Genetics of Cardiomyopathy. J. Am. Coll. Cardiol. 2016, 68, 2871–2886. [Google Scholar] [CrossRef] [PubMed]
  2. Jarcho, J.A.; McKenna, W.; Pare, J.A.P.; Solomon, S.D.; Holcombe, R.F.; Dickie, S.; Levi, T.; Donis-Keller, H.; Seidman, J.G.; Seidman, C.E. Mapping a Gene for Familial Hypertrophic Cardiomyopathy to Chromosome 14q1. NEJM 1989, 321, 1372–1378. [Google Scholar] [CrossRef] [PubMed]
  3. Geisterfer-Lowrance, A.A.; Kass, S.; Tanigawa, G.; Vosberg, H.P.; McKenna, W.; Seidman, C.E.; Seidman, J.G. A Molecular Basis for Familial Hypertrophic Cardiomyopathy: A Beta Cardiac Myosin Heavy Chain Gene Missense Mutation. Cell 1990, 62, 999–1006. [Google Scholar] [CrossRef] [PubMed]
  4. Hershberger, R.E.; Givertz, M.M.; Ho, C.Y.; Judge, D.P.; Kantor, P.F.; McBride, K.L.; Morales, A.; Taylor, M.R.G.; Vatta, M.; Ware, S.M. Genetic Evaluation of Cardiomyopathy-A Heart Failure Society of America Practice Guideline. J. Card. Fail. 2018, 24, 281–302. [Google Scholar] [CrossRef] [PubMed]
  5. Schwartz, P.J.; Ackerman, M.J.; George, A.L., Jr.; Wilde, A.A.M. Impact of Genetics on the Clinical Management of Channelopathies. J. Am. Coll. Cardiol. 2013, 62, 169–180. [Google Scholar] [CrossRef]
  6. Micolonghi, C.; Perrone, F.; Fabiani, M.; Caroselli, S.; Savio, C.; Pizzuti, A.; Germani, A.; Visco, V.; Petrucci, S.; Rubattu, S.; et al. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int. J. Mol. Sci. 2024, 25, 9787. [Google Scholar] [CrossRef]
  7. Walsh, R.; Thomson, K.L.; Ware, J.S.; Funke, B.H.; Woodley, J.; McGuire, K.J.; Mazzarotto, F.; Blair, E.; Seller, A.; Taylor, J.C.; et al. Reassessment of Mendelian Gene Pathogenicity Using 7,855 Cardiomyopathy Cases and 60,706 Reference Samples. Genet. Med. 2017, 19, 192–203. [Google Scholar] [CrossRef]
  8. Jordan, E.; Peterson, L.; Ai, T.; Asatryan, B.; Bronicki, L.; Brown, E.; Celeghin, R.; Edwards, M.; Fan, J.; Ingles, J.; et al. Evidence-Based Assessment of Genes in Dilated Cardiomyopathy. Circulation 2021, 144, 7–19. [Google Scholar] [CrossRef]
  9. Ingles, J.; Goldstein, J.; Thaxton, C.; Caleshu, C.; Corty, E.W.; Crowley, S.B.; Dougherty, K.; Harrison, S.M.; McGlaughon, J.; Milko, L.V.; et al. Evaluating the Clinical Validity of Hypertrophic Cardiomyopathy Genes. Circ. Genom. Precis. Med. 2019, 12, e002460. [Google Scholar] [CrossRef]
  10. Richardson, P.; McKenna, W.; Bristow, M.; Maisch, B.; Mautner, B.; O’Connell, J.; Olsen, E.; Thiene, G.; Goodwin, J.; Gyarfas, I.; et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of Cardiomyopathies. Circulation 1996, 93, 841–842. [Google Scholar] [CrossRef]
  11. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [PubMed]
  12. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef] [PubMed]
  13. Dec, G.W.; Fuster, V. Idiopathic Dilated Cardiomyopathy. N. Engl. J. Med. 1994, 331, 1564–1575. [Google Scholar] [CrossRef] [PubMed]
  14. Felker, G.M.; Thompson, R.E.; Hare, J.M.; Hruban, R.H.; Clemetson, D.E.; Howard, D.L.; Baughman, K.L.; Kasper, E.K. Underlying Causes and Long-Term Survival in Patients with Initially Unexplained Cardiomyopathy. N. Engl. J. Med. 2000, 342, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  15. Bagger, J.P.; Baandrup, U.; Rasmussen, K.; Møller, M.; Vesterlund, T. Cardiomyopathy in Western Denmark. Br. Heart J. 1984, 52, 327–331. [Google Scholar] [CrossRef]
  16. Williams, D.G.; Olsen, E.G. Prevalence of Overt Dilated Cardiomyopathy in Two Regions of England. Br. Heart J. 1985, 54, 153–155. [Google Scholar] [CrossRef]
  17. Codd, M.B.; Sugrue, D.D.; Gersh, B.J.; Melton, L.J. Epidemiology of Idiopathic Dilated and Hypertrophic Cardiomyopathy. A Population-based Study in Olmsted County, Minnesota, 1975–1984. Circulation 1989, 80, 564–572. [Google Scholar] [CrossRef]
  18. Hershberger, R.E.; Hedges, D.J.; Morales, A. Dilated Cardiomyopathy: The Complexity of a Diverse Genetic Architecture. Nat. Rev. Cardiol. 2013, 10, 531–547. [Google Scholar] [CrossRef]
  19. McGurk, K.A.; Zhang, X.; Theotokis, P.; Thomson, K.; Harper, A.; Buchan, R.J.; Mazaika, E.; Ormondroyd, E.; Wright, W.T.; Macaya, D.; et al. The Penetrance of Rare Variants in Cardiomyopathy-Associated Genes: A Cross-sectional Approach to Estimating Penetrance for Secondary Findings. Am. J. Hum. Genet. 2023, 110, 1482–1495. [Google Scholar] [CrossRef]
  20. Hammersley, D.J.; Jones, R.E.; Owen, R.; Mach, L.; Lota, A.S.; Khalique, Z.; De Marvao, A.; Androulakis, E.; Hatipoglu, S.; Gulati, A.; et al. Phenotype, Outcomes and Natural History of Early-Stage Non-Ischaemic Cardiomyopathy. Eur. J. Heart Fail. 2023, 25, 2050–2059. [Google Scholar] [CrossRef]
  21. Merlo, M.; Cannatà, A.; Pio Loco, C.; Stolfo, D.; Barbati, G.; Artico, J.; Gentile, P.; De Paris, V.; Ramani, F.; Zecchin, M.; et al. Contemporary Survival Trends and Etiological Characterization in Non-Ischemic Dilated Cardiomyopathy. Eur. J. Heart Fail. 2020, 22, 1111–1121. [Google Scholar] [CrossRef] [PubMed]
  22. Hazebroek, M.R.; Krapels, I.; Verdonschot, J.; van den Wijngaard, A.; Vanhoutte, E.; Hoos, M.; Snijders, L.; van Montfort, L.; Witjens, M.; Dennert, R.; et al. Prevalence of Pathogenic Gene Mutations and Prognosis Do Not Differ in Isolated Left Ventricular Dysfunction Compared with Dilated Cardiomyopathy. Circ. Heart Fail. 2018, 11, e004682. [Google Scholar] [CrossRef] [PubMed]
  23. Escobar-Lopez, L.; Ochoa, J.P.; Mirelis, J.G.; Espinosa, M.; Navarro, M.; Gallego-Delgado, M.; Barriales-Villa, R.; Robles-Mezcua, A.; Basurte-Elorz, M.T.; Gutiérrez García-Moreno, L.; et al. Association of Genetic Variants with Outcomes in Patients with Nonischemic Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2021, 78, 1682–1699. [Google Scholar] [CrossRef] [PubMed]
  24. Verdonschot, J.A.J.; Hazebroek, M.R.; Krapels, I.P.C.; Henkens, M.; Raafs, A.; Wang, P.; Merken, J.J.; Claes, G.R.F.; Vanhoutte, E.K.; van den Wijngaard, A.; et al. Implications of Genetic Testing in Dilated Cardiomyopathy. Circ. Genom. Precis. Med. 2020, 13, 476–487. [Google Scholar] [CrossRef] [PubMed]
  25. Merlo, M.; Pivetta, A.; Pinamonti, B.; Stolfo, D.; Zecchin, M.; Barbati, G.; Di Lenarda, A.; Sinagra, G. Long-Term Prognostic Impact of Therapeutic Strategies in Patients with Idiopathic Dilated Cardiomyopathy: Changing Mortality over the Last 30 Years. Eur. J. Heart Fail. 2014, 16, 317–324. [Google Scholar] [CrossRef]
  26. Michels, V.V.; Moll, P.P.; Miller, F.A.; Tajik, A.J.; Chu, J.S.; Driscoll, D.J.; Burnett, J.C.; Rodeheffer, R.J.; Chesebro, J.H.; Tazelaar, H.D. The Frequency of Familial Dilated Cardiomyopathy in a Series of Patients with Idiopathic Dilated Cardiomyopathy. N. Engl. J. Med. 1992, 326, 77–82. [Google Scholar] [CrossRef]
  27. Hey, T.M.; Rasmussen, T.B.; Madsen, T.; Aagaard, M.M.; Harbo, M.; Mølgaard, H.; Nielsen, S.K.; Haas, J.; Meder, B.; Møller, J.E.; et al. Clinical and Genetic Investigations of 109 Index Patients with Dilated Cardiomyopathy and 445 of Their Relatives. Circ. Heart Fail. 2020, 13, e006701. [Google Scholar] [CrossRef]
  28. Ebert, M.; Wijnmaalen, A.P.; de Riva, M.; Trines, S.A.; Androulakis, A.F.A.; Glashan, C.A.; Schalij, M.J.; Peter van Tintelen, J.; Jongbloed, J.D.H.; Zeppenfeld, K. Prevalence and Prognostic Impact of Pathogenic Variants in Patients with Dilated Cardiomyopathy Referred for Ventricular Tachycardia Ablation. JACC Clin. Electrophysiol. 2020, 6, 1103–1114. [Google Scholar] [CrossRef]
  29. Crispell, K.A.; Wray, A.; Ni, H.; Nauman, D.J.; Hershberger, R.E. Clinical Profiles of Four Large Pedigrees with Familial Dilated Cardiomyopathy: Preliminary Recommendations for Clinical Practice. J. Am. Coll. Cardiol. 1999, 34, 837–847. [Google Scholar] [CrossRef]
  30. Lukas Laws, J.; Lancaster, M.C.; Ben Shoemaker, M.; Stevenson, W.G.; Hung, R.R.; Wells, Q.; Marshall Brinkley, D.; Hughes, S.; Anderson, K.; Roden, D.; et al. Arrhythmias as Presentation of Genetic Cardiomyopathy. Circ. Res. 2022, 130, 1698–1722. [Google Scholar] [CrossRef]
  31. Mahon, N.G.; Murphy, R.T.; MacRae, C.A.; Caforio, A.L.; Elliott, P.M.; McKenna, W.J. Echocardiographic Evaluation in Asymptomatic Relatives of Patients with Dilated Cardiomyopathy Reveals Preclinical Disease. Ann. Intern. Med. 2005, 143, 108–115. [Google Scholar] [CrossRef] [PubMed]
  32. Huggins, G.S.; Kinnamon, D.D.; Haas, G.J.; Jordan, E.; Hofmeyer, M.; Kransdorf, E.; Ewald, G.A.; Morris, A.A.; Owens, A.; Lowes, B.; et al. Prevalence and Cumulative Risk of Familial Idiopathic Dilated Cardiomyopathy. JAMA 2022, 327, 454–463. [Google Scholar] [CrossRef] [PubMed]
  33. Ni, H.; Jordan, E.; Kinnamon, D.D.; Cao, J.; Haas, G.J.; Hofmeyer, M.; Kransdorf, E.; Ewald, G.A.; Morris, A.A.; Owens, A.; et al. Screening for Dilated Cardiomyopathy in At-Risk First-Degree Relatives. J. Am. Coll. Cardiol. 2023, 81, 2059–2071. [Google Scholar] [CrossRef] [PubMed]
  34. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2022, 79, e263–e421. [Google Scholar] [CrossRef] [PubMed]
  35. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed]
  36. Olson, T.M.; Michels, V.V.; Thibodeau, S.N.; Tai, Y.-S.; Keating, M.T. Actin Mutations in Dilated Cardiomyopathy, a Heritable Form of Heart Failure. Science 1998, 280, 750–752. [Google Scholar] [CrossRef]
  37. Kamisago, M.; Sharma, S.D.; DePalma, S.R.; Solomon, S.; Sharma, P.; McDonough, B.; Smoot, L.; Mullen, M.P.; Woolf, P.K.; Wigle, E.D.; et al. Mutations in Sarcomere Protein Genes as a Cause of Dilated Cardiomyopathy. N. Engl. J. Med. 2000, 343, 1688–1696. [Google Scholar] [CrossRef]
  38. Villard, E.; Duboscq-Bidot, L.; Charron, P.; Benaiche, A.; Conraads, V.; Sylvius, N.; Komajda, M. Mutation Screening in Dilated Cardiomyopathy: Prominent Role of the Beta Myosin Heavy Chain Gene. Eur. Heart J. 2005, 26, 794–803. [Google Scholar] [CrossRef]
  39. Tanigawa, G.; Jarcho, J.A.; Kass, S.; Solomon, S.D.; Vosberg, H.P.; Seidman, J.G.; Seidman, C.E. A Molecular Basis for Familial Hypertrophic Cardiomyopathy: An Alpha/Beta Cardiac Myosin Heavy Chain Hybrid Gene. Cell 1990, 62, 991–998. [Google Scholar] [CrossRef]
  40. Budde, B.S.; Binner, P.; Waldmüller, S.; Höhne, W.; Blankenfeldt, W.; Hassfeld, S.; Brömsen, J.; Dermintzoglou, A.; Wieczorek, M.; May, E.; et al. Noncompaction of the Ventricular Myocardium is Associated with a de novo Mutation in the Beta-Myosin Heavy Chain Gene. PLoS ONE 2007, 2, e1362. [Google Scholar] [CrossRef]
  41. Hoedemaekers, Y.M.; Caliskan, K.; Majoor-Krakauer, D.; van de Laar, I.; Michels, M.; Witsenburg, M.; ten Cate, F.J.; Simoons, M.L.; 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. [Google Scholar] [CrossRef] [PubMed]
  42. Watkins, H.; McKenna, W.J.; Thierfelder, L.; Suk, H.J.; Anan, R.; O’Donoghue, A.; Spirito, P.; Matsumori, A.; Moravec, C.S.; Seidman, J.G.; et al. Mutations in the Genes for Cardiac Troponin T and Alpha-Tropomyosin in Hypertrophic Cardiomyopathy. N. Engl. J. Med. 1995, 332, 1058–1064. [Google Scholar] [CrossRef] [PubMed]
  43. Thierfelder, L.; Watkins, H.; MacRae, C.; Lamas, R.; McKenna, W.; Vosberg, H.P.; Seidman, J.G.; Seidman, C.E. Alpha-Tropomyosin and Cardiac Troponin T Mutations Cause Familial Hypertrophic Cardiomyopathy: A Disease of the Sarcomere. Cell 1994, 77, 701–712. [Google Scholar] [CrossRef] [PubMed]
  44. Mogensen, J.; Murphy, R.T.; Shaw, T.; Bahl, A.; Redwood, C.; Watkins, H.; Burke, M.; Elliott, P.M.; McKenna, W.J. Severe Disease Expression of Cardiac Troponin C and T Mutations in Patients with Idiopathic Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2004, 44, 2033–2040. [Google Scholar] [CrossRef] [PubMed]
  45. Carballo, S.; Robinson, P.; Otway, R.; Fatkin, D.; Jongbloed, J.D.; de Jonge, N.; Blair, E.; van Tintelen, J.P.; Redwood, C.; Watkins, H. Identification and functional characterization of cardiac troponin I as a novel disease gene in autosomal dominant dilated cardiomyopathy. Circ. Res. 2009, 105, 375–382. [Google Scholar] [CrossRef]
  46. Kimura, A.; Harada, H.; Park, J.E.; Nishi, H.; Satoh, M.; Takahashi, M.; Hiroi, S.; Sasaoka, T.; Ohbuchi, N.; Nakamura, T.; et al. Mutations in the Cardiac Troponin I Gene Associated with Hypertrophic Cardiomyopathy. Nat. Genet. 1997, 16, 379–382. [Google Scholar] [CrossRef]
  47. Hoffmann, B.; Schmidt-Traub, H.; Perrot, A.; Osterziel, K.J.; Gessner, R. First Mutation in Cardiac Troponin C, L29Q, in a Patient with Hypertrophic Cardiomyopathy. Hum. Mutat. 2001, 17, 524. [Google Scholar] [CrossRef]
  48. Landstrom, A.P.; Parvatiyar, M.S.; Pinto, J.R.; Marquardt, M.L.; Bos, J.M.; Tester, D.J.; Ommen, S.R.; Potter, J.D.; Ackerman, M.J. Molecular and Functional Characterization of Novel Hypertrophic Cardiomyopathy Susceptibility Mutations in TNNC1-Encoded Troponin C. J. Mol. Cell Cardiol. 2008, 45, 281–288. [Google Scholar] [CrossRef]
  49. Parvatiyar, M.S.; Landstrom, A.P.; Figueiredo-Freitas, C.; Potter, J.D.; Ackerman, M.J.; Pinto, J.R. A Mutation in TNNC1-Encoded Cardiac Troponin C, TNNC1-A31S, Predisposes to Hypertrophic Cardiomyopathy and Ventricular Fibrillation. J. Biol. Chem. 2012, 287, 31845–31855. [Google Scholar] [CrossRef]
  50. Lakdawala, N.K.; Dellefave, L.; Redwood, C.S.; Sparks, E.; Cirino, A.L.; Depalma, S.; Colan, S.D.; Funke, B.; Zimmerman, R.S.; Robinson, P.; et al. Familial Dilated Cardiomyopathy Caused by an Alpha-Tropomyosin Mutation: The Distinctive Natural History of Sarcomeric Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2010, 55, 320–329. [Google Scholar] [CrossRef]
  51. Daehmlow, S.; Erdmann, J.; Knueppel, T.; Gille, C.; Froemmel, C.; Hummel, M.; Hetzer, R.; Regitz-Zagrosek, V. Novel Mutations in Sarcomeric Protein Genes in Dilated Cardiomyopathy. Biochem. Biophys. Res. Commun. 2002, 298, 116–120. [Google Scholar] [CrossRef] [PubMed]
  52. Møller, D.V.; Andersen, P.S.; Hedley, P.; Ersbøll, M.K.; Bundgaard, H.; Moolman-Smook, J.; Christiansen, M.; Køber, L. The Role of Sarcomere Gene Mutations in Patients with Idiopathic Dilated Cardiomyopathy. Eur. J. Hum. Genet. 2009, 17, 1241–1249. [Google Scholar] [CrossRef]
  53. Bonne, G.; Carrier, L.; Bercovici, J.; Cruaud, C.; Richard, P.; Hainque, B.; Gautel, M.; Labeit, S.; James, M.; Beckmann, J.; et al. Cardiac Myosin Binding Protein-C Gene Splice Acceptor Site Mutation Is Associated with Familial Hypertrophic Cardiomyopathy. Nat. Genet. 1995, 11, 438–440. [Google Scholar] [CrossRef] [PubMed]
  54. Watkins, H.; Conner, D.; Thierfelder, L.; Jarcho, J.A.; MacRae, C.; McKenna, W.J.; Maron, B.J.; Seidman, J.G.; Seidman, C.E. Mutations in the Cardiac Myosin Binding Protein-C Gene on Chromosome 11 Cause Familial Hypertrophic Cardiomyopathy. Nat. Genet. 1995, 11, 434–437. [Google Scholar] [CrossRef] [PubMed]
  55. Gerull, B.; Gramlich, M.; Atherton, J.; McNabb, M.; Trombitás, K.; Sasse-Klaassen, S.; Seidman, J.G.; Seidman, C.; Granzier, H.; Labeit, S.; et al. Mutations of TTN, Encoding the Giant Muscle Filament Titin, Cause Familial Dilated Cardiomyopathy. Nat. Genet. 2002, 30, 201–204. [Google Scholar] [CrossRef]
  56. Herman, D.S.; Lam, L.; Taylor, M.R.; Wang, L.; Teekakirikul, P.; Christodoulou, D.; Conner, L.; DePalma, S.R.; McDonough, B.; Sparks, E.; et al. Truncations of Titin Causing Dilated Cardiomyopathy. N. Engl. J. Med. 2012, 366, 619–628. [Google Scholar] [CrossRef]
  57. Taylor, M.; Graw, S.; Sinagra, G.; Barnes, C.; Slavov, D.; Brun, F.; Pinamonti, B.; Salcedo, E.E.; Sauer, W.; Pyxaras, S.; et al. Genetic Variation in Titin in Arrhythmogenic Right Ventricular Cardiomyopathy-Overlap Syndromes. Circulation 2011, 124, 876–885. [Google Scholar] [CrossRef]
  58. Hackman, P.; Vihola, A.; Haravuori, H.; Marchand, S.; Sarparanta, J.; De Seze, J.; Labeit, S.; Witt, C.; Peltonen, L.; Richard, I.; et al. Tibial Muscular Dystrophy Is a Titinopathy Caused by Mutations in TTN, the Gene Encoding the Giant Skeletal-Muscle Protein Titin. Am. J. Hum. Genet. 2002, 71, 492–500. [Google Scholar] [CrossRef]
  59. Hackman, P.; Marchand, S.; Sarparanta, J.; Vihola, A.; Pénisson-Besnier, I.; Eymard, B.; Pardal-Fernández, J.M.; Hammouda, E.-H.; Richard, I.; Illa, I.; et al. Truncating mutations in C-terminal titin may cause more severe tibial muscular dystrophy (TMD). Neuromuscul. Disord. 2008, 18, 922–928. [Google Scholar] [CrossRef]
  60. Pfeffer, G.; Elliott, H.R.; Griffin, H.; Barresi, R.; Miller, J.; Marsh, J.; Evilä, A.; Vihola, A.; Hackman, P.; Straub, V.; et al. Titin Mutation Segregates with Hereditary Myopathy with Early Respiratory Failure. Brain 2012, 135, 1695–1713. [Google Scholar] [CrossRef]
  61. Carmignac, V.; Salih, M.A.; Quijano-Roy, S.; Marchand, S.; Al Rayess, M.M.; Mukhtar, M.M.; Urtizberea, J.A.; Labeit, S.; Guicheney, P.; Leturcq, F.; et al. C-Terminal Titin Deletions Cause a Novel Early-Onset Myopathy with Fatal Cardiomyopathy. Ann. Neurol. 2007, 61, 340–351. [Google Scholar] [CrossRef] [PubMed]
  62. Huang, W.; Liang, J.; Yuan, C.C.; Kazmierczak, K.; Zhou, Z.; Morales, A.; McBride, K.L.; Fitzgerald-Butt, S.M.; Hershberger, R.E.; Szczesna-Cordary, D. Novel Familial Dilated Cardiomyopathy Mutation in MYL2 Affects the Structure and Function of Myosin Regulatory Light Chain. FEBS J. 2015, 282, 2379–2393. [Google Scholar] [CrossRef] [PubMed]
  63. Poetter, K.; Jiang, H.; Hassanzadeh, S.; Master, S.R.; Chang, A.; Dalakas, M.C.; Rayment, I.; Sellers, J.R.; Fananapazir, L.; Epstein, N.D. 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. [Google Scholar] [CrossRef] [PubMed]
  64. Purevjav, E.; Arimura, T.; Augustin, S.; Huby, A.C.; Takagi, K.; Nunoda, S.; Kearney, D.L.; Taylor, M.D.; Terasaki, F.; Bos, J.M.; et al. Molecular Basis for Clinical Heterogeneity in Inherited Cardiomyopathies Due to Myopalladin Mutations. Hum. Mol. Genet. 2012, 21, 2039–2053. [Google Scholar] [CrossRef]
  65. Duboscq-Bidot, L.; Xu, P.; Charron, P.; Neyroud, N.; Dilanian, G.; Millaire, A.; Bors, V.; Komajda, M.; Villard, E. Mutations in the Z-Band Protein Myopalladin Gene and Idiopathic Dilated Cardiomyopathy. Cardiovasc. Res. 2008, 77, 118–125. [Google Scholar] [CrossRef]
  66. Jakobs, P.M.; Hanson, E.L.; Crispell, K.A.; Toy, W.; Keegan, H.; Schilling, K.; Icenogle, T.B.; Litt, M.; Hershberger, R.E. Novel Lamin A/C Mutations in Two Families with Dilated Cardiomyopathy and Conduction System Disease. J. Card. Fail. 2001, 7, 249–256. [Google Scholar] [CrossRef]
  67. Fatkin, D.; MacRae, C.; Sasaki, T.; Wolff, M.R.; Porcu, M.; Frenneaux, M.; Atherton, J.; Vidaillet, H.J., Jr.; Spudich, S.; De Girolami, U.; et al. Missense Mutations in the Rod Domain of the Lamin A/C Gene as Causes of Dilated Cardiomyopathy and Conduction-System Disease. N. Engl. J. Med. 1999, 341, 1715–1724. [Google Scholar] [CrossRef]
  68. Quarta, G.; Syrris, P.; Ashworth, M.; Jenkins, S.; Zuborne Alapi, K.; Morgan, J.; Muir, A.; Pantazis, A.; McKenna, W.J.; Elliott, P.M. Mutations in the Lamin A/C Gene Mimic Arrhythmogenic Right Ventricular Cardiomyopathy. Eur. Heart J. 2012, 33, 1128–1136. [Google Scholar] [CrossRef]
  69. Bonne, G.; Di Barletta, M.R.; Varnous, S.; Bécane, H.M.; Hammouda, E.H.; Merlini, L.; Muntoni, F.; Greenberg, C.R.; Gary, F.; Urtizberea, J.A.; et al. Mutations in the Gene Encoding Lamin A/C Cause Autosomal Dominant Emery-Dreifuss Muscular Dystrophy. Nat. Genet. 1999, 21, 285–288. [Google Scholar] [CrossRef]
  70. Muchir, A.; Bonne, G.; van der Kooi, A.J.; van Meegen, M.; Baas, F.; Bolhuis, P.A.; de Visser, M.; Schwartz, K. Identification of Mutations in the Gene Encoding Lamins A/C in Autosomal Dominant Limb Girdle Muscular Dystrophy with Atrioventricular Conduction Disturbances (LGMD1B). Hum. Mol. Genet. 2000, 9, 1453–1459. [Google Scholar] [CrossRef]
  71. Quijano-Roy, S.; Mbieleu, B.; Bönnemann, C.G.; Jeannet, P.Y.; Colomer, J.; Clarke, N.F.; Cuisset, J.M.; Roper, H.; De Meirleir, L.; D’Amico, A.; et al. De Novo LMNA Mutations Cause a New Form of Congenital Muscular Dystrophy. Ann. Neurol. 2008, 64, 177–186. [Google Scholar] [CrossRef] [PubMed]
  72. Boone, P.M.; Yuan, B.; Gu, S.; Ma, Z.; Gambin, T.; Gonzaga-Jauregui, C.; Jain, M.; Murdock, T.J.; White, J.J.; Jhangiani, S.N.; et al. Hutterite-type cataract maps to chromosome 6p21.32-p21.31, cosegregates with a homozygous mutation in LEMD2, and is associated with sudden cardiac death. Mol. Genet. Genom. Med. 2016, 4, 77–94. [Google Scholar] [CrossRef] [PubMed]
  73. Abdelfatah, N.; Chen, R.; Duff, H.J.; Seifer, C.M.; Buffo, I.; Huculak, C.; Clarke, S.; Clegg, R.; Jassal, D.S.; Gordon, P.M.K.; et al. Characterization of a Unique Form of Arrhythmic Cardiomyopathy Caused by Recessive Mutation in LEMD2. JACC Basic Transl. Sci. 2019, 4, 204–221. [Google Scholar] [CrossRef] [PubMed]
  74. Ortiz-Genga, M.F.; Cuenca, S.; Dal Ferro, M.; Zorio, E.; Salgado-Aranda, R.; Climent, V.; Padrón-Barthe, L.; Duro-Aguado, I.; Jiménez-Jáimez, J.; Hidalgo-Olivares, V.M.; et al. Truncating FLNC Mutations Are Associated with High-Risk Dilated and Arrhythmogenic Cardiomyopathies. J. Am. Coll. Cardiol. 2016, 68, 2440–2451. [Google Scholar] [CrossRef] [PubMed]
  75. Begay, R.L.; Graw, S.L.; Sinagra, G.; Asimaki, A.; Rowland, T.J.; Slavov, D.B.; Gowan, K.; Jones, K.L.; Brun, F.; Merlo, M.; et al. Filamin C Truncation Mutations Are Associated with Arrhythmogenic Dilated Cardiomyopathy and Changes in the Cell-Cell Adhesion Structures. JACC Clin. Electrophysiol. 2018, 4, 504–514. [Google Scholar] [CrossRef]
  76. Valdés-Mas, R.; Gutiérrez-Fernández, A.; Gómez, J.; Coto, E.; Astudillo, A.; Puente, D.A.; Reguero, J.R.; Álvarez, V.; Morís, C.; León, D.; et al. Mutations in Filamin C Cause a New Form of Familial Hypertrophic Cardiomyopathy. Nat. Commun. 2014, 5, 5326. [Google Scholar] [CrossRef]
  77. Gómez, J.; Lorca, R.; Reguero, J.R.; Morís, C.; Martín, M.; Tranche, S.; Alonso, B.; Iglesias, S.; Alvarez, V.; Díaz-Molina, B.; et al. Screening of the Filamin C Gene in a Large Cohort of Hypertrophic Cardiomyopathy Patients. Circ. Cardiovasc. Genet. 2017, 10, e001584. [Google Scholar] [CrossRef]
  78. Vorgerd, M.; van der Ven, P.F.; Bruchertseifer, V.; Löwe, T.; Kley, R.A.; Schröder, R.; Lochmüller, H.; Himmel, M.; Koehler, K.; Fürst, D.O.; et al. A Mutation in the Dimerization Domain of Filamin C Causes a Novel Type of Autosomal Dominant Myofibrillar Myopathy. Am. J. Hum. Genet. 2005, 77, 297–304. [Google Scholar] [CrossRef]
  79. Muntoni, F.; Cau, M.; Ganau, A.; Congiu, R.; Arvedi, G.; Mateddu, A.; Marrosu, M.G.; Cianchetti, C.; Realdi, G.; Cao, A.; et al. Brief Report: Deletion of the Dystrophin Muscle-Promoter Region Associated with X-linked Dilated Cardiomyopathy. N. Engl. J. Med. 1993, 329, 921–925. [Google Scholar] [CrossRef]
  80. Ortiz-Lopez, R.; Li, H.; Su, J.; Goytia, V.; Towbin, J.A. Evidence for a Dystrophin Missense Mutation as a Cause of X-linked Dilated Cardiomyopathy. Circulation 1997, 95, 2434–2440. [Google Scholar] [CrossRef]
  81. Malhotra, S.B.; Hart, K.A.; Klamut, H.J.; Thomas, N.S.T.; Bodrug, S.E.; Burghes, A.H.M.; Bobrow, M.; Harper, P.S.; Thompson, M.W.; Ray, P.N.; et al. Frame-Shift Deletions in Patients with Duchenne and Becker Muscular Dystrophy. Science 1988, 242, 755–759. [Google Scholar] [CrossRef] [PubMed]
  82. Bione, S.; Maestrini, E.; Rivella, S.; Mancini, M.; Regis, S.; Romeo, G.; Toniolo, D. Identification of a Novel X-linked Gene Responsible for Emery-Dreifuss Muscular Dystrophy. Nat. Genet. 1994, 8, 323–327. [Google Scholar] [CrossRef]
  83. Bione, S.; Small, K.; Aksmanovic, V.M.; D’Urso, M.; Ciccodicola, A.; Merlini, L.; Morandi, L.; Kress, W.; Yates, J.R.; Warren, S.T.; et al. Identification of New Mutations in the Emery-Dreifuss Muscular Dystrophy Gene and Evidence for Genetic Heterogeneity of the Disease. Hum. Mol. Genet. 1995, 4, 1859–1863. [Google Scholar] [CrossRef] [PubMed]
  84. Goldfarb, L.G.; Park, K.Y.; Cervenáková, L.; Gorokhova, S.; Lee, H.S.; Vasconcelos, O.; Nagle, J.W.; Semino-Mora, C.; Sivakumar, K.; Dalakas, M.C. Missense Mutations in Desmin Associated with Familial Cardiac and Skeletal Myopathy. Nat. Genet. 1998, 19, 402–403. [Google Scholar] [CrossRef] [PubMed]
  85. Li, D.; Tapscoft, T.; Gonzalez, O.; Burch, P.E.; Quiñones, M.A.; Zoghbi, W.A.; Hill, R.; Bachinski, L.L.; Mann, D.L.; Roberts, R. Desmin Mutation Responsible for Idiopathic Dilated Cardiomyopathy. Circulation 1999, 100, 461–464. [Google Scholar] [CrossRef] [PubMed]
  86. Dalakas, M.C.; Park, K.Y.; Semino-Mora, C.; Lee, H.S.; Sivakumar, K.; Goldfarb, L.G. Desmin Myopathy, a Skeletal Myopathy with Cardiomyopathy Caused by Mutations in the Desmin Gene. N. Engl. J. Med. 2000, 342, 770–780. [Google Scholar] [CrossRef]
  87. Brodehl, A.; Dieding, M.; Biere, N.; Unger, A.; Klauke, B.; Walhorn, V.; Gummert, J.; Schulz, U.; Linke, W.A.; Gerull, B.; et al. Functional Characterization of the Novel DES Mutation p.L136P Associated with Dilated Cardiomyopathy Reveals a Dominant Filament Assembly Defect. J. Mol. Cell Cardiol. 2016, 91, 207–214. [Google Scholar] [CrossRef]
  88. Kulikova, O.; Brodehl, A.; Kiseleva, A.; Myasnikov, R.; Meshkov, A.; Stanasiuk, C.; Gärtner, A.; Divashuk, M.; Sotnikova, E.; Koretskiy, S.; et al. The Desmin (DES) Mutation p.A337P Is Associated with Left-Ventricular Non-Compaction Cardiomyopathy. Genes 2021, 12, 121. [Google Scholar] [CrossRef]
  89. Marakhonov, A.V.; Brodehl, A.; Myasnikov, R.P.; Sparber, P.A.; Kiseleva, A.V.; Kulikova, O.V.; Meshkov, A.N.; Zharikova, A.A.; Koretsky, S.N.; Kharlap, M.S.; et al. Noncompaction Cardiomyopathy Is Caused by a Novel In-Frame Desmin (DES) Deletion Mutation within the 1A Coiled-Coil Rod Segment Leading to a Severe Filament Assembly Defect. Hum. Mutat. 2019, 40, 734–741. [Google Scholar] [CrossRef]
  90. Brodehl, A.; Hain, C.; Flottmann, F.; Ratnavadivel, S.; Gaertner, A.; Klauke, B.; Kalinowski, J.; Körperich, H.; Gummert, J.; Paluszkiewicz, L.; et al. The Desmin Mutation DES-c.735G>C Causes Severe Restrictive Cardiomyopathy by Inducing In-Frame Skipping of Exon-3. Biomedicines 2021, 9, 1400. [Google Scholar] [CrossRef]
  91. Bione, S.; D’Adamo, P.; Maestrini, E.; Gedeon, A.K.; Bolhuis, P.A.; Toniolo, D. A Novel X-linked Gene, G4.5. is Responsible for Barth Syndrome. Nat. Genet. 1996, 12, 385–389. [Google Scholar] [CrossRef] [PubMed]
  92. Davey, K.M.; Parboosingh, J.S.; McLeod, D.R.; Chan, A.; Casey, R.; Ferreira, P.; Snyder, F.F.; Bridge, P.J.; Bernier, F.P. Mutation of DNAJC19, a Human Homologue of Yeast Inner Mitochondrial Membrane Co-Chaperones, Causes DCMA Syndrome, a Novel Autosomal Recessive Barth Syndrome-Like Condition. J. Med. Genet. 2006, 43, 385–393. [Google Scholar] [CrossRef]
  93. Ojala, T.; Polinati, P.; Manninen, T.; Hiippala, A.; Rajantie, J.; Karikoski, R.; Suomalainen, A.; Tyni, T. New Mutation of Mitochondrial DNAJC19 Causing Dilated and Noncompaction Cardiomyopathy, Anemia, Ataxia, and Male Genital Anomalies. Pediatr. Res. 2012, 72, 432–437. [Google Scholar] [CrossRef] [PubMed]
  94. Arimura, T.; Ishikawa, T.; Nunoda, S.; Kawai, S.; Kimura, A. Dilated Cardiomyopathy-Associated BAG3 Mutations Impair Z-Disc Assembly and Enhance Sensitivity to Apoptosis in Cardiomyocytes. Hum. Mutat. 2011, 32, 1481–1491. [Google Scholar] [CrossRef] [PubMed]
  95. Villard, E.; Perret, C.; Gary, F.; Proust, C.; Dilanian, G.; Hengstenberg, C.; Ruppert, V.; Arbustini, E.; Wichter, T.; Germain, M.; et al. A Genome-Wide Association Study Identifies Two Loci Associated with Heart Failure Due to Dilated Cardiomyopathy. Eur. Heart J. 2011, 32, 1065–1076. [Google Scholar] [CrossRef] [PubMed]
  96. Brauch, K.M.; Karst, M.L.; Herron, K.J.; de Andrade, M.; Pellikka, P.A.; Rodeheffer, R.J.; Michels, V.V.; Olson, T.M. Mutations in Ribonucleic Acid Binding Protein Gene Cause Familial Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2009, 54, 930–941. [Google Scholar] [CrossRef]
  97. Parikh, V.N.; Caleshu, C.; Reuter, C.; Lazzeroni, L.C.; Ingles, J.; Garcia, J.; McCaleb, K.; Adesiyun, T.; Sedaghat-Hamedani, F.; Kumar, S.; et al. Regional Variation in RBM20 Causes a Highly Penetrant Arrhythmogenic Cardiomyopathy. Circ. Heart Fail. 2019, 12, e005371. [Google Scholar] [CrossRef]
  98. Kirk, E.P.; Sunde, M.; Costa, M.W.; Rankin, S.A.; Wolstein, O.; Castro, M.L.; Butler, T.L.; Hyun, C.; Guo, G.; Otway, R.; et al. Mutations in Cardiac T-Box Factor Gene TBX20 Are Associated with Diverse Cardiac Pathologies, Including Defects of Septation and Valvulogenesis and Cardiomyopathy. Am. J. Hum. Genet. 2007, 81, 280–291. [Google Scholar] [CrossRef]
  99. Amor-Salamanca, A.; Santana Rodríguez, A.; Rasoul, H.; Rodríguez-Palomares, J.F.; Moldovan, O.; Hey, T.M.; Delgado, M.G.; Cuenca, D.L.; de Castro Campos, D.; Basurte-Elorz, M.T.; et al. Role of TBX20 Truncating Variants in Dilated Cardiomyopathy and Left Ventricular Noncompaction. Circ. Genom. Precis. Med. 2024, 17, e004404. [Google Scholar] [CrossRef]
  100. Garcia-Pavia, P.; Syrris, P.; Salas, C.; Evans, A.; Mirelis, J.G.; Cobo-Marcos, M.; Vilches, C.; Bornstein, B.; Segovia, J.; Alonso-Pulpon, L.; et al. Desmosomal Protein Gene Mutations in Patients with Idiopathic Dilated Cardiomyopathy Undergoing Cardiac Transplantation: A Clinicopathological Study. Heart 2011, 97, 1744–1752. [Google Scholar] [CrossRef]
  101. Coonar, A.S.; Protonotarios, N.; Tsatsopoulou, A.; Needham, E.W.; Houlston, R.S.; Cliff, S.; Otter, M.I.; Murday, V.A.; Mattu, R.K.; McKenna, W.J. Gene for Arrhythmogenic Right Ventricular Cardiomyopathy with Diffuse Nonepidermolytic Palmoplantar Keratoderma and Woolly Hair (Naxos Disease) Maps to 17q21. Circulation 1998, 97, 2049–2058. [Google Scholar] [CrossRef] [PubMed]
  102. McKoy, G.; Protonotarios, N.; Crosby, A.; Tsatsopoulou, A.; Anastasakis, A.; Coonar, A.; Norman, M.; Baboonian, C.; Jeffery, S.; McKenna, W.J. Identification of a Deletion in Plakoglobin in Arrhythmogenic Right Ventricular Cardiomyopathy with Palmoplantar Keratoderma and Woolly Hair (Naxos Disease). Lancet 2000, 355, 2119–2124. [Google Scholar] [CrossRef] [PubMed]
  103. Asimaki, A.; Syrris, P.; Wichter, T.; Matthias, P.; Saffitz, J.E.; McKenna, W.J. A Novel Dominant Mutation in Plakoglobin Causes Arrhythmogenic Right Ventricular Cardiomyopathy. Am. J. Hum. Genet. 2007, 81, 964–973. [Google Scholar] [CrossRef] [PubMed]
  104. Norgett, E.E.; Hatsell, S.J.; Carvajal-Huerta, L.; Cabezas, J.C.; Common, J.; Purkis, P.E.; Whittock, N.; Leigh, I.M.; Stevens, H.P.; Kelsell, D.P. Recessive Mutation in Desmoplakin Disrupts Desmoplakin-Intermediate Filament Interactions and Causes Dilated Cardiomyopathy, Woolly Hair and Keratoderma. Hum. Mol. Genet. 2000, 9, 2761–2766. [Google Scholar] [CrossRef] [PubMed]
  105. Rampazzo, A.; Nava, A.; Malacrida, S.; Beffagna, G.; Bauce, B.; Rossi, V.; Zimbello, R.; Simionati, B.; Basso, C.; Thiene, G.; et al. Mutation in Human Desmoplakin Domain Binding to Plakoglobin Causes a Dominant Form of Arrhythmogenic Right Ventricular Cardiomyopathy. Am. J. Hum. Genet. 2002, 71, 1200–1206. [Google Scholar] [CrossRef]
  106. Alcalai, R.; Metzger, S.; Rosenheck, S.; Meiner, V.; Chajek-Shaul, T. A Recessive Mutation in Desmoplakin Causes Arrhythmogenic Right Ventricular Dysplasia, Skin Disorder, and Woolly Hair. J. Am. Coll. Cardiol. 2003, 42, 319–327. [Google Scholar] [CrossRef]
  107. Elliott, P.; O’Mahony, C.; Syrris, P.; Evans, A.; Rivera Sorensen, C.; Sheppard, M.N.; Carr-White, G.; Pantazis, A.; McKenna, W.J. Prevalence of Desmosomal Protein Gene Mutations in Patients with Dilated Cardiomyopathy. Circ. Cardiovasc. Genet. 2010, 3, 314–322. [Google Scholar] [CrossRef]
  108. Gerull, B.; Heuser, A.; Wichter, T.; Paul, M.; Basson, C.T.; McDermott, D.A.; Lerman, B.B.; Markowitz, S.M.; Ellinor, P.T.; MacRae, C.A.; et al. Mutations in the Desmosomal Protein Plakophilin-2 Are Common in Arrhythmogenic Right Ventricular Cardiomyopathy. Nat. Genet. 2004, 36, 1162–1164. [Google Scholar] [CrossRef]
  109. Syrris, P.; Ward, D.; Asimaki, A.; Sen-Chowdhry, S.; Ebrahim, H.Y.; Evans, A.; Hitomi, N.; Norman, M.; Pantazis, A.; Shaw, A.L.; et al. Clinical Expression of Plakophilin-2 Mutations in Familial Arrhythmogenic Right Ventricular Cardiomyopathy. Circulation 2006, 113, 356–364. [Google Scholar] [CrossRef]
  110. Dalal, D.; Molin, L.H.; Piccini, J.; Tichnell, C.; James, C.; Bomma, C.; Prakasa, K.; Towbin, J.A.; Marcus, F.I.; Spevak, P.J.; et al. Clinical features of arrhythmogenic right ventricular dysplasia/cardiomyopathy associated with mutations in plakophilin-2. Circulation 2006, 113, 1641–1649. [Google Scholar] [CrossRef]
  111. Pilichou, K.; Nava, A.; Basso, C.; Beffagna, G.; Bauce, B.; Lorenzon, A.; Frigo, G.; Vettori, A.; Valente, M.; Towbin, J.; et al. Mutations in Desmoglein-2 Gene Are Associated with Arrhythmogenic Right Ventricular Cardiomyopathy. Circulation 2006, 113, 1171–1179. [Google Scholar] [CrossRef] [PubMed]
  112. Awad, M.M.; Dalal, D.; Cho, E.; Amat-Alarcon, N.; James, C.; Tichnell, C.; Tucker, A.; Russell, S.D.; Bluemke, D.A.; Dietz, H.C.; et al. DSG2 Mutations Contribute to Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy. Am. J. Hum. Genet. 2006, 79, 136–142. [Google Scholar] [CrossRef] [PubMed]
  113. Brodehl, A.; Meshkov, A.; Myasnikov, R.; Kiseleva, A.; Kulikova, O.; Klauke, B.; Sotnikova, E.; Stanasiuk, C.; Divashuk, M.; Pohl, G.M.; et al. Hemi- and Homozygous Loss-of-Function Mutations in DSG2 (Desmoglein-2) Cause Recessive Arrhythmogenic Cardiomyopathy with an Early Onset. Int. J. Mol. Sci. 2021, 22, 3786. [Google Scholar] [CrossRef] [PubMed]
  114. Heuser, A.; Plovie, E.R.; Ellinor, P.T.; Grossmann, K.S.; Shin, J.T.; Wichter, T.; Basson, C.T.; Lerman, B.B.; Sasse-Klaassen, S.; Thierfelder, L.; et al. Mutant Desmocollin-2 Causes Arrhythmogenic Right Ventricular Cardiomyopathy. Am. J. Hum. Genet. 2006, 79, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  115. Syrris, P.; Ward, D.; Evans, A.; Asimaki, A.; Gandjbakhch, E.; Sen-Chowdhry, S.; McKenna, W.J. Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy Associated with Mutations in the Desmosomal Gene Desmocollin-2. Am. J. Hum. Genet. 2006, 79, 978–984. [Google Scholar] [CrossRef]
  116. Brodehl, A.; Weiss, J.; Debus, J.D.; Stanasiuk, C.; Klauke, B.; Deutsch, M.A.; Fox, H.; Bax, J.; Ebbinghaus, H.; Gärtner, A.; et al. A Homozygous DSC2 Deletion Associated with Arrhythmogenic Cardiomyopathy Is Caused by Uniparental Isodisomy. J. Mol. Cell Cardiol. 2020, 141, 17–29. [Google Scholar] [CrossRef]
  117. Merner, N.D.; Hodgkinson, K.A.; Haywood, A.F.; Connors, S.; French, V.M.; Drenckhahn, J.D.; Kupprion, C.; Ramadanova, K.; Thierfelder, L.; McKenna, W.; et al. Arrhythmogenic Right Ventricular Cardiomyopathy Type 5 is a Fully Penetrant, Lethal Arrhythmic Disorder Caused by a Missense Mutation in the TMEM43 Gene. Am. J. Hum. Genet. 2008, 82, 809–821. [Google Scholar] [CrossRef]
  118. Baskin, B.; Skinner, J.R.; Sanatani, S.; Terespolsky, D.; Krahn, A.D.; Ray, P.N.; Scherer, S.W.; Hamilton, R.M. TMEM43 Mutations Associated with Arrhythmogenic Right Ventricular Cardiomyopathy in Non-Newfoundland Populations. Hum. Genet. 2013, 132, 1245–1252. [Google Scholar] [CrossRef]
  119. Christensen, A.H.; Andersen, C.B.; Tybjaerg-Hansen, A.; Haunso, S.; Svendsen, J.H. Mutation Analysis and Evaluation of the Cardiac Localization of TMEM43 in Arrhythmogenic Right Ventricular Cardiomyopathy. Clin. Genet. 2011, 80, 256–264. [Google Scholar] [CrossRef]
  120. Brodehl, A.; Rezazadeh, S.; Williams, T.; Munsie, N.M.; Liedtke, D.; Oh, T.; Ferrier, R.; Shen, Y.; Jones, S.J.M.; Stiegler, A.L.; et al. Mutations in ILK, Encoding Integrin-Linked Kinase, Are Associated with Arrhythmogenic Cardiomyopathy. Transl. Res. 2019, 208, 15–29. [Google Scholar] [CrossRef]
  121. Haghighi, K.; Kolokathis, F.; Pater, L.; Lynch, R.A.; Asahi, M.; Gramolini, A.O.; Fan, G.C.; Tsiapras, D.; Hahn, H.S.; Adamopoulos, S.; et al. Human Phospholamban Null Results in Lethal Dilated Cardiomyopathy Revealing a Critical Difference Between Mouse and Human. J. Clin. Investig. 2003, 111, 869–876. [Google Scholar] [CrossRef] [PubMed]
  122. Haghighi, K.; Kolokathis, F.; Gramolini, A.O.; Waggoner, J.R.; Pater, L.; Lynch, R.A.; Fan, G.C.; Tsiapras, D.; Parekh, R.R.; Dorn, G.W., 2nd; et al. A Mutation in the Human Phospholamban Gene, Deleting Arginine 14, Results in Lethal, Hereditary Cardiomyopathy. Proc. Natl. Acad. Sci. USA 2006, 103, 1388–1393. [Google Scholar] [CrossRef]
  123. Schmitt, J.P.; Kamisago, M.; Asahi, M.; Li, G.H.; Ahmad, F.; Mende, U.; Kranias, E.G.; MacLennan, D.H.; Seidman, J.G.; Seidman, C.E. Dilated Cardiomyopathy and Heart Failure Caused by a Mutation in Phospholamban. Science 2003, 299, 1410–1413. [Google Scholar] [CrossRef] [PubMed]
  124. McNair, W.P.; Ku, L.; Taylor, M.R.; Fain, P.R.; Dao, D.; Wolfel, E.; Mestroni, L. SCN5A Mutation Associated with Dilated Cardiomyopathy, Conduction Disorder, and Arrhythmia. Circulation 2004, 110, 2163–2167. [Google Scholar] [CrossRef] [PubMed]
  125. Olson, T.M.; Michels, V.V.; Ballew, J.D.; Reyna, S.P.; Karst, M.L.; Herron, K.J.; Horton, S.C.; Rodeheffer, R.J.; Anderson, J.L. Sodium Channel Mutations and Susceptibility to Heart Failure and Atrial Fibrillation. JAMA 2005, 293, 447–454. [Google Scholar] [CrossRef]
  126. Olson, T.M.; Keating, M.T. Mapping a Cardiomyopathy Locus to Chromosome 3p22-p25. J. Clin. Investig. 1996, 97, 528–532. [Google Scholar] [CrossRef]
  127. Walsh, R.; Buchan, R.; Wilk, A.; John, S.; Felkin, L.E.; Thomson, K.L.; Chiaw, T.H.; Loong, C.C.W.; Pua, C.J.; Raphael, C.; et al. Defining the Genetic Architecture of Hypertrophic Cardiomyopathy: Re-evaluating the Role of Non-Sarcomeric Genes. Eur. Heart J. 2017, 38, 3461–3468. [Google Scholar] [CrossRef]
  128. Thomson, K.L.; Ormondroyd, E.; Harper, A.R.; Dent, T.; McGuire, K.; Baksi, J.; Blair, E.; Brennan, P.; Buchan, R.; Bueser, T.; et al. Analysis of 51 Proposed Hypertrophic Cardiomyopathy Genes from Genome Sequencing Data in Sarcomere Negative Cases Has Negligible Diagnostic Yield. Genet. Med. 2019, 21, 1576–1584. [Google Scholar] [CrossRef]
  129. van Spaendonck-Zwarts, K.Y.; van Rijsingen, I.A.; van den Berg, M.P.; Lekanne Deprez, R.H.; Post, J.G.; van Mil, A.M.; Asselbergs, F.W.; Christiaans, I.; van Langen, I.M.; Wilde, A.A.; et al. Genetic Analysis in 418 Index Patients with Idiopathic Dilated Cardiomyopathy: Overview of 10 Years’ Experience. Eur. J. Heart Fail. 2013, 15, 628–636. [Google Scholar] [CrossRef]
  130. Pugh, T.J.; Kelly, M.A.; Gowrisankar, S.; Hynes, E.; Seidman, M.A.; Baxter, S.M.; Bowser, M.; Harrison, B.; Aaron, D.; Mahanta, L.M.; et al. The Landscape of Genetic Variation in Dilated Cardiomyopathy as Surveyed by Clinical DNA Sequencing. Genet. Med. 2014, 16, 601–608. [Google Scholar] [CrossRef]
  131. Ader, F.; De Groote, P.; Réant, P.; Rooryck-Thambo, C.; Dupin-Deguine, D.; Rambaud, C.; Khraiche, D.; Perret, C.; Pruny, J.F.; Mathieu-Dramard, M.; et al. FLNC Pathogenic Variants in Patients with Cardiomyopathies: Prevalence and Genotype-Phenotype Correlations. Clin. Genet. 2019, 96, 317–329. [Google Scholar] [CrossRef] [PubMed]
  132. Mazzarotto, F.; Tayal, U.; Buchan, R.J.; Midwinter, W.; Wilk, A.; Whiffin, N.; Govind, R.; Mazaika, E.; de Marvao, A.; Dawes, T.J.W.; et al. Reevaluating the Genetic Contribution of Monogenic Dilated Cardiomyopathy. Circulation 2020, 141, 387–398. [Google Scholar] [CrossRef] [PubMed]
  133. Tadros, R.; Francis, C.; Xu, X.; Vermeer, A.M.C.; Harper, A.R.; Huurman, R.; Kelu Bisabu, K.; Walsh, R.; Hoorntje, E.T.; te Rijdt, W.P.; et al. Shared Genetic Pathways Contribute to Risk of Hypertrophic and Dilated Cardiomyopathies with Opposite Directions of Effect. Nat. Genet. 2021, 53, 128–134. [Google Scholar] [CrossRef] [PubMed]
  134. Davis, J.; Davis, L.C.; Correll, R.N.; Makarewich, C.A.; Schwanekamp, J.A.; Moussavi-Harami, F.; Wang, D.; York, A.J.; Wu, H.; Houser, S.R.; et al. A Tension-Based Model Distinguishes Hypertrophic versus Dilated Cardiomyopathy. Cell 2016, 165, 1147–1159. [Google Scholar] [CrossRef] [PubMed]
  135. Dechat, T.; Pfleghaar, K.; Sengupta, K.; Shimi, T.; Shumaker, D.K.; Solimando, L.; Goldman, R.D. Nuclear Lamins: Major Factors in the Structural Organization and Function of the Nucleus and Chromatin. Genes. Dev. 2008, 22, 832–853. [Google Scholar] [CrossRef]
  136. Hasselberg, N.E.; Haland, T.F.; Saberniak, J.; Brekke, P.H.; Berge, K.E.; Leren, T.P.; Edvardsen, T.; Haugaa, K.H. Lamin A/C Cardiomyopathy: Young Onset, High Penetrance, and Frequent Need for Heart Transplantation. Eur. Heart J. 2018, 39, 853–860. [Google Scholar] [CrossRef]
  137. Arbustini, E.; Pilotto, A.; Repetto, A.; Grasso, M.; Negri, A.; Diegoli, M.; Campana, C.; Scelsi, L.; Baldini, E.; Gavazzi, A.; et al. Autosomal Dominant Dilated Cardiomyopathy with Atrioventricular Block: A Lamin A/C Defect-Related Disease. J. Am. Coll. Cardiol. 2002, 39, 981–990. [Google Scholar] [CrossRef]
  138. Kumar, S.; Baldinger, S.H.; Gandjbakhch, E.; Maury, P.; Sellal, J.M.; Androulakis, A.F.; Waintraub, X.; Charron, P.; Rollin, A.; Richard, P.; et al. Long-Term Arrhythmic and Nonarrhythmic Outcomes of Lamin A/C Mutation Carriers. J. Am. Coll. Cardiol. 2016, 68, 2299–2307. [Google Scholar] [CrossRef]
  139. Al-Khatib, S.M.; Stevenson, W.G.; Ackerman, M.J.; Bryant, W.J.; Callans, D.J.; Curtis, A.B.; Deal, B.J.; Dickfeld, T.; Field, M.E.; Fonarow, G.C.; et al. 2017 AHA/ACC/HRS Guideline for Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation 2018, 138, e272–e391. [Google Scholar] [CrossRef]
  140. Kusumoto, F.M.; Schoenfeld, M.H.; Barrett, C.; Edgerton, J.R.; Ellenbogen, K.A.; Gold, M.R.; Goldschlager, N.F.; Hamilton, R.M.; Joglar, J.A.; Kim, R.J.; et al. 2018 ACC/AHA/HRS Guideline on the Evaluation and Management of Patients with Bradycardia and Cardiac Conduction Delay: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation 2019, 140, e382–e482. [Google Scholar] [CrossRef]
  141. Zhou, Y.; Chen, Z.; Zhang, L.; Zhu, M.; Tan, C.; Zhou, X.; Evans, S.M.; Fang, X.; Feng, W.; Chen, J. Loss of Filamin C Is Catastrophic for Heart Function. Circulation 2020, 141, 869–871. [Google Scholar] [CrossRef] [PubMed]
  142. Agarwal, R.; Paulo, J.A.; Toepfer, C.N.; Ewoldt, J.K.; Sundaram, S.; Chopra, A.; Zhang, Q.; Gorham, J.; DePalma, S.R.; Chen, C.S.; et al. Filamin C Cardiomyopathy Variants Cause Protein and Lysosome Accumulation. Circ. Res. 2021, 129, 751–766. [Google Scholar] [CrossRef] [PubMed]
  143. Gao, S.; He, L.; Lam, C.K.; Taylor, M.R.G.; Mestroni, L.; Lombardi, R.; Chen, S.N. Filamin C Deficiency Impairs Sarcomere Stability and Activates Focal Adhesion Kinase through PDGFRA Signaling in Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2024, 13, 278. [Google Scholar] [CrossRef]
  144. Chen, S.N.; Lam, C.K.; Wan, Y.W.; Gao, S.; Malak, O.A.; Zhao, S.R.; Lombardi, R.; Ambardekar, A.V.; Bristow, M.R.; Cleveland, J.; et al. Activation of PDGFRA Signaling Contributes to Filamin C-Related Arrhythmogenic Cardiomyopathy. Sci. Adv. 2022, 8, eabk0052. [Google Scholar] [CrossRef]
  145. Janin, A.; N’Guyen, K.; Habib, G.; Dauphin, C.; Chanavat, V.; Bouvagnet, P.; Eschalier, R.; Streichenberger, N.; Chevalier, P.; Millat, G. Truncating Mutations on Myofibrillar Myopathies Causing Genes as Prevalent Molecular Explanations in Patients with Dilated Cardiomyopathy. Clin. Genet. 2017, 92, 616–623. [Google Scholar] [CrossRef] [PubMed]
  146. Cannie, D.E.; Syrris, P.; Protonotarios, A.; Bakalakos, A.; Pruny, J.F.; Ditaranto, R.; Martinez-Veira, C.; Larrañaga-Moreira, J.M.; Medo, K.; Bermúdez-Jiménez, F.J.; et al. Emery-Dreifuss Muscular Dystrophy Type 1 is Associated with a High Risk of Malignant Ventricular Arrhythmias and End-Stage Heart Failure. Eur. Heart J. 2023, 44, 5064–5073. [Google Scholar] [CrossRef] [PubMed]
  147. Skriver, S.V.; Krett, B.; Poulsen, N.S.; Krag, T.; Walas, H.R.; Christensen, A.H.; Bundgaard, H.; Vissing, J.; Vissing, C.R. Skeletal Muscle Involvement in Patients with Truncations of Titin and Familial Dilated Cardiomyopathy. JACC Heart Fail. 2024, 12, 740–753. [Google Scholar] [CrossRef] [PubMed]
  148. Garlid, A.O.; Schaffer, C.T.; Kim, J.; Bhatt, H.; Guevara-Gonzalez, V.; Ping, P. TAZ Encodes Tafazzin, a Transacylase Essential for Cardiolipin Formation and Central to the Etiology of Barth Syndrome. Gene 2020, 726, 144148. [Google Scholar] [CrossRef]
  149. Machiraju, P.; Degtiarev, V.; Patel, D.; Hazari, H.; Lowry, R.B.; Bedard, T.; Sinasac, D.; Brundler, M.A.; Greenway, S.C.; Khan, A. Phenotype and Pathology of Dilated Cardiomyopathy with Ataxia Syndrome in Children. J. Inherit. Metab. Dis. 2022, 45, 366–376. [Google Scholar] [CrossRef]
  150. Brenner, C.M.; Choudhary, M.; McCormick, M.G.; Cheung, D.; Landesberg, G.P.; Wang, J.F.; Song, J.; Martin, T.G.; Cheung, J.Y.; Qu, H.Q.; et al. BAG3: Nature’s Quintessential Multi-Functional Protein Functions as a Ubiquitous Intra-Cellular Glue. Cells 2023, 12, 937. [Google Scholar] [CrossRef]
  151. Feldman, A.M.; Begay, R.L.; Knezevic, T.; Myers, V.D.; Slavov, D.B.; Zhu, W.; Gowan, K.; Graw, S.L.; Jones, K.L.; Tilley, D.G.; et al. Decreased Levels of BAG3 in a Family with a Rare Variant and in Idiopathic Dilated Cardiomyopathy. J. Cell Physiol. 2014, 229, 1697–1702. [Google Scholar] [CrossRef] [PubMed]
  152. Domínguez, F.; Cuenca, S.; Bilińska, Z.; Toro, R.; Villard, E.; Barriales-Villa, R.; Ochoa, J.P.; Asselbergs, F.; Sammani, A.; Franaszczyk, M.; et al. Dilated Cardiomyopathy Due to BLC2-Associated Athanogene 3 (BAG3) Mutations. J. Am. Coll. Cardiol. 2018, 72, 2471–2481. [Google Scholar] [CrossRef] [PubMed]
  153. LeWinter, M.M.; Wu, Y.; Labeit, S.; Granzier, H. Cardiac Titin: Structure, Functions and Role in Disease. Clin. Chim. Acta 2007, 375, 1–9. [Google Scholar] [CrossRef] [PubMed]
  154. van der Ven, P.F.; Bartsch, J.W.; Gautel, M.; Jockusch, H.; Fürst, D.O. A Functional Knock-Out of Titin Results in Defective Myofibril Assembly. J. Cell Sci. 2000, 113, 1405–1414. [Google Scholar] [CrossRef]
  155. Musa, H.; Meek, S.; Gautel, M.; Peddie, D.; Smith, A.J.; Peckham, M. Targeted Homozygous Deletion of M-Band Titin in Cardiomyocytes Prevents Sarcomere Formation. J. Cell Sci. 2006, 119, 4322–4331. [Google Scholar] [CrossRef]
  156. Linke, W.A.; Popov, V.I.; Pollack, G.H. Passive and Active Tension in Single Cardiac Myofibrils. Biophys. J. 1994, 67, 782–792. [Google Scholar] [CrossRef]
  157. Helmes, M.; Trombitás, K.; Granzier, H. Titin Develops Restoring Force in Rat Cardiac Myocytes. Circ. Res. 1996, 79, 619–626. [Google Scholar] [CrossRef]
  158. Roberts, A.M.; Ware, J.S.; Herman, D.S.; Schafer, S.; Baksi, J.; Bick, A.G.; Buchan, R.J.; Walsh, R.; John, S.; Wilkinson, S.; et al. Integrated Allelic, Transcriptional, and Phenomic Dissection of the Cardiac Effects of Titin Truncations in Health and Disease. Sci. Transl. Med. 2015, 7, 270ra276. [Google Scholar] [CrossRef]
  159. Schafer, S.; de Marvao, A.; Adami, E.; Fiedler, L.R.; Ng, B.; Khin, E.; Rackham, O.J.; van Heesch, S.; Pua, C.J.; Kui, M.; et al. Titin-Truncating Variants Affect Heart Function in Disease Cohorts and the General Population. Nat. Genet. 2017, 49, 46–53. [Google Scholar] [CrossRef]
  160. Tayal, U.; Newsome, S.; Buchan, R.; Whiffin, N.; Halliday, B.; Lota, A.; Roberts, A.; Baksi, A.J.; Voges, I.; Midwinter, W.; et al. Phenotype and Clinical Outcomes of Titin Cardiomyopathy. J. Am. Coll. Cardiol. 2017, 70, 2264–2274. [Google Scholar] [CrossRef]
  161. Anderson, J.L.; Christensen, G.B.; Escobar, H.; Horne, B.D.; Knight, S.; Jacobs, V.; Afshar, K.; Hebl, V.B.; Muhlestein, J.B.; Knowlton, K.U.; et al. Discovery of TITIN Gene Truncating Variant Mutations and 5-Year Outcomes in Patients with Nonischemic Dilated Cardiomyopathy. Am. J. Cardiol. 2020, 137, 97–102. [Google Scholar] [CrossRef]
  162. Xiao, L.; Li, C.; Sun, Y.; Chen, Y.; Wei, H.; Hu, D.; Yu, T.; Li, X.; Jin, L.; Shi, L.; et al. Clinical Significance of Variants in the TTN Gene in a Large Cohort of Patients with Sporadic Dilated Cardiomyopathy. Front. Cardiovasc. Med. 2021, 8, 657689. [Google Scholar] [CrossRef]
  163. Heliö, K.; Cicerchia, M.; Hathaway, J.; Tommiska, J.; Huusko, J.; Saarinen, I.; Koskinen, L.; Muona, M.; Kytölä, V.; Djupsjöbacka, J.; et al. Diagnostic Yield of Genetic Testing in a Multinational Heterogeneous Cohort of 2088 DCM Patients. Front. Cardiovasc. Med. 2023, 10, 1254272. [Google Scholar] [CrossRef] [PubMed]
  164. Patel, P.N.; Ito, K.; Willcox, J.A.L.; Haghighi, A.; Jang, M.Y.; Gorham, J.M.; DePalma, S.R.; Lam, L.; McDonough, B.; Johnson, R.; et al. Contribution of Noncanonical Splice Variants to TTN Truncating Variant Cardiomyopathy. Circ. Genom. Precis. Med. 2021, 14, e003389. [Google Scholar] [CrossRef] [PubMed]
  165. Hastings, R.; de Villiers, C.P.; Hooper, C.; Ormondroyd, L.; Pagnamenta, A.; Lise, S.; Salatino, S.; Knight, S.J.; Taylor, J.C.; Thomson, K.L.; et al. Combination of Whole Genome Sequencing, Linkage, and Functional Studies Implicates a Missense Mutation in Titin as a Cause of Autosomal Dominant Cardiomyopathy with Features of Left Ventricular Noncompaction. Circ. Cardiovasc. Genet. 2016, 9, 426–435. [Google Scholar] [CrossRef]
  166. Domínguez, F.; Lalaguna, L.; Martínez-Martín, I.; Piqueras-Flores, J.; Rasmussen, T.B.; Zorio, E.; Giovinazzo, G.; Prados, B.; Ochoa, J.P.; Bornstein, B.; et al. Titin Missense Variants as a Cause of Familial Dilated Cardiomyopathy. Circulation 2023, 147, 1711–1713. [Google Scholar] [CrossRef] [PubMed]
  167. Hinson, J.T.; Chopra, A.; Nafissi, N.; Polacheck, W.J.; Benson, C.C.; Swist, S.; Gorham, J.; Yang, L.; Schafer, S.; Sheng, C.C.; et al. Heart Disease: Titin Mutations in iPS Cells Define Sarcomere Insufficiency as a Cause of Dilated Cardiomyopathy. Science 2015, 349, 982–986. [Google Scholar] [CrossRef] [PubMed]
  168. Begay, R.L.; Graw, S.; Sinagra, G.; Merlo, M.; Slavov, D.; Gowan, K.; Jones, K.L.; Barbati, G.; Spezzacatene, A.; Brun, F.; et al. Role of Titin Missense Variants in Dilated Cardiomyopathy. J. Am. Heart Assoc. 2015, 4, 11. [Google Scholar] [CrossRef] [PubMed]
  169. Akinrinade, O.; Heliö, T.; Lekanne Deprez, R.H.; Jongbloed, J.D.H.; Boven, L.G.; van den Berg, M.P.; Pinto, Y.M.; Alastalo, T.P.; Myllykangas, S.; Spaendonck-Zwarts, K.V.; et al. Relevance of Titin Missense and Non-Frameshifting Insertions/Deletions Variants in Dilated Cardiomyopathy. Sci. Rep. 2019, 9, 4093. [Google Scholar] [CrossRef]
  170. Norton, N.; Li, D.; Rampersaud, E.; Morales, A.; Martin, E.R.; Zuchner, S.; Guo, S.; Gonzalez, M.; Hedges, D.J.; Robertson, P.D.; et al. Exome Sequencing and Genome-Wide Linkage Analysis in 17 Families Illustrate the Complex Contribution of TTN Truncating Variants to Dilated Cardiomyopathy. Circ. Cardiovasc. Genet. 2013, 6, 144–153. [Google Scholar] [CrossRef]
  171. Kornienko, J.; Rodríguez-Martínez, M.; Fenzl, K.; Hinze, F.; Schraivogel, D.; Grosch, M.; Tunaj, B.; Lindenhofer, D.; Schraft, L.; Kueblbeck, M.; et al. Mislocalization of Pathogenic RBM20 Variants in Dilated Cardiomyopathy is Caused by Loss-of-Interaction with Transportin-3. Nat. Commun. 2023, 14, 4312. [Google Scholar] [CrossRef] [PubMed]
  172. Guo, W.; Schafer, S.; Greaser, M.L.; Radke, M.H.; Liss, M.; Govindarajan, T.; Maatz, H.; Schulz, H.; Li, S.; Parrish, A.M.; et al. RBM20, a Gene for Hereditary Cardiomyopathy, Regulates Titin Splicing. Nat. Med. 2012, 18, 766–773. [Google Scholar] [CrossRef] [PubMed]
  173. Beqqali, A.; Bollen, I.A.; Rasmussen, T.B.; van den Hoogenhof, M.M.; van Deutekom, H.W.; Schafer, S.; Haas, J.; Meder, B.; Sørensen, K.E.; van Oort, R.J.; et al. A Mutation in the Glutamate-Rich Region of RNA-Binding Motif Protein 20 Causes Dilated Cardiomyopathy Through Missplicing of Titin and Impaired Frank-Starling Mechanism. Cardiovasc. Res. 2016, 112, 452–463. [Google Scholar] [CrossRef] [PubMed]
  174. Greaser, M.L.; Warren, C.M.; Esbona, K.; Guo, W.; Duan, Y.; Parrish, A.M.; Krzesinski, P.R.; Norman, H.S.; Dunning, S.; Fitzsimons, D.P.; et al. Mutation That Dramatically Alters Rat Titin Isoform Expression and Cardiomyocyte Passive Tension. J. Mol. Cell Cardiol. 2008, 44, 983–991. [Google Scholar] [CrossRef] [PubMed]
  175. Wyles, S.P.; Li, X.; Hrstka, S.C.; Reyes, S.; Oommen, S.; Beraldi, R.; Edwards, J.; Terzic, A.; Olson, T.M.; Nelson, T.J. Modeling Structural and Functional Deficiencies of RBM20 Familial Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Hum. Mol. Genet. 2016, 25, 254–265. [Google Scholar] [CrossRef]
  176. Cannie, D.E.; Protonotarios, A.; Bakalakos, A.; Syrris, P.; Lorenzini, M.; De Stavola, B.; Bjerregaard, L.; Dybro, A.M.; Hey, T.M.; Hansen, F.G.; et al. Risks of Ventricular Arrhythmia and Heart Failure in Carriers of RBM20 Variants. Circ. Genom. Precis. Med. 2023, 16, 434–441. [Google Scholar] [CrossRef]
  177. Marcus, F.I.; Fontaine, G.H.; Guiraudon, G.; Frank, R.; Laurenceau, J.L.; Malergue, C.; Grosgogeat, Y. Right Ventricular Dysplasia: A Report of 24 Adult Cases. Circulation 1982, 65, 384–398. [Google Scholar] [CrossRef]
  178. Corrado, D.; Link, M.S.; Calkins, H. Arrhythmogenic Right Ventricular Cardiomyopathy. N. Engl. J. Med. 2017, 376, 61–72. [Google Scholar] [CrossRef]
  179. Kapplinger, J.D.; Landstrom, A.P.; Salisbury, B.A.; Callis, T.E.; Pollevick, G.D.; Tester, D.J.; Cox, M.G.; Bhuiyan, Z.; Bikker, H.; Wiesfeld, A.C.; et al. Distinguishing Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia-Associated Mutations From Background Genetic Noise. J. Am. Coll. Cardiol. 2011, 57, 2317–2327. [Google Scholar] [CrossRef]
  180. Groeneweg, J.A.; Bhonsale, A.; James, C.A.; te Riele, A.S.; Dooijes, D.; Tichnell, C.; Murray, B.; Wiesfeld, A.C.; Sawant, A.C.; Kassamali, B.; et al. Clinical Presentation, Long-Term Follow-Up, and Outcomes of 1001 Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy Patients and Family Members. Circ. Cardiovasc. Genet. 2015, 8, 437–446. [Google Scholar] [CrossRef]
  181. Sen-Chowdhry, S.; Syrris, P.; Prasad, S.K.; Hughes, S.E.; Merrifield, R.; Ward, D.; Pennell, D.J.; McKenna, W.J. Left-Dominant Arrhythmogenic Cardiomyopathy: An Under-Recognized Clinical Entity. J. Am. Coll. Cardiol. 2008, 52, 2175–2187. [Google Scholar] [CrossRef] [PubMed]
  182. Bhonsale, A.; Groeneweg, J.A.; James, C.A.; Dooijes, D.; Tichnell, C.; Jongbloed, J.D.; Murray, B.; te Riele, A.S.; van den Berg, M.P.; Bikker, H.; et al. Impact of Genotype on Clinical Course in Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy-Associated Mutation Carriers. Eur. Heart J. 2015, 36, 847–855. [Google Scholar] [CrossRef] [PubMed]
  183. Berte, B.; Denis, A.; Amraoui, S.; Yamashita, S.; Komatsu, Y.; Pillois, X.; Sacher, F.; Mahida, S.; Wielandts, J.Y.; Sellal, J.M.; et al. Characterization of the Left-Sided Substrate in Arrhythmogenic Right Ventricular Cardiomyopathy. Circ. Arrhythm. Electrophysiol. 2015, 8, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  184. Brun, F.; Gigli, M.; Graw, S.L.; Judge, D.P.; Merlo, M.; Murray, B.; Calkins, H.; Sinagra, G.; Taylor, M.R.; Mestroni, L.; et al. FLNC Truncations Cause Arrhythmogenic Right Ventricular Cardiomyopathy. J. Med. Genet. 2020, 57, 254–257. [Google Scholar] [CrossRef] [PubMed]
  185. Hall, C.L.; Akhtar, M.M.; Sabater-Molina, M.; Futema, M.; Asimaki, A.; Protonotarios, A.; Dalageorgou, C.; Pittman, A.M.; Suarez, M.P.; Aguilera, B.; et al. Filamin C Variants Are Associated with a Distinctive Clinical and Immunohistochemical Arrhythmogenic Cardiomyopathy Phenotype. Int. J. Cardiol. 2020, 307, 101–108. [Google Scholar] [CrossRef]
  186. Towbin, J.A.; McKenna, W.J.; Abrams, D.J.; Ackerman, M.J.; Calkins, H.; Darrieux, F.C.C.; Daubert, J.P.; de Chillou, C.; DePasquale, E.C.; Desai, M.Y.; et al. 2019 HRS Expert Consensus Statement on Evaluation, Risk Stratification, and Management of Arrhythmogenic Cardiomyopathy. Heart Rhythm. 2019, 16, e301–e372. [Google Scholar] [CrossRef]
  187. Wilde, A.A.M.; Amin, A.S. Clinical Spectrum of SCN5A Mutations: Long QT Syndrome, Brugada Syndrome, and Cardiomyopathy. JACC Clin. Electrophysiol. 2018, 4, 569–579. [Google Scholar] [CrossRef]
  188. Norman, M.; Simpson, M.; Mogensen, J.; Shaw, A.; Hughes, S.; Syrris, P.; Sen-Chowdhry, S.; Rowland, E.; Crosby, A.; McKenna, W.J. Novel Mutation in Desmoplakin Causes Arrhythmogenic Left Ventricular Cardiomyopathy. Circulation 2005, 112, 636–642. [Google Scholar] [CrossRef]
  189. Smith, E.D.; Lakdawala, N.K.; Papoutsidakis, N.; Aubert, G.; Mazzanti, A.; McCanta, A.C.; Agarwal, P.P.; Arscott, P.; Dellefave-Castillo, L.M.; Vorovich, E.E.; et al. Desmoplakin Cardiomyopathy, a Fibrotic and Inflammatory Form of Cardiomyopathy Distinct From Typical Dilated or Arrhythmogenic Right Ventricular Cardiomyopathy. Circulation 2020, 141, 1872–1884. [Google Scholar] [CrossRef]
  190. McColl, H.; Cordina, R.; Lal, S.; Parker, M.; Hunyor, I.; Medi, C.; Gray, B. Recurrent Immunosuppressive-Responsive Myocarditis in a Patient with Desmoplakin Cardiomyopathy: A Case Report. Eur. Heart J. Case Rep. 2024, 8, ytae129. [Google Scholar] [CrossRef]
  191. Sedmera, D.; Pexieder, T.; Vuillemin, M.; Thompson, R.P.; Anderson, R.H. Developmental Patterning of the Myocardium. Anat. Rec. 2000, 258, 319–337. [Google Scholar] [CrossRef]
  192. Weir-McCall, J.R.; Yeap, P.M.; Papagiorcopulo, C.; Fitzgerald, K.; Gandy, S.J.; Lambert, M.; Belch, J.J.; Cavin, I.; Littleford, R.; Macfarlane, J.A.; et al. Left Ventricular Noncompaction: Anatomical Phenotype or Distinct Cardiomyopathy? J. Am. Coll. Cardiol. 2016, 68, 2157–2165. [Google Scholar] [CrossRef] [PubMed]
  193. de la Chica, J.A.; Gómez-Talavera, S.; García-Ruiz, J.M.; García-Lunar, I.; Oliva, B.; Fernández-Alvira, J.M.; López-Melgar, B.; Sánchez-González, J.; de la Pompa, J.L.; Mendiguren, J.M.; et al. Association Between Left Ventricular Noncompaction and Vigorous Physical Activity. J. Am. Coll. Cardiol. 2020, 76, 1723–1733. [Google Scholar] [CrossRef] [PubMed]
  194. Gati, S.; Chandra, N.; Bennett, R.L.; Reed, M.; Kervio, G.; Panoulas, V.F.; Ghani, S.; Sheikh, N.; Zaidi, A.; Wilson, M.; et al. Increased Left Ventricular Trabeculation in Highly Trained Athletes: Do We Need More Stringent Criteria for the Diagnosis of Left Ventricular Non-Compaction in Athletes? Heart 2013, 99, 401–408. [Google Scholar] [CrossRef] [PubMed]
  195. Caselli, S.; Ferreira, D.; Kanawati, E.; Di Paolo, F.; Pisicchio, C.; Attenhofer Jost, C.; Spataro, A.; Jenni, R.; Pelliccia, A. Prominent Left Ventricular Trabeculations in Competitive Athletes: A Proposal for Risk Stratification and Management. Int. J. Cardiol. 2016, 223, 590–595. [Google Scholar] [CrossRef]
  196. Gati, S.; Papadakis, M.; Papamichael, N.D.; Zaidi, A.; Sheikh, N.; Reed, M.; Sharma, R.; Thilaganathan, B.; Sharma, S. Reversible De Novo Left Ventricular Trabeculations in Pregnant Women: Implications for the Diagnosis of Left Ventricular Noncompaction in Low-Risk Populations. Circulation 2014, 130, 475–483. [Google Scholar] [CrossRef]
  197. D’Ascenzi, F.; Pelliccia, A.; Natali, B.M.; Bonifazi, M.; Mondillo, S. Exercise-Induced Left-Ventricular Hypertrabeculation in Athlete’s Heart. Int. J. Cardiol. 2015, 181, 320–322. [Google Scholar] [CrossRef]
  198. Petersen, S.E.; Jensen, B.; Aung, N.; Friedrich, M.G.; McMahon, C.J.; Mohiddin, S.A.; Pignatelli, R.H.; Ricci, F.; Anderson, R.H.; Bluemke, D.A. Excessive Trabeculation of the Left Ventricle: JACC: Cardiovascular Imaging Expert Panel Paper. JACC Cardiovasc. Imaging 2023, 16, 408–425. [Google Scholar] [CrossRef]
  199. Ross, S.B.; Jones, K.; Blanch, B.; Puranik, R.; McGeechan, K.; Barratt, A.; Semsarian, C. A Systematic Review and Meta-Analysis of the Prevalence of Left Ventricular Non-Compaction in Adults. Eur. Heart J. 2020, 41, 1428–1436. [Google Scholar] [CrossRef]
  200. Biagini, E.; Ragni, L.; Ferlito, M.; Pasquale, F.; Lofiego, C.; Leone, O.; Rocchi, G.; Perugini, E.; Zagnoni, S.; Branzi, A.; et al. Different Types of Cardiomyopathy Associated with Isolated Ventricular Noncompaction. Am. J. Cardiol. 2006, 98, 821–824. [Google Scholar] [CrossRef]
  201. Kelley-Hedgepeth, A.; Towbin, J.A.; Maron, M.S. Images in Cardiovascular Medicine: Overlapping Phenotypes—Left Ventricular Noncompaction and Hypertrophic Cardiomyopathy. Circulation 2009, 119, e588–e589. [Google Scholar] [CrossRef] [PubMed]
  202. Gabra, B.; Anavekar, N.S.; Haaf, P. Hypertrophic Cardiomyopathy and Left Ventricular Non-Compaction Cardiomyopathy: Two in One. Eur. Heart J. 2023, 44, 327. [Google Scholar] [CrossRef] [PubMed]
  203. Luxán, G.; Casanova, J.C.; Martínez-Poveda, B.; Prados, B.; D’Amato, G.; MacGrogan, D.; Gonzalez-Rajal, A.; Dobarro, D.; Torroja, C.; Martinez, F.; et al. Mutations in the NOTCH Pathway Regulator MIB1 Cause Left Ventricular Noncompaction Cardiomyopathy. Nat. Med. 2013, 19, 193–201. [Google Scholar] [CrossRef] [PubMed]
  204. Siguero-Álvarez, M.; Salguero-Jiménez, A.; Grego-Bessa, J.; de la Barrera, J.; MacGrogan, D.; Prados, B.; Sánchez-Sáez, F.; Piñeiro-Sabarís, R.; Felipe-Medina, N.; Torroja, C.; et al. A Human Hereditary Cardiomyopathy Shares a Genetic Substrate with Bicuspid Aortic Valve. Circulation 2023, 147, 47–65. [Google Scholar] [CrossRef] [PubMed]
  205. Vatta, M.; Mohapatra, B.; Jimenez, S.; Sanchez, X.; Faulkner, G.; Perles, Z.; Sinagra, G.; Lin, J.H.; Vu, T.M.; Zhou, Q.; et al. Mutations in Cypher/ZASP in Patients with Dilated Cardiomyopathy and Left Ventricular Non-Compaction. J. Am. Coll. Cardiol. 2003, 42, 2014–2027. [Google Scholar] [CrossRef]
  206. Hermida-Prieto, M.; Monserrat, L.; Castro-Beiras, A.; Laredo, R.; Soler, R.; Peteiro, J.; Rodríguez, E.; Bouzas, B.; Alvarez, N.; Muñiz, J.; et al. Familial Dilated Cardiomyopathy and Isolated Left Ventricular Noncompaction Associated with Lamin A/C Gene Mutations. Am. J. Cardiol. 2004, 94, 50–54. [Google Scholar] [CrossRef]
  207. Monserrat, L.; Hermida-Prieto, M.; Fernandez, X.; Rodríguez, I.; Dumont, C.; Cazón, L.; Cuesta, M.G.; Gonzalez-Juanatey, C.; Peteiro, J.; Alvarez, N.; et al. Mutation in the Alpha-Cardiac Actin Gene Associated with Apical Hypertrophic Cardiomyopathy, Left Ventricular Non-Compaction, and Septal Defects. Eur. Heart J. 2007, 28, 1953–1961. [Google Scholar] [CrossRef]
  208. Frustaci, A.; De Luca, A.; Guida, V.; Biagini, T.; Mazza, T.; Gaudio, C.; Letizia, C.; Russo, M.A.; Galea, N.; Chimenti, C. Novel α-Actin Gene Mutation p.(Ala21Val) Causing Familial Hypertrophic Cardiomyopathy, Myocardial Noncompaction, and Transmural Crypts. Clinical-Pathologic Correlation. J. Am. Heart Assoc. 2018, 7, 4. [Google Scholar] [CrossRef]
  209. Sedaghat-Hamedani, F.; Haas, J.; Zhu, F.; Geier, C.; Kayvanpour, E.; Liss, M.; Lai, A.; Frese, K.; Pribe-Wolferts, R.; Amr, A.; et al. Clinical Genetics and Outcome of Left Ventricular Non-Compaction Cardiomyopathy. Eur. Heart J. 2017, 38, 3449–3460. [Google Scholar] [CrossRef]
  210. van Waning, J.I.; Caliskan, K.; Hoedemaekers, Y.M.; van Spaendonck-Zwarts, K.Y.; Baas, A.F.; Boekholdt, S.M.; van Melle, J.P.; Teske, A.J.; Asselbergs, F.W.; Backx, A.; et al. Genetics, Clinical Features, and Long-Term Outcome of Noncompaction Cardiomyopathy. J. Am. Coll. Cardiol. 2018, 71, 711–722. [Google Scholar] [CrossRef]
  211. Kayvanpour, E.; Sedaghat-Hamedani, F.; Gi, W.T.; Tugrul, O.F.; Amr, A.; Haas, J.; Zhu, F.; Ehlermann, P.; Uhlmann, L.; Katus, H.A.; et al. Clinical and Genetic Insights into Non-Compaction: A Meta-Analysis and Systematic Review on 7598 Individuals. Clin. Res. Cardiol. 2019, 108, 1297–1308. [Google Scholar] [CrossRef] [PubMed]
  212. Mazzarotto, F.; Hawley, M.H.; Beltrami, M.; Beekman, L.; de Marvao, A.; McGurk, K.A.; Statton, B.; Boschi, B.; Girolami, F.; Roberts, A.M.; et al. Systematic large-scale assessment of the genetic architecture of left ventricular noncompaction reveals diverse etiologies. Genet. Med. 2021, 23, 856–864. [Google Scholar] [CrossRef] [PubMed]
  213. Casas, G.; Limeres, J.; Oristrell, G.; Gutierrez-Garcia, L.; Andreini, D.; Borregan, M.; Larrañaga-Moreira, J.M.; Lopez-Sainz, A.; Codina-Solà, M.; Teixido-Tura, G.; et al. Clinical Risk Prediction in Patients with Left Ventricular Myocardial Noncompaction. J. Am. Coll. Cardiol. 2021, 78, 643–662. [Google Scholar] [CrossRef] [PubMed]
  214. Hershberger, R.E.; Lindenfeld, J.; Mestroni, L.; Seidman, C.E.; Taylor, M.R.G.; Towbin, J.A. Genetic Evaluation of Cardiomyopathy—A Heart Failure Society of America Practice Guideline. J. Card. Fail. 2009, 15, 83–97. [Google Scholar] [CrossRef]
  215. Arbelo, E.; Protonotarios, A.; Gimeno, J.R.; Arbustini, E.; Barriales-Villa, R.; Basso, C.; Bezzina, C.R.; Biagini, E.; Blom, N.A.; de Boer, R.A.; et al. 2023 ESC Guidelines for the Management of Cardiomyopathies. Eur. Heart J. 2023, 44, 3503–3626. [Google Scholar] [CrossRef]
  216. Mogensen, J.; Kubo, T.; Duque, M.; Uribe, W.; Shaw, A.; Murphy, R.; Gimeno, J.R.; Elliott, P.; McKenna, W.J. Idiopathic Restrictive Cardiomyopathy Is Part of the Clinical Expression of Cardiac Troponin I Mutations. J. Clin. Investig. 2003, 111, 209–216. [Google Scholar] [CrossRef]
  217. Kaski, J.P.; Syrris, P.; Burch, M.; Tomé-Esteban, M.T.; Fenton, M.; Christiansen, M.; Andersen, P.S.; Sebire, N.; Ashworth, M.; Deanfield, J.E.; et al. Idiopathic Restrictive Cardiomyopathy in Children Is Caused by Mutations in Cardiac Sarcomere Protein Genes. Heart 2008, 94, 1478–1484. [Google Scholar] [CrossRef]
  218. Bagnall, R.D.; Yeates, L.; Semsarian, C. The Role of Large Gene Deletions and Duplications in MYBPC3 and TNNT2 in Patients with Hypertrophic Cardiomyopathy. Int. J. Cardiol. 2010, 145, 150–153. [Google Scholar] [CrossRef]
  219. Menon, S.C.; Michels, V.V.; Pellikka, P.A.; Ballew, J.D.; Karst, M.L.; Herron, K.J.; Nelson, S.M.; Rodeheffer, R.J.; Olson, T.M. Cardiac Troponin T Mutation in Familial Cardiomyopathy with Variable Remodeling and Restrictive Physiology. Clin. Genet. 2008, 74, 445–454. [Google Scholar] [CrossRef]
  220. De Bortoli, M.; Vio, R.; Basso, C.; Calore, M.; Minervini, G.; Angelini, A.; Melacini, P.; Vitiello, L.; Vazza, G.; Thiene, G.; et al. Novel Missense Variant in MYL2 Gene Associated with Hypertrophic Cardiomyopathy Showing High Incidence of Restrictive Physiology. Circ. Genom. Precis. Med. 2020, 13, e002824. [Google Scholar] [CrossRef]
  221. Kubo, T.; Gimeno, J.R.; Bahl, A.; Steffensen, U.; Steffensen, M.; Osman, E.; Thaman, R.; Mogensen, J.; Elliott, P.M.; Doi, Y.; et al. Prevalence, clinical significance, and genetic basis of hypertrophic cardiomyopathy with restrictive phenotype. J. Am. Coll. Cardiol. 2007, 49, 2419–2426. [Google Scholar] [CrossRef]
  222. Biagini, E.; Spirito, P.; Rocchi, G.; Ferlito, M.; Rosmini, S.; Lai, F.; Lorenzini, M.; Terzi, F.; Bacchi-Reggiani, L.; Boriani, G.; et al. Prognostic Implications of the Doppler Restrictive Filling Pattern in Hypertrophic Cardiomyopathy. Am. J. Cardiol. 2009, 104, 1727–1731. [Google Scholar] [CrossRef] [PubMed]
  223. Coppini, R.; Ho, C.Y.; Ashley, E.; Day, S.; Ferrantini, C.; Girolami, F.; Tomberli, B.; Bardi, S.; Torricelli, F.; Cecchi, F.; et al. Clinical Phenotype and Outcome of Hypertrophic Cardiomyopathy Associated with Thin-Filament Gene Mutations. J. Am. Coll. Cardiol. 2014, 64, 2589–2600. [Google Scholar] [CrossRef] [PubMed]
  224. Honey, M. A Case of Fatal Peripartum Cardiomyopathy. Br. Heart J. 1986, 55, 114. [Google Scholar] [CrossRef]
  225. Ware, J.S.; Li, J.; Mazaika, E.; Yasso, C.M.; DeSouza, T.; Cappola, T.P.; Tsai, E.J.; Hilfiker-Kleiner, D.; Kamiya, C.A.; Mazzarotto, F.; et al. Shared Genetic Predisposition in Peripartum and Dilated Cardiomyopathies. N. Engl. J. Med. 2016, 374, 233–241. [Google Scholar] [CrossRef]
  226. Goli, R.; Li, J.; Brandimarto, J.; Levine, L.D.; Riis, V.; McAfee, Q.; DePalma, S.; Haghighi, A.; Seidman, J.G.; Seidman, C.E.; et al. Genetic and Phenotypic Landscape of Peripartum Cardiomyopathy. Circulation 2021, 143, 1852–1862. [Google Scholar] [CrossRef] [PubMed]
  227. Garcia-Pavia, P.; Kim, Y.; Restrepo-Cordoba, M.A.; Lunde, I.G.; Wakimoto, H.; Smith, A.M.; Toepfer, C.N.; Getz, K.; Gorham, J.; Patel, P.; et al. Genetic Variants Associated with Cancer Therapy-Induced Cardiomyopathy. Circulation 2019, 140, 31–41. [Google Scholar] [CrossRef] [PubMed]
  228. Ware, J.S.; Amor-Salamanca, A.; Tayal, U.; Govind, R.; Serrano, I.; Salazar-Mendiguchía, J.; García-Pinilla, J.M.; Pascual-Figal, D.A.; Nuñez, J.; Guzzo-Merello, G.; et al. Genetic Etiology for Alcohol-Induced Cardiac Toxicity. J. Am. Coll. Cardiol. 2018, 71, 2293–2302. [Google Scholar] [CrossRef]
  229. Khan, R.S.; Pahl, E.; Dellefave-Castillo, L.; Rychlik, K.; Ing, A.; Yap, K.L.; Brew, C.; Johnston, J.R.; McNally, E.M.; Webster, G. Genotype and Cardiac Outcomes in Pediatric Dilated Cardiomyopathy. J. Am. Heart Assoc. 2022, 11, e022854. [Google Scholar] [CrossRef]
  230. Heidecker, B.; Williams, S.H.; Jain, K.; Oleynik, A.; Patriki, D.; Kottwitz, J.; Berg, J.; Garcia, J.A.; Baltensperger, N.; Lovrinovic, M.; et al. Virome Sequencing in Patients with Myocarditis. Circ. Heart Fail. 2020, 13, e007103. [Google Scholar] [CrossRef]
  231. Artico, J.; Merlo, M.; Delcaro, G.; Cannatà, A.; Gentile, P.; De Angelis, G.; Paldino, A.; Bussani, R.; Ferro, M.D.; Sinagra, G. Lymphocytic Myocarditis: A Genetically Predisposed Disease? J. Am. Coll. Cardiol. 2020, 75, 3098–3100. [Google Scholar] [CrossRef] [PubMed]
  232. Kontorovich, A.R.; Patel, N.; Moscati, A.; Richter, F.; Peter, I.; Purevjav, E.; Selejan, S.R.; Kindermann, I.; Towbin, J.A.; Bohm, M.; et al. Myopathic Cardiac Genotypes Increase Risk for Myocarditis. JACC Basic Transl. Sci. 2021, 6, 584–592. [Google Scholar] [CrossRef]
  233. Seidel, F.; Holtgrewe, M.; Al-Wakeel-Marquard, N.; Opgen-Rhein, B.; Dartsch, J.; Herbst, C.; Beule, D.; Pickardt, T.; Klingel, K.; Messroghli, D.; et al. Pathogenic Variants Associated with Dilated Cardiomyopathy Predict Outcome in Pediatric Myocarditis. Circ. Genom. Precis. Med. 2021, 14, e003250. [Google Scholar] [CrossRef] [PubMed]
  234. Tiron, C.; Campuzano, O.; Fernández-Falgueras, A.; Alcalde, M.; Loma-Osorio, P.; Zamora, E.; Caballero, A.; Sarquella-Brugada, G.; Cesar, S.; Garcia-Cuenllas, L.; et al. Prevalence of Pathogenic Variants in Cardiomyopathy-Associated Genes in Myocarditis. Circ. Genom. Precis. Med. 2022, 15, e003408. [Google Scholar] [CrossRef]
  235. Lota, A.S.; Hazebroek, M.R.; Theotokis, P.; Wassall, R.; Salmi, S.; Halliday, B.P.; Tayal, U.; Verdonschot, J.; Meena, D.; Owen, R.; et al. Genetic Architecture of Acute Myocarditis and the Overlap with Inherited Cardiomyopathy. Circulation 2022, 146, 1123–1134. [Google Scholar] [CrossRef] [PubMed]
  236. Jansweijer, J.A.; van Spaendonck-Zwarts, K.Y.; Tanck, M.W.T.; van Tintelen, J.P.; Christiaans, I.; van der Smagt, J.; Vermeer, A.; Bos, J.M.; Moss, A.J.; Swan, H.; et al. Heritability in Genetic Heart Disease: The Role of Genetic Background. Open Heart 2019, 6, e000929. [Google Scholar] [CrossRef] [PubMed]
  237. Rao, R.A.; Kozaily, E.; Jawaid, O.; Sabra, M.; El-Am, E.A.; Chaaya, R.G.B.; Woiewodski, L.; Elsemesmani, H.; Ramchandani, J.; Shah, C.; et al. Comparison of and Frequency of Mortality, Left Ventricular Assist Device Implantation, Ventricular Arrhythmias, and Heart Transplantation in Patients with Familial Versus Nonfamilial Idiopathic Dilated Cardiomyopathy. Am. J. Cardiol. 2022, 179, 83–89. [Google Scholar] [CrossRef]
  238. Stroeks, S.; Hellebrekers, D.; Claes, G.R.F.; Tayal, U.; Krapels, I.P.C.; Vanhoutte, E.K.; van den Wijngaard, A.; Henkens, M.; Ware, J.S.; Heymans, S.R.B.; et al. Clinical impact of re-evaluating genes and variants implicated in dilated cardiomyopathy. Genet. Med. 2021, 23, 2186–2193. [Google Scholar] [CrossRef]
  239. Hazebroek, M.R.; Moors, S.; Dennert, R.; van den Wijngaard, A.; Krapels, I.; Hoos, M.; Verdonschot, J.; Merken, J.J.; de Vries, B.; Wolffs, P.F.; et al. Prognostic Relevance of Gene-Environment Interactions in Patients with Dilated Cardiomyopathy: Applying the MOGE(S) Classification. J. Am. Coll. Cardiol. 2015, 66, 1313–1323. [Google Scholar] [CrossRef]
  240. Jansweijer, J.A.; Nieuwhof, K.; Russo, F.; Hoorntje, E.T.; Jongbloed, J.D.; Lekanne Deprez, R.H.; Postma, A.V.; Bronk, M.; van Rijsingen, I.A.; de Haij, S.; et al. Truncating Titin Mutations Are Associated with a Mild and Treatable Form of Dilated Cardiomyopathy. Eur. J. Heart Fail. 2017, 19, 512–521. [Google Scholar] [CrossRef]
  241. Gigli, M.; Merlo, M.; Graw, S.L.; Barbati, G.; Rowland, T.J.; Slavov, D.B.; Stolfo, D.; Haywood, M.E.; Dal Ferro, M.; Altinier, A.; et al. Genetic Risk of Arrhythmic Phenotypes in Patients with Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2019, 74, 1480–1490. [Google Scholar] [CrossRef] [PubMed]
  242. Dal Ferro, M.; Stolfo, D.; Altinier, A.; Gigli, M.; Perrieri, M.; Ramani, F.; Barbati, G.; Pivetta, A.; Brun, F.; Monserrat, L.; et al. Association Between Mutation Status and Left Ventricular Reverse Remodelling in Dilated Cardiomyopathy. Heart 2017, 103, 1704–1710. [Google Scholar] [CrossRef] [PubMed]
  243. Pasotti, M.; Klersy, C.; Pilotto, A.; Marziliano, N.; Rapezzi, C.; Serio, A.; Mannarino, S.; Gambarin, F.; Favalli, V.; Grasso, M.; et al. Long-Term Outcome and Risk Stratification in Dilated Cardiolaminopathies. J. Am. Coll. Cardiol. 2008, 52, 1250–1260. [Google Scholar] [CrossRef] [PubMed]
  244. Cabrera-Romero, E.; Ochoa, J.P.; Barriales-Villa, R.; Bermúdez-Jiménez, F.J.; Climent-Payá, V.; Zorio, E.; Espinosa, M.A.; Gallego-Delgado, M.; Navarro-Peñalver, M.; Arana-Achaga, X.; et al. Penetrance of Dilated Cardiomyopathy in Genotype-Positive Relatives. J. Am. Coll. Cardiol. 2024, 83, 1640–1651. [Google Scholar] [CrossRef]
  245. Akhtar, M.M.; Lorenzini, M.; Pavlou, M.; Ochoa, J.P.; O’Mahony, C.; Restrepo-Cordoba, M.A.; Segura-Rodriguez, D.; Bermúdez-Jiménez, F.; Molina, P.; Cuenca, S.; et al. Association of Left Ventricular Systolic Dysfunction Among Carriers of Truncating Variants in Filamin C with Frequent Ventricular Arrhythmia and End-stage Heart Failure. JAMA Cardiol. 2021, 6, 891–901. [Google Scholar] [CrossRef]
  246. Gigli, M.; Stolfo, D.; Graw, S.L.; Merlo, M.; Gregorio, C.; Nee Chen, S.; Dal Ferro, M.; Paldino, M.A.; De Angelis, G.; Brun, F.; et al. Phenotypic Expression, Natural History, and Risk Stratification of Cardiomyopathy Caused by Filamin C Truncating Variants. Circulation 2021, 144, 1600–1611. [Google Scholar] [CrossRef]
  247. de Frutos, F.; Ochoa, J.P.; Navarro-Peñalver, M.; Baas, A.; Bjerre, J.V.; Zorio, E.; Méndez, I.; Lorca, R.; Verdonschot, J.A.J.; García-Granja, P.E.; et al. Natural History of MYH7-Related Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2022, 80, 1447–1461. [Google Scholar] [CrossRef]
  248. Jansen, M.; de Brouwer, R.; Hassanzada, F.; Schoemaker, A.E.; Schmidt, A.F.; Kooijman-Reumerman, M.D.; Bracun, V.; Slieker, M.G.; Dooijes, D.; Vermeer, A.M.C.; et al. Penetrance and Prognosis of MYH7 Variant-Associated Cardiomyopathies: Results From a Dutch Multicenter Cohort Study. JACC Heart Fail. 2024, 12, 134–147. [Google Scholar] [CrossRef]
  249. Dorn, G.W., 2nd; McNally, E.M. Two Strikes and You’re Out: Gene-Gene Mutation Interactions in HCM. Circ. Res. 2014, 115, 208–210. [Google Scholar] [CrossRef]
  250. Ijaz, T.; Burke, M.A. BET Protein-Mediated Transcriptional Regulation in Heart Failure. Int. J. Mol. Sci. 2021, 22, 6059. [Google Scholar] [CrossRef]
  251. Golbus, J.R.; Puckelwartz, M.J.; Fahrenbach, J.P.; Dellefave-Castillo, L.M.; Wolfgeher, D.; McNally, E.M. Population-Based Variation in Cardiomyopathy Genes. Circ. Cardiovasc. Genet. 2012, 5, 391–399. [Google Scholar] [CrossRef] [PubMed]
  252. Norton, N.; Robertson, P.D.; Rieder, M.J.; Züchner, S.; Rampersaud, E.; Martin, E.; Li, D.; Nickerson, D.A.; Hershberger, R.E. Evaluating Pathogenicity of Rare Variants From Dilated Cardiomyopathy in the Exome Era. Circ. Cardiovasc. Genet. 2012, 5, 167–174. [Google Scholar] [CrossRef] [PubMed]
  253. Shah, R.A.; Asatryan, B.; Sharaf Dabbagh, G.; Aung, N.; Khanji, M.Y.; Lopes, L.R.; van Duijvenboden, S.; Holmes, A.; Muser, D.; Landstrom, A.P.; et al. Frequency, Penetrance, and Variable Expressivity of Dilated Cardiomyopathy-Associated Putative Pathogenic Gene Variants in UK Biobank Participants. Circulation 2022, 146, 110–124. [Google Scholar] [CrossRef] [PubMed]
  254. Meder, B.; Rühle, F.; Weis, T.; Homuth, G.; Keller, A.; Franke, J.; Peil, B.; Lorenzo Bermejo, J.; Frese, K.; Huge, A.; et al. A Genome-Wide Association Study Identifies 6p21 as Novel Risk Locus for Dilated Cardiomyopathy. Eur. Heart J. 2014, 35, 1069–1077. [Google Scholar] [CrossRef] [PubMed]
  255. Esslinger, U.; Garnier, S.; Korniat, A.; Proust, C.; Kararigas, G.; Müller-Nurasyid, M.; Empana, J.P.; Morley, M.P.; Perret, C.; Stark, K.; et al. Exome-Wide Association Study Reveals Novel Susceptibility Genes to Sporadic Dilated Cardiomyopathy. PLoS ONE 2017, 12, e0172995. [Google Scholar] [CrossRef]
  256. Garnier, S.; Harakalova, M.; Weiss, S.; Mokry, M.; Regitz-Zagrosek, V.; Hengstenberg, C.; Cappola, T.P.; Isnard, R.; Arbustini, E.; Cook, S.A.; et al. Genome-Wide Association Analysis in Dilated Cardiomyopathy Reveals Two New Players in Systolic Heart Failure on Chromosomes 3p25.1 and 22q11.23. Eur. Heart J. 2021, 42, 2000–2011. [Google Scholar] [CrossRef]
  257. Wild, P.S.; Felix, J.F.; Schillert, A.; Teumer, A.; Chen, M.H.; Leening, M.J.G.; Völker, U.; Großmann, V.; Brody, J.A.; Irvin, M.R.; et al. Large-Scale Genome-Wide Analysis Identifies Genetic Variants Associated with Cardiac Structure and Function. J. Clin. Investig. 2017, 127, 1798–1812. [Google Scholar] [CrossRef]
  258. Pirruccello, J.P.; Bick, A.; Wang, M.; Chaffin, M.; Friedman, S.; Yao, J.; Guo, X.; Venkatesh, B.A.; Taylor, K.D.; Post, W.S.; et al. Analysis of Cardiac Magnetic Resonance Imaging in 36,000 Individuals Yields Genetic Insights into Dilated Cardiomyopathy. Nat. Commun. 2020, 11, 2254. [Google Scholar] [CrossRef]
  259. Wilde, A.A.M.; Semsarian, C.; Márquez, M.F.; Sepehri Shamloo, A.; Ackerman, M.J.; Ashley, E.A.; Sternick, E.B.; Barajas-Martinez, H.; Behr, E.R.; Bezzina, C.R.; et al. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the State of Genetic Testing for Cardiac Diseases. Heart Rhythm. 2022, 19, e1–e60. [Google Scholar] [CrossRef]
  260. Waddell-Smith, K.E.; Donoghue, T.; Oates, S.; Graham, A.; Crawford, J.; Stiles, M.K.; Aitken, A.; Skinner, J.R. Inpatient Detection of Cardiac-Inherited Disease: The Impact of Improving Family History Taking. Open Heart 2016, 3, e000329. [Google Scholar] [CrossRef]
  261. Morales, A.; Cowan, J.; Dagua, J.; Hershberger, R.E. Family History: An Essential Tool for Cardiovascular Genetic Medicine. Congest. Heart Fail. 2008, 14, 37–45. [Google Scholar] [CrossRef] [PubMed]
  262. Dellefave-Castillo, L.M.; Cirino, A.L.; Callis, T.E.; Esplin, E.D.; Garcia, J.; Hatchell, K.E.; Johnson, B.; Morales, A.; Regalado, E.; Rojahn, S.; et al. Assessment of the Diagnostic Yield of Combined Cardiomyopathy and Arrhythmia Genetic Testing. JAMA Cardiol. 2022, 7, 966–974. [Google Scholar] [CrossRef] [PubMed]
  263. Catchpool, M.; Ramchand, J.; Martyn, M.; Hare, D.L.; James, P.A.; Trainer, A.H.; Knight, J.; Goranitis, I. A Cost-Effectiveness Model of Genetic Testing and Periodical Clinical Screening for the Evaluation of Families with Dilated Cardiomyopathy. Genet. Med. 2019, 21, 2815–2822. [Google Scholar] [CrossRef] [PubMed]
  264. Stroeks, S.; Verdonschot, J.A.J. The Next Step Toward Personalized Recommendations for Genetic Cardiomyopathies. Eur. J. Hum. Genet. 2023, 31, 1201–1203. [Google Scholar] [CrossRef]
  265. Zhang, X.; Walsh, R.; Whiffin, N.; Buchan, R.; Midwinter, W.; Wilk, A.; Govind, R.; Li, N.; Ahmad, M.; Mazzarotto, F.; et al. Disease-Specific Variant Pathogenicity Prediction Significantly Improves Variant Interpretation in Inherited Cardiac Conditions. Genet. Med. 2021, 23, 69–79. [Google Scholar] [CrossRef]
  266. Braunwald, E.; Lambrew, C.T.; Rockoff, S.D.; Ross, J., Jr.; Morrow, A.G. Idiopathic Hypertrophic Subaortic Stenosis. I. A Description of the Disease Based Upon an Analysis of 64 Pateints. Circulation 1964, 30 (Suppl. 4), 3–119. [Google Scholar] [CrossRef]
  267. Grodzicker, T.; Williams, J.; Sharp, P.; Sambrook, J. Physical Mapping of Temperature-Sensitive Mutations of Adenoviruses. Cold Spring Harb. Symp. Quant. Biol. 1974, 39, 439–446. [Google Scholar] [CrossRef]
  268. Sanger, F.; Nicklen, S.; Coulson, A.R. DNA Sequencing with Chain-Terminating Inhibitors. Proc. Natl. Acad. Sci. USA 1977, 74, 5463–5467. [Google Scholar] [CrossRef]
  269. Gusella, J.F.; Wexler, N.S.; Conneally, P.M.; Naylor, S.L.; Anderson, M.A.; Tanzi, R.E.; Watkins, P.C.; Ottina, K.; Wallace, M.R.; Sakaguchi, A.Y.; et al. A Polymorphic DNA Marker Genetically Linked to Huntington’s Disease. Nature 1983, 306, 234–238. [Google Scholar] [CrossRef]
  270. Donis-Keller, H.; Green, P.; Helms, C.; Cartinhour, S.; Weiffenbach, B.; Stephens, K.; Keith, T.P.; Bowden, D.W.; Smith, D.R.; Lander, E.S.; et al. A Genetic Linkage Map of the Human Genome. Cell 1987, 51, 319–337. [Google Scholar] [CrossRef]
  271. Watson, J.D. The Human Genome Project: Past, Present, and Future. Science 1990, 248, 44–49. [Google Scholar] [CrossRef] [PubMed]
  272. Collins, F.S.; Green, E.D.; Guttmacher, A.E.; Guyer, M.S. A Vision for the Future of Genomics Research. Nature 2003, 422, 835–847. [Google Scholar] [CrossRef] [PubMed]
  273. Margulies, M.; Egholm, M.; Altman, W.E.; Attiya, S.; Bader, J.S.; Bemben, L.A.; Berka, J.; Braverman, M.S.; Chen, Y.J.; Chen, Z.; et al. Genome Sequencing in Microfabricated High-Density Picolitre Reactors. Nature 2005, 437, 376–380. [Google Scholar] [CrossRef] [PubMed]
  274. Fu, W.; O’Connor, T.D.; Jun, G.; Kang, H.M.; Abecasis, G.; Leal, S.M.; Gabriel, S.; Rieder, M.J.; Altshuler, D.; Shendure, J.; et al. Analysis of 6,515 Exomes Reveals the Recent Origin of Most Human Protein-Coding Variants. Nature 2013, 493, 216–220. [Google Scholar] [CrossRef]
  275. Auton, A.; Brooks, L.D.; Durbin, R.M.; Garrison, E.P.; Kang, H.M.; Korbel, J.O.; Marchini, J.L.; McCarthy, S.; McVean, G.A.; Abecasis, G.R. A Global Reference for Human Genetic Variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef]
  276. Lek, M.; Karczewski, K.J.; Minikel, E.V.; Samocha, K.E.; Banks, E.; Fennell, T.; O’Donnell-Luria, A.H.; Ware, J.S.; Hill, A.J.; Cummings, B.B.; et al. Analysis of Protein-Coding Genetic Variation in 60,706 Humans. Nature 2016, 536, 285–291. [Google Scholar] [CrossRef]
  277. Karczewski, K.J.; Francioli, L.C.; Tiao, G.; Cummings, B.B.; Alföldi, J.; Wang, Q.; Collins, R.L.; Laricchia, K.M.; Ganna, A.; Birnbaum, D.P.; et al. The Mutational Constraint Spectrum Quantified From Variation in 141,456 Humans. Nature 2020, 581, 434–443. [Google Scholar] [CrossRef]
  278. Taliun, D.; Harris, D.N.; Kessler, M.D.; Carlson, J.; Szpiech, Z.A.; Torres, R.; Taliun, S.A.G.; Corvelo, A.; Gogarten, S.M.; Kang, H.M.; et al. Sequencing of 53,831 Diverse Genomes From the NHLBI TOPMed Program. Nature 2021, 590, 290–299. [Google Scholar] [CrossRef]
  279. Harper, A.R.; Goel, A.; Grace, C.; Thomson, K.L.; Petersen, S.E.; Xu, X.; Waring, A.; Ormondroyd, E.; Kramer, C.M.; Ho, C.Y.; et al. Common Genetic Variants and Modifiable Risk Factors Underpin Hypertrophic Cardiomyopathy Susceptibility and Expressivity. Nat. Genet. 2021, 53, 135–142. [Google Scholar] [CrossRef]
  280. Martin, A.R.; Kanai, M.; Kamatani, Y.; Okada, Y.; Neale, B.M.; Daly, M.J. Clinical Use of Current Polygenic Risk Scores May Exacerbate Health Disparities. Nat. Genet. 2019, 51, 584–591. [Google Scholar] [CrossRef]
  281. Garcia-Pavia, P.; Grogan, M.; Kale, P.; Berk, J.L.; Maurer, M.S.; Conceição, I.; Di Carli, M.; Solomon, S.D.; Chen, C.; Yureneva, E.; et al. Impact of Vutrisiran on Exploratory Cardiac Parameters in Hereditary Transthyretin-Mediated Amyloidosis with Polyneuropathy. Eur. J. Heart Fail. 2024, 26, 397–410. [Google Scholar] [CrossRef] [PubMed]
  282. Nishiyama, T.; Zhang, Y.; Cui, M.; Li, H.; Sanchez-Ortiz, E.; McAnally, J.R.; Tan, W.; Kim, J.; Chen, K.; Xu, L.; et al. Precise Genomic Editing of Pathogenic Mutations in RBM20 Rescues Dilated Cardiomyopathy. Sci. Transl. Med. 2022, 14, eade1633. [Google Scholar] [CrossRef] [PubMed]
  283. Xu, L.; Lau, Y.S.; Gao, Y.; Li, H.; Han, R. Life-Long AAV-Mediated CRISPR Genome Editing in Dystrophic Heart Improves Cardiomyopathy without Causing Serious Lesions in mdx Mice. Mol. Ther. 2019, 27, 1407–1414. [Google Scholar] [CrossRef] [PubMed]
  284. Haroldson, J.; Harrison, W.; Lombardi, L.; Argast, G.; Duclos, Z.; Nelson, S.; Sethi, S.; Tomlinson, L.; Paterson, N.; Pollman, M.; et al. MyPeak-1: A Phase 1b Study to Evaluate Safety and Efficacy of TN-201, an Adeno-Associated Virus Serotype 9 (AAV9) Investigational Gene Therapy, in Adults with MYBPC3-Associated Hypertrophic Cardiomyopathy (HCM). J. Card. Fail. 2024, 30, S5. [Google Scholar] [CrossRef]
  285. Grosch, M.; Schraft, L.; Chan, A.; Küchenhoff, L.; Rapti, K.; Ferreira, A.M.; Kornienko, J.; Li, S.; Radke, M.H.; Krämer, C.; et al. Striated Muscle-Specific Base Editing Enables Correction of Mutations Causing Dilated Cardiomyopathy. Nat. Commun. 2023, 14, 3714. [Google Scholar] [CrossRef]
Figure 1. Visual illustration of the breadth of gene ontologies in dilated cardiomyopathy. Summarized are genes involved in (A) cell junctions, (B) the sarcomere, (C) mitochondria, (D) nuclear architecture and protein trafficking, and (E) cytoskeletal architecture (bold font—definitively pathogenic genes; standard font—putatively pathogenic genes).
Figure 1. Visual illustration of the breadth of gene ontologies in dilated cardiomyopathy. Summarized are genes involved in (A) cell junctions, (B) the sarcomere, (C) mitochondria, (D) nuclear architecture and protein trafficking, and (E) cytoskeletal architecture (bold font—definitively pathogenic genes; standard font—putatively pathogenic genes).
Ijms 25 11460 g001
Figure 2. Summary of the genetic underpinnings of the various cardiomyopathy syndromes. Genes that produce channelopathies, ACM, LVNC, and mitochondrial disease can also produce a DCM phenotype. Similarly, genes implicated in RCM and mitochondrial disease can also produce an HCM phenotype. ACM arrhythmogenic cardiomyopathy, ARVC arrhythmogenic right ventricular cardiomyopathy, ALVC arrhythmogenic left ventricular cardiomyopathy, BiV biventricular, DCM dilated cardiomyopathy, HCM hypertrophic cardiomyopathy, LVNC left ventricular non-compaction, and RCM restrictive cardiomyopathy.
Figure 2. Summary of the genetic underpinnings of the various cardiomyopathy syndromes. Genes that produce channelopathies, ACM, LVNC, and mitochondrial disease can also produce a DCM phenotype. Similarly, genes implicated in RCM and mitochondrial disease can also produce an HCM phenotype. ACM arrhythmogenic cardiomyopathy, ARVC arrhythmogenic right ventricular cardiomyopathy, ALVC arrhythmogenic left ventricular cardiomyopathy, BiV biventricular, DCM dilated cardiomyopathy, HCM hypertrophic cardiomyopathy, LVNC left ventricular non-compaction, and RCM restrictive cardiomyopathy.
Ijms 25 11460 g002
Figure 3. The relationship between variant effect size and its frequency in a population. Effect size refers to the magnitude of influence the variant has on a phenotype, whereas allele frequency describes the prevalence of the variant in a population. DCM is an example of a complex genetic disorder with a causal genetic basis ranging from monogenic, whereby very rare variants in specific genes have a marked effect on cardiac phenotype, to polygenic, where more prevalent minor variants, which individually have little effect on phenotype, can in aggregate result in the disease.
Figure 3. The relationship between variant effect size and its frequency in a population. Effect size refers to the magnitude of influence the variant has on a phenotype, whereas allele frequency describes the prevalence of the variant in a population. DCM is an example of a complex genetic disorder with a causal genetic basis ranging from monogenic, whereby very rare variants in specific genes have a marked effect on cardiac phenotype, to polygenic, where more prevalent minor variants, which individually have little effect on phenotype, can in aggregate result in the disease.
Ijms 25 11460 g003
Figure 4. Summary of major guidelines recommendations to genetics screening in DCM. (Left panel) demonstrates steps provider should take for proband (index) patient. (Right panel) demonstrates algorithmic approach to first-degree relatives [4,34,259].
Figure 4. Summary of major guidelines recommendations to genetics screening in DCM. (Left panel) demonstrates steps provider should take for proband (index) patient. (Right panel) demonstrates algorithmic approach to first-degree relatives [4,34,259].
Ijms 25 11460 g004
Figure 5. A 60-year history of advances in cardiomyopathy genetics. Our understanding of the genetic basis of the cardiomyopathies has paralleled major advances in genetics research tools. HCM hypertrophic cardiomyopathy; RFLP restriction fragment length polymorphism; MD muscular dystrophy; DCM dilated cardiomyopathy; ARVC arrhythmogenic right ventricular cardiomyopathy; NGS next-generation sequencing; GWAS genome-wide association study; ESP the NHLBI Exome Sequencing Project; ExAC exome aggregation consortium; gnomAD genome aggregation database consortium; TOPmed the NHLBI Trans-omics for precision medicine study. References in the figure include [2,3,36,43,53,54,56,81,95,105,133,266,267,268,269,270,271,272,273,274,275,276,277,278,279].
Figure 5. A 60-year history of advances in cardiomyopathy genetics. Our understanding of the genetic basis of the cardiomyopathies has paralleled major advances in genetics research tools. HCM hypertrophic cardiomyopathy; RFLP restriction fragment length polymorphism; MD muscular dystrophy; DCM dilated cardiomyopathy; ARVC arrhythmogenic right ventricular cardiomyopathy; NGS next-generation sequencing; GWAS genome-wide association study; ESP the NHLBI Exome Sequencing Project; ExAC exome aggregation consortium; gnomAD genome aggregation database consortium; TOPmed the NHLBI Trans-omics for precision medicine study. References in the figure include [2,3,36,43,53,54,56,81,95,105,133,266,267,268,269,270,271,272,273,274,275,276,277,278,279].
Ijms 25 11460 g005
Table 1. Challenges and solutions to identifying familial disease.
Table 1. Challenges and solutions to identifying familial disease.
Challenge Solution
Inadequate family history-Take at least a three-generation family history (i.e., current, prior, and subsequent generations)
-Clarify potentially vague cardiovascular events (e.g., heart attack)
-Obtain family medical records for review
Incomplete penetrance and variable expressivity-Evaluation by a cardiologist
-Genetic testing of P/LP genes
-Echocardiogram
-ECG
De novo variant-Cascade genetic testing in offspring
P/LP pathogenic/likely pathogenic, ECG electrocardiogram.
Table 2. Gene ontologies in dilated cardiomyopathy.
Table 2. Gene ontologies in dilated cardiomyopathy.
Gene OMIM Protein Associated Phenotype(s) Other than DCM
Sarcomere
ACTC1 [36]102540Cardiac actin HCM
MYH7 [37,38]160760β-myosin heavy chainHCM [3,39] and LVNC [40,41]
TNNT2 [37]191045Troponin-THCM [42,43]
TNNI3 [44,45]191044Troponin-I HCM [46]
TNNC1 [44]191040Troponin-C HCM (putative) [47,48,49]
TPM1 [50]191010ɑ-TropomyosinHCM [42,43]
MYBPC3 [51,52]600958Myosin binding protein CHCM [53,54]
TTN [55,56]188840TitinACM [57], Tibial Muscular Dystrophy [58], LGMD2J [59], Hereditary Myopathy with Early Respiratory Failure [60], and Salih Myopathy [61]
MYL2 [62]160781Myosin light chain 2HCM [63]
MYPN [64,65]608517MyopalladinHCM (putative) [64]
Nuclear and cytoskeletal architecture
LMNA [66,67]150330 Lamin A/C ACM [68], EDMD type 2 [69], LGMD1B [70], and Congenital Muscular Dystrophy [71]
LEM2616312LEM domain-containing protein 2 (LEMD2)ACM [72,73]
FLNC [74]102565Filamin CACM [74,75], HCM (putative) [76,77], and MFM [78]
DMD [79,80] 300377DystrophinDuchenne muscular dystrophy and Becker muscular dystrophy [81]
EMD [82]300384EmerinEDMD type 1 [83]
DES [84,85,86,87]125660Desmin LVNC [88,89], RCM [90], ACM and MFM [84,86]
Mitochondrial
TAZ [91]300394TaffazinBarth syndrome [91]
DNAJC19 [92,93]608977DnaJ heat shock protein family (Hsp40) member C19 Dilated cardiomyopathy with ataxia syndrome [92,93]
Protein trafficking
BAG3 [94,95]603883Bcl2-associated athanogene 3 ACM, MFM
Gene expression
RBM20 [96]613171Ribonucleic acid binding protein 20 ACM [97]
TBX20 [98]606061T-box protein 20 Congenital heart defects [98], LVNC [99]
Desmosomal proteins
JUP [100]173325PlakoglobinARVC/ACM and Naxos syndrome [101,102,103]
DSP [104]125647DesmoplakinARVC/ACM [105] and Carvajal syndrome [104,106]
PKP2 [107]602861PlakophilinARVC/ACM [108,109,110]
DSG2 [107]125671Desmoglein 2ARVC/ACM [111,112,113]
DSC2 [100,107]125645Desmocollin 2ARVC/ACM [114,115,116]
Membrane proteins
TMEM43 [117]612048Transmembrane protein 43 ARVC/ACM [117,118,119]
ILK602366Integrin-linked kinaseARVC/ACM [120]
Sarcoplasmic reticulum
PLN [121,122,123]172405Phospholamban
Channels
SCN5A [124,125,126]600163Voltage-gated sodium channel α-subunit ACM, long-QT syndrome (type 3), Brugada syndrome, conduction delay, ectopic Purkinje foci, sinus node dysfunction [124], and atrial fibrillation [125]
HCM hypertrophic cardiomyopathy, LVNC left ventricular non-compaction, ACM arrhythmogenic cardiomyopathy, ARVC arrhythmogenic right ventricular cardiomyopathy, LGMD limb-girdle muscular dystrophy, EDMD Emery-Dreifuss muscular dystrophy, MFM myofibrillar myopathy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Newman, N.A.; Burke, M.A. Dilated Cardiomyopathy: A Genetic Journey from Past to Future. Int. J. Mol. Sci. 2024, 25, 11460. https://doi.org/10.3390/ijms252111460

AMA Style

Newman NA, Burke MA. Dilated Cardiomyopathy: A Genetic Journey from Past to Future. International Journal of Molecular Sciences. 2024; 25(21):11460. https://doi.org/10.3390/ijms252111460

Chicago/Turabian Style

Newman, Noah A., and Michael A. Burke. 2024. "Dilated Cardiomyopathy: A Genetic Journey from Past to Future" International Journal of Molecular Sciences 25, no. 21: 11460. https://doi.org/10.3390/ijms252111460

APA Style

Newman, N. A., & Burke, M. A. (2024). Dilated Cardiomyopathy: A Genetic Journey from Past to Future. International Journal of Molecular Sciences, 25(21), 11460. https://doi.org/10.3390/ijms252111460

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop