Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Cardiol. Aug 26, 2025; 17(8): 107847
Published online Aug 26, 2025. doi: 10.4330/wjc.v17.i8.107847
Relationship between genotype and clinical phenotype of hypertrophic cardiomyopathy
Lan-Lan Zhang, Bo Wang, Jing Wang, Jia Zhao, Li-Wen Liu, Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Fourth Military Medical University, Xi’an 710000, Shaanxi Province, China
ORCID number: Lan-Lan Zhang (0009-0003-7360-2037); Li-Wen Liu (0000-0001-9838-5727).
Co-first authors: Lan-Lan Zhang and Bo Wang.
Author contributions: Zhang LL and Wang B sorted out the published literature and wrote the manuscript, they contributed equally to this article, they are the co-first authors of this manuscript; Zhang LL, Wang B, Wang J, Zhao J, and Liu LW provided critical input, critically reviewed the manuscript, and contributed to discussion; Liu LW conceived the project; and all authors have read and approved the final manuscript.
Supported by National Natural Science Foundation of China, No. 82230065, No. 82371974 and No. 82272009.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Li-Wen Liu, Chief Physician, Full Professor, Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Fourth Military Medical University, No. 127 Changle West Road, Xi’an 710000, Shaanxi Province, China. liuliwen@fmmu.edu.cn
Received: April 2, 2025
Revised: May 12, 2025
Accepted: August 12, 2025
Published online: August 26, 2025
Processing time: 144 Days and 23.7 Hours

Abstract

Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiovascular disease, mostly inherited in an autosomal dominant manner. It is a global heart disease with complex clinical phenotypes and gene expression. The prevalence rate in the population is 1:500-1:200. This article mainly introduces the diagnostic criteria, pathological manifestations, and genetic basis of HCM, which is the leading cause of sudden death in adolescents and athletes due to exercise, with 60%-70% showing familial clustering. It also discusses the latest progress in the relationship between different genotypes and clinical phenotypes of HCM patients.

Key Words: Hypertrophic cardiomyopathy; Genotype; Phenotype; Sarcomere; Thick myofilament; Thin myofilament

Core Tip: The presence of sarcomere mutations makes the prognosis of hypertrophic cardiomyopathy (HCM) patients worse. Compared with the two most common thick myofilament genes, MYH7 carriers have an earlier onset of HCM. Compared with patients with thick myofilament gene mutations, patients with thin myofilament mutations have a worse prognosis. Among patients with thin myofilament gene mutations, TNNC1 positive probands have the worst prognosis. In this paper, the close relationship between HCM hypertrophy pattern and genetic variation was combed and insights were put forward, which provided a theoretical basis for genetic testing and personalized management of HCM patients.



INTRODUCTION
Diagnostic criteria

Hypertrophic cardiomyopathy (HCM) is a heart disease characterized by myocardial hypertrophy, mainly manifested as thickening of the left ventricular (LV) wall. According to the 2023 “Chinese Guidelines for Diagnosis and Treatment of Adult HCM”[1] and the 2024 “AHA/ACC/AMSSM/HRS/PACES/SCMR Guideline for the Management of HCM”[2], the clinical diagnosis of adult HCM is confirmed through imaging tests (echocardiography, cardiac magnetic resonance (CMR) imaging, computed tomography). Any of the above imaging tests shows a maximum end diastolic wall thickness of ≥ 15 mm in any part of the left ventricle. After excluding other causes of hypertrophy (such as increased afterload induced cardiac hypertrophy and athlete’s cardiac hypertrophy), it can be diagnosed as HCM; genetic test shows carrying a pathogenic sarcomere gene mutation (Sarc) or having a family history of HCM, the diagnosis can be made with LV wall thickness ≥ 13 mm. The diagnostic criteria for children use a body surface area adjusted Z-score that is twice the standard deviation of the mean. The guidelines recommend that the diagnosis of HCM in children should consider the screening situation and the probability of disease prediction: A threshold of Z > 2.5 may be suitable for early identification of HCM in asymptomatic children without a family history, while for children with a clear family history or pathogenic Sarcs, a threshold of Z > 2 may be sufficient for early diagnosis[3].

Clinical and pathological findings

HCM is a heterogeneous disease that encompasses a range of disease characteristics (inner phenotypes) limited to the heart, which converge to produce clinically recognized disease phenotypes[4]. The phenotype of HCM is characterized by LV hypertrophy, excessive myocardial contraction, decreased compliance, myofibrillar disorder, and fibrosis. Patients often experience decreased mobility, exertional dyspnea, and/or chest pain symptoms due to impaired LV diastolic function and reduced compliance, LV outflow tract (LVOT) obstruction and mitral regurgitation, as well as microvascular dysfunction and subendocardial ischemia. In addition to increased interventricular septal thickness, abnormalities in the mitral and sub-valvular structures can also lead to systolic anterior displacement and LVOT obstruction, which are characteristics of obstructive HCM[5].

GENETIC BASIS

Molecular genetic studies have confirmed that 40%-60%[6] are mutations in genes encoding sarcomere structural proteins. Twenty-seven pathogenic genes have been found to be associated with HCM. These genes encode thick myofilament, thin myofilament, Z-disk structural proteins or calcium regulation related proteins. The remaining 50% of HCM do not find a clear pathogenic gene mutation, the cause of which is unknown. In some previous HCM diagnostic guidelines[6,7], it is believed that congenital metabolic diseases, neuromuscular diseases, mitochondrial diseases, malformation syndrome, systemic amyloidosis, etc.[7]. These rare clinical diseases are also classified as HCM, but the latest guidelines[1,2] exclude these diseases from HCM because of their clear etiology, prognosis and treatment are quite different from HCM.

Sarc

HCM, as a Mendelian autosomal dominant genetic disorder, has variable penetrance and is associated with genetic variations in the encoding of myocardial sarcomere proteins involved in contractile function[8]. The first gene related to HCM (MYH7) was identified in 1989[9], and its gene library expanded to include seven additional sarcomere genes (MYBPC3,TNNT2, TPM1, MYL2, MYL3, TNNI3, ACTC1) in the 1990s[10]. In patients with HCM and pathogenic sarcomere gene variants, the two most common genes are β - MYH7 and MYBPC3, which are found in 70% of variant positive patients, while other genes (TNNI3, TNNT2, TPM1, MYL2, MYL3, ACTC1) each account for a small proportion of patients (1% to 5%). Among these genes, over 2000 variants have been identified[11], most of which are “proprietary” (specific to individual families). Each descendant of affected family members has a 50% chance of inheriting this variant[12].

Established causal genes for HCM and their population frequencies are listed in Figure 1[12]. A schematic structure of a sarcomere composed of thick and thin filaments and Z discs is depicted along with its protein constituents involved in HCM. Lorenzini et al[13] conducted a retrospective analysis of carriers of sarcomere protein gene mutations identified during home screening. The estimated HCM penetrance rate at 15 years follow-up was 46%, and the frequency of the pathogenic gene was as follows: MYBPC3 (43.2%), MYH7 (24.2%), TNNI3 (13.7%), TNNT2 (11.9%), TPM1 (3.2%), MYL2 (2.1%), ACTC1 (0.4%), MYL3 (0%). In addition, 4 individuals (1.4%) carried multiple pathogenicity/Likely pathogenicity variants (3 MYBPC3 and TNNI3, and 1 MYH7 and TNNT2). According to the family line, the frequency of pathogenic genes is: MYBPC3 (45.5%), MYH7 (28.2%), TNNI3 (10.9%), TNNT2 (9.6%), TPM1 (1.9%), MYL2 (1.9%), ACTC1 (0.6%), MYL3 (0%); two families (1.3%) carry multiple pathogenicity/Likely pathogenicity variants. In a multivariate model adjusted for age and stratified by CMR, the independent predictors of HCM development were male gender and electrocardiographic abnormalities, with the TNNI3 variant having the lowest risk.

Figure 1
Figure 1 Hypertrophic cardiomyopathy as a disease of sarcomere proteins[12]. Citation: Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 2017; 121: 749-770. Copyright© 2017 American Heart Association, Inc.. Published by Wolters Kluwer Health, Inc. ( https://www.ahajournals.org/open-access-information).
Non-Sarc

In the Human Gene Mutation Database, professional (version 2015.3), there are 51 gene variants associated with HCM, including 8 fully validated sarcomere genes, 31 non-sarcomere genes, and 12 genes associated with complex phenotypes including left ventricular hypertrophy. The 8 core sarcomere genes (MYH7, MYBPC3, TNNT2, TPM1, MYL2, MYL3, TNNI3, and ACTC1) were validated completely because a detailed familial linkage study identified them as HCM genes. In addition, variants in 5 genes (ACTA1, COX15, MRPL3, MYO6 and SLC25A5) have been reported in other rare diseases, and most of these variants show a recessive mode of inheritance. There is evidence[14] to suggest that 11 genes play a major pathogenic role in HCM: Strong evidence (PLN, CSRP3[15], FHL1[16]), moderate evidence (CRYAB, ACTN2[17,18], FLNC[19], MYOZ2[20]), or weak evidence (TNNC1, MYH6, TRIM55[21], TRIM63[21]). In recent years, other non-sarcomere genes have also been found to be associated with the pathogenesis of HCM, such as the sodium channel gene SCN5A[22], which was confirmed to play an important role in the pathogenesis of HCM. In addition, our center found that the calcium channel gene CACNA1C has a gene accumulation effect with MYH7 gene in a large family with HCM[23,24].

Genetic screening

Echocardiography and CMR have a synergistic effect on the diagnosis of proband and family screening. Genetic testing can identify affected individuals without HCM phenotype, and its important role is to perform next-generation (cascade) testing on high-risk and usually asymptomatic family members. When the proband’s pathological sarcomere mutation test is positive, relatives who test negative for the same (or other) sarcomere mutation and are considered unaffected may be released from lifelong imaging examinations and clinical monitoring. In addition, the targeted gene testing team can identify non sarcomere cardiac, metabolic, or systemic diseases clinically similar to HCM that are associated with LV hypertrophy, such as lysosomal storage diseases like lysosome-associated membrane protein 2 (Danon) cardiomyopathy, (protein kinase, AMP-activated, non-catalytic, gamma-2) cardiac syndrome, Fabry disease, Noonan syndrome, and other RASopathy, as well as transthyretin cardiac amyloidosis. Due to the significant differences in natural course and treatment strategies between these phenotypic copies and sarcomere HCM, differential diagnosis defines the key role of genetic testing in such patients.

HCM is mainly a sarcomere disease, therefore, first-line gene testing mainly includes the detection of pathogenic HCM genes[25]: MYH7, MYBPC3, TNNI3, TNNT2, TPM1, MYL2, MYL3, and ACTC1. Pathogenic variants are typically identified in approximately 30% of sporadic cases and 60% of familial cases[26]. The identification of uncertain significant variations is not a clinically actionable outcome, but further research can be conducted at the clinical or research level to elucidate the pathogenicity of the variation (for example, through co segregation analysis of family members, DNA testing of parents to determine whether uncertain significant variations is a functional study from scratch). Genetic testing can be performed using various technical platforms, including gene panels, whole exome sequencing, or whole genome sequencing[27]. Generally, after the detection of eight first-line gene panels in the clinic, the whole exome gene detection or even the whole genome detection can be selected to find the possible new disease causing gene after the relevant disease causing gene is not found. However, the whole exome and whole genome detection have certain difficulties in the subsequent interpretation of gene data, and they can be applied to the interpretation of clinical data only after the large family and subsequent functional validation.

Recent studies have emphasized that specific genetic variations associated with the development of cardiomyopathy, particularly HCM, can guide rational drug design and the development of novel drug therapies[28]. For example, long-term administration of the small molecule MYK-461 has been shown to slow down the development of HCM and decrease pro-fibrotic gene expression in mice with myosin heavy chain heterozygous variants[29]. In addition, determining the diagnostic and prognostic impact of known pathological variations on sarcomere genes has laid the foundation for potential gene therapy. For example, clustered regularly interspaced short palindromic repeats - clustered regularly interspaced short palindromic repeats-associated protein 9 targets the GAGT deletion in MYBPC3 in human embryos and expresses functional cMYPPC in MYBPC3 gene knockout mice using adeno-associated virus therapy[30,31], to achieve the effect gene therapy. Overall, we found that specific genetic variations are associated with specific prognostic outcomes, which reinforces the role of genetic testing in personalized care for cardiomyopathy. In addition, to investigate the impact of common genetic variations on the risk of HCM, Harper et al[32] conducted two independent, unrelated, multi lineage case-control genome-wide association studies recruiting HCM patients from the HCM registry (2541 unselected cases compared to 40283 United Kingdom Biobank controls) and a comparative disease of rare biological resources (239 sarcomere negative cases compared to 7203 controls). Multi gene risk distribution, combined with pathogenic risk factors, drives individual susceptibility, and the management of sarcomere negativity is also an important aspect.

Genetic testing in HCM has several clinical benefits, including confirming diagnosis, preclinical diagnosis, cascading genetic testing within families, and guiding reproductive decision-making. The cascade gene testing in this family can identify individuals who carry pathogenic variants and require continuous monitoring, while those who do not carry the variant can be freed from lifelong clinical monitoring. After genetic testing, clinically actionable results (i.e., possible pathogenicity or pathogenicity) can provide diagnostic clarification for the proband and offer the possibility of cascading (predictive) testing for high-risk family members[12,33,34]. The most important is that the current assisted reproductive technology can screen embryos that do not carry disease causing genes in vitro and implant them into the mother, fundamentally reducing the number of patients with HCM and achieving true eugenics.

CLINICAL PHENOTYPE DIFFERENCES AMONG DIFFERENT GENOTYPES

The clinical first-line genes of HCM include mutations in genes encoding MYH7 and MYBPC3, accounting for about 80% of the positive cases of HCM by genetic testing; Next, TNNI3 and TNNT2 accounted for about 15% of HCM patients with positive genetic testing, and finally TPM1, ACTC1, MYL2 and MYL3 were relatively rare.

Morphological differences

The baseline data roughly divides patients into two categories. One group is sarcomere mutation positive, more likely to exhibit ventricular septal curvature with more fibrosis but less resting obstruction, while the other group is sarcomere mutation negative, more likely to exhibit isolated basal septal hypertrophy with obstruction but less fibrosis[35].

Compared with patients with sarcomere mutation negative HCM, those with sarcomere mutation positive HCM have a higher decrease in global longitudinal strain and a higher degree of LV myocardial fibrosis, indicating that sarcomere mutations may lead to LV diastolic dysfunction, thereby causing early progression of late stage heart failure[36]. Research has confirmed that pathogenic or potentially pathogenic sarcomere mutations are significantly associated with a large number of LV replacement fibrosis and extracellular volume fraction, especially in the interventricular septum. Similarly, Wang et al[37] found that late gadolinium enhancement ≥ 15% is three times associated with sudden cardiac death (SCD) risk, which supports the association between poor prognosis in HCM patients and sarcomere mutations[38]. More evidence also suggested that HCM patients carrying genetic mutations are more susceptible to increased interventricular septum and interventricular septal thickness compared to patients without genetic mutations[39]. It was found that mid septal hypertrophy is mainly associated with MYBPC3 variant, while a higher septum-to-posterior wall ratio correlated with MYH7 variant. Mutations in MYH7, MYBPC3, and other sarcomere or myofilament genes (TNNI3, TPM1, and TNNT2) have been shown to be associated with increased hypertrophy of the LV anterior wall, interventricular septum, and lateral wall. In contrast, ALPK3-associated hypertrophy chiefly presented in the apical region, while hypertrophy related to TTN and OBSCN mutations exhibited a uniform distribution across the myocardium. The pattern of hypertrophy varies with the type and category of gene mutation, which provides valuable diagnostic insights for early clinical identification of patients with sarcomere variation, so as to carry out genetic testing and family protection as soon as possible.

Pathological differences

Sarcomeric HCM is associated with worse outcomes than diseases caused by non sarcomeric mutations. Patients carrying sarcomere mutations, with strong evidence of pathogenicity, show clinical disease at an earlier age and have a greater burden, HCM related complications occurred earlier. After controlling for gender and ethnicity, the presence of sarcomere mutations increased the risk of all adverse outcomes by more than 2-fold, with the highest risk of ventricular arrhythmias[40]. Moreover, in recent years, our center’s research found that carrying pathogenic sarcomere mutations while carrying rare calcium gene variants (even without sufficient evidence to prove the pathogenicity of these variants) would have a more severe HCM phenotype and prognosis, suggesting that carrying pathogenicity is one of the important factors for the poor prognosis of HCM patients[24].

There is a direct link between Sarcs and poor microcirculation remodeling in HCM. HCM patients with sarcomere filament mutations are characterized by more severe microvascular dysfunction, which is the reason for the increased long-term prevalence of ventricular dysfunction and heart failure in genotype positive patients[41]. Patients with sarcomere mutation positive HCM have a higher degree of myocardial fibrosis and more severe ventricular diastolic dysfunction, which may lead to early onset of late stage heart failure[36]. Research has shown that they suffer from fatigue limitation and congestion symptoms, which are mainly the result of diastolic dysfunction and increased ventricular stiffness, leading to elevated left atrial and LV end diastolic pressure, resulting in pulmonary congestion and decreased exercise capacity[42,43]. Research has found that Sarc + patients with HCM have a higher lifetime risk than Sarc - patients and develop late diastolic heart failure (New York Heart Association III/IV symptoms) at an earlier age. CMR examination confirms that Sarc + patients have more severe LV diastolic dysfunction, providing a reasonable link between late LV diastolic dysfunction and Sarcs[36].

Overall, patients carrying sarcomere mutations have strong evidence of pathogenicity and exhibit clinical symptoms at an earlier age[40]. Several studies have reported that compared to HCM patients with negative sarcomere mutations, HCM patients carrying pathogenic/potentially pathogenic sarcomere mutations have a poorer prognosis. This includes early onset, high incidence rate of SCD, high incidence rate and overall mortality of atrial fibrillation, ventricular arrhythmia, heart failure[44-48], and they also tend to have more serious hypertrophy, microvascular dysfunction and myocardial fibrosis[7,49].

In addition, our center’s research has found that compared with patients with single Sarcs, patients with dual gene mutations are older, have a higher percentage of HCM family history, thicker walls, higher LVOT pressure gradients, more pathological Q waves, and more bundle branch block. During the follow-up period, the primary endpoint of patients with dual gene mutations was higher than the other three groups. Multivariate analysis showed that dual gene variation was an independent predictor of the primary endpoint event in patients, confirming the cumulative effect of dual gene variation, which resulted in more severe HCM phenotype and prognosis in patients[24].

Thin-filament mutations vs thick-filament mutations in Sarc + patients

Sarcomere is the basic unit of myocardial fiber, and its components include thick myofilament, thin myofilament, Z-line, M-line, etc. Gene variation can translate proteins with biological function defects by changing the amino acid sequence. It can also cause structural or functional abnormalities of sarcomere or sarcomere related proteins by reducing the expression level of the encoded protein, which will lead to abnormal myocardial contraction, impaired diastolic function, increased energy consumption, and ultimately cardiac hypertrophy. The thick myofilament are mainly composed of myosin, with the head and tail of myosin molecules arranged in a spiral shape to form the thick myofilament. The myosin head has ATPase activity and can bind to actin and generate force. Common HCM pathogenic thick myofilament genes include MYH7, MYL2, MYL3 and MYBPC3. The thin myofilament is mainly composed of actin, tropomyosin and troponin complex. Common HCM pathogenic thin myofilament genes include TNNT2, TNNI3, TPM1 and ACTC1.

Coppini et al[49] showed that patients with thin-filament mutations exhibited: (1) Milder and atypical distribution of LV hypertrophy and fewer occurrences of outflow tract obstruction compared to those with thick filamentous HCM; (2) The progression rate is higher in functional class III or IV of the New York Heart Association; (3) The incidence of systolic dysfunction or restrictive LV filling was higher during the last evaluation; (4) The prevalence of three-phase LV filling mode increased by 2.4 times; and (5) The incidence of malignant ventricular arrhythmia and SCD is similar[50].

In patients with HCM, thin filament mutations are associated with an increased likelihood of late stage LV dysfunction and heart failure compared to thick filament disease, but the risk of arrhythmia is comparable. Three phase LV filling is particularly common in filamentous HCM, reflecting severe diastolic dysfunction. The study supports the hypothesis that thin-filament HCM is phenotypically different from the more common thick-filament HCM[51-54]. Specific differences in LV morphology, function, and remodeling have been identified, indicating unique potential pathophysiological mechanisms. In the initial assessment, patients with thin-filaments showed smaller LV hypertrophy, typically developing in an apical or concentric pattern, while patients with thick-filaments almost universally exhibited classic asymmetric LV hypertrophy, including basal septum and anterior wall[55]. Therefore, dynamic LVOT obstruction is less common in the thin-filament cohort. The unique clinical and biophysical features of HCM associated with thin-filament mutations are different from the more common thick-filament mutations.

Thin-filament HCM is more likely to be associated with non significant and atypical distribution of LV hypertrophy, increased fibrosis, poor LV remodeling leading to functional deterioration, and more frequent occurrence of three-phase LV filling, reflecting severe diastolic dysfunction[51-54]. The management strategy should consider sufficient monitoring to detect LV dysfunction and symptom progression early, but the risk of arrhythmia does not seem to increase solely due to thin-filament mutation genotype.

MYH7 mutation vs MYBPC3 mutation in thick-filament

It is presumed that mutations in the mechanical enzyme β-cardiac myosin, which drives ventricular contraction, cause HCM by affecting the power output of myosin movement[56]. Β-myosin is a hexamer composed of two heavy chains, two essential light chains and two regulatory light chains. Heavy chains can be further divided into heavy meromyosin and light meromyosin. Heavy meromyosinconsists of sub-fragment 1 and sub-fragment 2, the former is the motor or head domain of myosin and the latter is the first about 40% of the α - helical coiled coil tail[57]. Light meromyosin is the distal tail of myosin, which contains the sequences responsible for thin filament formation. MYH7 181-937 is recognized as an important functional region of the head and neck, and the variation is likely to affect the protein function, and the clinical phenotype is more severe. There are relatively few pathogenic mutations in the rod, even if the disease occurs, the clinical phenotype caused is relatively mild.

Truncated mutations (encoding cMyPPC) or myosin missense mutations result in high contraction and low relaxation. Genetic and biochemical methods have shown that the gradual loss of cMyPPC leads to mutual enhancement of myosin contractility and changes in the dynamic conformation of myosin during relaxation. A treatment strategy that weakens cMyPPC activity may rescue the decrease in myocardial contractility in dilated cardiomyopathy patients, while MYK-461 inhibition of myosin benefits most mutated HCM patients[58].

Generally, there is no significant difference between the location of mutations in the encoded protein domain and the HCM phenotype. However, mutations in the MYH7 gene exhibit a strong preference for the globular head and hinge regions of myosin heavy chain protein, although there have also been descriptions of mutations leading to changes in the rod domain[59]. Phenotypically, mutations in the converter domain and flat surface area of the spherical head of MYH7 are associated with early disease onset. Overall, MYH7 and MYBPC3 are not only the two most common pathogenic genes involved in the basal septum in the typical form of HCM, but also the two most common genes in apical HCM[60].

Previous research data has shown that carriers of MYBPC3 mutations are older than carriers of MYH7 mutations (P = 0.01)[61]. Compared with MYBPC3 or other Sarcs, patients with pathogenic or potentially pathogenic MYH7 mutations tend to have higher late gadolinium enhancement and extracellular volume fractions%[38].

Troponin gene specific prognostic association in thin-filament mutations

The variation of troponin encoding genes is a clear cause of cardiomyopathy, but there is significant clinical heterogeneity. Early studies have clearly shown that compared to sarcomere negative patients, sarcomere positive patients have a poorer prognosis and higher endpoints for cardiovascular death, progression to end-stage heart failure, and non fatal stroke. At the individual gene level, some early correlations have been identified, such as the pathogenicity of residue 92 in TNNT2 and an increase in SCD, as well as an increase in the mutation rate of codon 145 in TNNI3 leading to restrictive cardiomyopathy (RCM) or HCM[60,62]. With the advancement of genetic testing and newly discovered variations, the Troponin gene specific prognostic association has been determined.

The data compiled by Coppini et al[49] shows that TNNC1 probands have the youngest age of diagnosis and the highest incidence of potentially fatal events. TNNT2 and TNNI3 probands have similar mortality, SCD, and ventricular fibrillation records, while TNNI3 probands tend to have RCM. Diagnostic hotspots were isolated in the N-terminus, tropomyosin binding domain, and alpha helic-1 of TNNT2, and probands carrying mutations at these locations showed significant differences in SCD/ventricular fibrillation; P = 0.004, SCD/death/transplantation (P = 0.053), and diagnostic age (P = 0.052); Most of the hotspots in TNNI3 probands are concentrated in the inhibitory region, switch peptide region, and C-terminus, with a higher incidence of heart failure (P = 0.030), as well as less common subtypes of cardiomyopathy such as RCM (P = 0.008). No diagnostic hotspots were found in the TNNC1 proband hotspots, possibly due to the small number of probands and/or the high pathogenicity of all hotspots.

PERSPECTIVE

Applying gene loading PheWAS to a large biobank based on healthcare may elucidate the medical impact of gene variations on human disease phenotypes[63]. The logical first step is to perform PheWAS only on pLOF variants, which has the advantage of being able to inquire about the maximum effect of gene burden on the relevant phenotype, but due to low frequency, it may lead to insufficient efficacy. Studies have shown that clustering pLOF alone for gene load association testing may be more suitable for certain genes (such as MYBPC3), while including other pDM variants selected based on computer prediction may be more advantageous in increasing the number of effect alleles for other genes (such as MYH7)[64].

A key goal of the precision medicine program is to promote a platform through which healthcare providers can make accurate diagnoses based on various personalized health data, including personal genetic information. Genetic testing for HCM patients can determine the exact genetic cause of the disease, improve diagnostic accuracy for patients with ambiguous diagnoses, and allow for cascading screening of family members[65]. However, pathogenic HCM variants discovered in more comprehensive sequencing may also be overlooked in clinical practice, and it is crucial to use the genome first approach of WES to review clinical charts of carriers of pathogenic and potentially harmful variants[66].

In addition, Green et al[29] inhibited the development of ventricular hypertrophy, myocardial cell disorder, and myocardial fibrosis through early and long-term administration of MYK-461, and reduced the expression of hypertrophy and pro fibrotic genes in mice carrying heterozygous human mutations in myosin heavy chain. Identified the small molecule MYK-461, which reduces contractility by decreasing the activity of adenosine triphosphatase in cardiac myosin heavy chain. High dynamic contraction is crucial for the pathology of HCM, and sarcomere contraction inhibitors may be a valuable treatment for HCM.

CONCLUSION

Multiple mechanisms reflecting the diversity of pathogenic genes and mutations are related to the pathogenesis of HCM. The mechanism events in HCM can be divided into four groups of interlocking mechanisms. The main flaw is mutation. The initial or proximal phenotype is defined as the phenotype resulting from the direct impact of mutations on the structure and function of sarcomere proteins. The intermediate (or secondary) phenotype includes molecular changes that occur in response to changes in the structure and function of sarcomere proteins. Examples of the latter include changes in gene expression and activation of signaling pathways, such as the mitogen-activated protein kinase and transforming growth factor beta 1 pathways. The tertiary effect is accompanied by histological and pathological phenotypes, which are the result of numerous secondary molecular events in the myocardium, such as activation of the hypertrophic signaling pathway. These molecular and histological changes lead to the clinical phenotype of HCM (grade four). It should be noted that there are mechanistic differences between HCM cases caused by sarcomere protein mutations and phenotypic conditions, as the latter’s ventricular hypertrophy may be at least partially due to the storage of substances (such as glycogen) and partially due to functional defects in muscle cells, such as impaired contraction.

Sarcomere mutations are associated with early onset age and high incidence rate, and the existence of sarcomere mutations makes the prognosis worse. Age and sarcomere mutation status at the time of diagnosis can predict prognosis, and patients with pathogenic/potentially pathogenic sarcomere mutations have a 2-fold higher risk of developing adverse outcomes compared to those without mutations.

In patients with sarcomere mutations, compared with patients with thick filament mutations, patients with thin filament mutations are associated with an increased likelihood of late stage LV dysfunction and heart failure, reflecting severe diastolic dysfunction. Comparing the two most common coarse muscle filament genes MYH7 and MYBPC3, MYBPC3 mutation carriers have a higher age, but the difference in myocardial fibrosis is not significant.

Among patients with thin myofilament gene mutations, TNNC1 positive probands had the worst prognosis. The mapping of troponin variants identified positions in TNNT2 and TNNI3 that are associated with enhanced pathogenicity, diagnosis of RCM, and increased risk of sudden death, that is, de novo variants, homozygotes, and compound heterozygotes have a poor prognosis. Of course, the research on the relationship between genotype and clinical phenotype is still controversial, and further large sample multi center long-term follow-up research is urgently needed. In this paper, the close relationship between HCM hypertrophy pattern and genetic variation was combed and insights were put forward, which provided a theoretical basis for genetic testing and personalized management of HCM patients.

ACKNOWLEDGEMENTS

We appreciate Professor Yang Yang, School of Life Sciences, Northwest University, China, for his guidance.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cardiac and cardiovascular systems

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade B, Grade C

Novelty: Grade B, Grade B, Grade B, Grade C

Creativity or Innovation: Grade B, Grade B, Grade C, Grade C

Scientific Significance: Grade A, Grade C, Grade C, Grade C

P-Reviewer: Hameed Y, PhD, Assistant Professor, Pakistan; Yu YW, Assistant Professor, PhD, China S-Editor: Bai Y L-Editor: A P-Editor: Wang WB

References
1.  Ommen SR, Ho CY, Asif IM, Balaji S, Burke MA, Day SM, Dearani JA, Epps KC, Evanovich L, Ferrari VA, Joglar JA, Khan SS, Kim JJ, Kittleson MM, Krittanawong C, Martinez MW, Mital S, Naidu SS, Saberi S, Semsarian C, Times S, Waldman CB; Peer Review Committee Members. 2024 AHA/ACC/AMSSM/HRS/PACES/SCMR Guideline for the Management of Hypertrophic Cardiomyopathy: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. Circulation. 2024;149:e1239-e1311.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 40]  [Cited by in RCA: 187]  [Article Influence: 187.0]  [Reference Citation Analysis (0)]
2.  Ommen SR, Mital S, Burke MA, Day SM, Deswal A, Elliott P, Evanovich LL, Hung J, Joglar JA, Kantor P, Kimmelstiel C, Kittleson M, Link MS, Maron MS, Martinez MW, Miyake CY, Schaff HV, Semsarian C, Sorajja P. 2020 AHA/ACC Guideline for the Diagnosis and Treatment of Patients With Hypertrophic Cardiomyopathy: Executive Summary: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol. 2020;76:3022-3055.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 124]  [Cited by in RCA: 194]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
3.  Maron BJ. Clinical Course and Management of Hypertrophic Cardiomyopathy. N Engl J Med. 2018;379:655-668.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 398]  [Cited by in RCA: 617]  [Article Influence: 88.1]  [Reference Citation Analysis (0)]
4.  Tuohy CV, Kaul S, Song HK, Nazer B, Heitner SB. Hypertrophic cardiomyopathy: the future of treatment. Eur J Heart Fail. 2020;22:228-240.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 33]  [Cited by in RCA: 124]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
5.  [Guidelines for the diagnosis and treatment for Chinese adult patients with hypertrophic cardiomyopathy]. Zhonghua Xin Xue Guan Bing Za Zhi. 2017;45:1015-1032.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
6.  Authors/Task Force members; Elliott PM, Anastasakis A, Borger MA, Borggrefe M, Cecchi F, Charron P, Hagege AA, Lafont A, Limongelli G, Mahrholdt H, McKenna WJ, Mogensen J, Nihoyannopoulos P, Nistri S, Pieper PG, Pieske B, Rapezzi C, Rutten FH, Tillmanns C, Watkins H. 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy: the Task Force for the Diagnosis and Management of Hypertrophic Cardiomyopathy of the European Society of Cardiology (ESC). Eur Heart J. 2014;35:2733-2779.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2292]  [Cited by in RCA: 3069]  [Article Influence: 279.0]  [Reference Citation Analysis (0)]
7.  Maron BJ, Desai MY, Nishimura RA, Spirito P, Rakowski H, Towbin JA, Rowin EJ, Maron MS, Sherrid MV. Diagnosis and Evaluation of Hypertrophic Cardiomyopathy: JACC State-of-the-Art Review. J Am Coll Cardiol. 2022;79:372-389.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 291]  [Article Influence: 97.0]  [Reference Citation Analysis (0)]
8.  Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, O'Donnell-Luria AH, Ware JS, Hill AJ, Cummings BB, Tukiainen T, Birnbaum DP, Kosmicki JA, Duncan LE, Estrada K, Zhao F, Zou J, Pierce-Hoffman E, Berghout J, Cooper DN, Deflaux N, DePristo M, Do R, Flannick J, Fromer M, Gauthier L, Goldstein J, Gupta N, Howrigan D, Kiezun A, Kurki MI, Moonshine AL, Natarajan P, Orozco L, Peloso GM, Poplin R, Rivas MA, Ruano-Rubio V, Rose SA, Ruderfer DM, Shakir K, Stenson PD, Stevens C, Thomas BP, Tiao G, Tusie-Luna MT, Weisburd B, Won HH, Yu D, Altshuler DM, Ardissino D, Boehnke M, Danesh J, Donnelly S, Elosua R, Florez JC, Gabriel SB, Getz G, Glatt SJ, Hultman CM, Kathiresan S, Laakso M, McCarroll S, McCarthy MI, McGovern D, McPherson R, Neale BM, Palotie A, Purcell SM, Saleheen D, Scharf JM, Sklar P, Sullivan PF, Tuomilehto J, Tsuang MT, Watkins HC, Wilson JG, Daly MJ, MacArthur DG; Exome Aggregation Consortium. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285-291.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7480]  [Cited by in RCA: 7721]  [Article Influence: 857.9]  [Reference Citation Analysis (0)]
9.  Bahcall OG. Genetic variation: ExAC boosts clinical variant interpretation in rare diseases. Nat Rev Genet. 2016;17:584.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
10.  de Marvao A, McGurk KA, Zheng SL, Thanaj M, Bai W, Duan J, Biffi C, Mazzarotto F, Statton B, Dawes TJW, Savioli N, Halliday BP, Xu X, Buchan RJ, Baksi AJ, Quinlan M, Tokarczuk P, Tayal U, Francis C, Whiffin N, Theotokis PI, Zhang X, Jang M, Berry A, Pantazis A, Barton PJR, Rueckert D, Prasad SK, Walsh R, Ho CY, Cook SA, Ware JS, O'Regan DP. Phenotypic Expression and Outcomes in Individuals With Rare Genetic Variants of Hypertrophic Cardiomyopathy. J Am Coll Cardiol. 2021;78:1097-1110.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
11.  Maurizi N, Michels M, Rowin EJ, Semsarian C, Girolami F, Tomberli B, Cecchi F, Maron MS, Olivotto I, Maron BJ. Clinical Course and Significance of Hypertrophic Cardiomyopathy Without Left Ventricular Hypertrophy. Circulation. 2019;139:830-833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 50]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
12.  Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res. 2017;121:749-770.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 491]  [Cited by in RCA: 922]  [Article Influence: 115.3]  [Reference Citation Analysis (0)]
13.  Lorenzini M, Norrish G, Field E, Ochoa JP, Cicerchia M, Akhtar MM, Syrris P, Lopes LR, Kaski JP, Elliott PM. Penetrance of Hypertrophic Cardiomyopathy in Sarcomere Protein Mutation Carriers. J Am Coll Cardiol. 2020;76:550-559.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 40]  [Cited by in RCA: 124]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
14.  Walsh R, Buchan R, Wilk A, John S, Felkin LE, Thomson KL, Chiaw TH, Loong CCW, Pua CJ, Raphael C, Prasad S, Barton PJ, Funke B, Watkins H, Ware JS, Cook SA. Defining the genetic architecture of hypertrophic cardiomyopathy: re-evaluating the role of non-sarcomeric genes. Eur Heart J. 2017;38:3461-3468.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 46]  [Cited by in RCA: 107]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
15.  Geier C, Gehmlich K, Ehler E, Hassfeld S, Perrot A, Hayess K, Cardim N, Wenzel K, Erdmann B, Krackhardt F, Posch MG, Osterziel KJ, Bublak A, Nägele H, Scheffold T, Dietz R, Chien KR, Spuler S, Fürst DO, Nürnberg P, Ozcelik C. Beyond the sarcomere: CSRP3 mutations cause hypertrophic cardiomyopathy. Hum Mol Genet. 2008;17:2753-2765.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 142]  [Cited by in RCA: 129]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
16.  Friedrich FW, Wilding BR, Reischmann S, Crocini C, Lang P, Charron P, Müller OJ, McGrath MJ, Vollert I, Hansen A, Linke WA, Hengstenberg C, Bonne G, Morner S, Wichter T, Madeira H, Arbustini E, Eschenhagen T, Mitchell CA, Isnard R, Carrier L. Evidence for FHL1 as a novel disease gene for isolated hypertrophic cardiomyopathy. Hum Mol Genet. 2012;21:3237-3254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 84]  [Cited by in RCA: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
17.  Chiu C, Bagnall RD, Ingles J, Yeates L, Kennerson M, Donald JA, Jormakka M, Lind JM, Semsarian C. Mutations in alpha-actinin-2 cause hypertrophic cardiomyopathy: a genome-wide analysis. J Am Coll Cardiol. 2010;55:1127-1135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 133]  [Cited by in RCA: 151]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
18.  Girolami F, Iascone M, Tomberli B, Bardi S, Benelli M, Marseglia G, Pescucci C, Pezzoli L, Sana ME, Basso C, Marziliano N, Merlini PA, Fornaro A, Cecchi F, Torricelli F, Olivotto I. Novel α-actinin 2 variant associated with familial hypertrophic cardiomyopathy and juvenile atrial arrhythmias: a massively parallel sequencing study. Circ Cardiovasc Genet. 2014;7:741-750.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 54]  [Cited by in RCA: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
19.  Valdés-Mas R, Gutiérrez-Fernández A, Gómez J, Coto E, Astudillo A, Puente DA, Reguero JR, Álvarez V, Morís C, León D, Martín M, Puente XS, López-Otín C. Mutations in filamin C cause a new form of familial hypertrophic cardiomyopathy. Nat Commun. 2014;5:5326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 123]  [Cited by in RCA: 149]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
20.  Osio A, Tan L, Chen SN, Lombardi R, Nagueh SF, Shete S, Roberts R, Willerson JT, Marian AJ. Myozenin 2 is a novel gene for human hypertrophic cardiomyopathy. Circ Res. 2007;100:766-768.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 161]  [Cited by in RCA: 130]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
21.  Su M, Wang J, Kang L, Wang Y, Zou Y, Feng X, Wang D, Ahmad F, Zhou X, Hui R, Song L. Rare variants in genes encoding MuRF1 and MuRF2 are modifiers of hypertrophic cardiomyopathy. Int J Mol Sci. 2014;15:9302-9313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
22.  Priganc M, Zigová M, Boroňová I, Bernasovská J, Dojčáková D, Szabadosová V, Mydlárová Blaščáková M, Tóthová I, Kmec J, Bernasovský I. Analysis of SCN5A Gene Variants in East Slovak Patients with Cardiomyopathy. J Clin Lab Anal. 2017;31:e22037.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
23.  Wang B, Guo RQ, Wang J, Yang F, Zuo L, Liu Y, Shao H, Ju Y, Sun C, Xu L, Zhang YM, Wang LF, Liu LW. The Cumulative Effects of the MYH7-V878A and CACNA1C-A1594V Mutations in a Chinese Family with Hypertrophic Cardiomyopathy. Cardiology. 2017;138:228-237.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
24.  Zhao J, Wang B, Ta S, Lu X, Zhao X, Liu J, Yuan J, Wang J, Liu L. Association between Hypertrophic Cardiomyopathy and Variations in Sarcomere Gene and Calcium Channel Gene in Adults. Cardiology. 2024;149:440-450.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
25.  Rowin EJ, Maron BJ, Haas TS, Garberich RF, Wang W, Link MS, Maron MS. Hypertrophic Cardiomyopathy With Left Ventricular Apical Aneurysm: Implications for Risk Stratification and Management. J Am Coll Cardiol. 2017;69:761-773.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 175]  [Cited by in RCA: 265]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
26.  Norrish G, Jager J, Field E, Quinn E, Fell H, Lord E, Cicerchia MN, Ochoa JP, Cervi E, Elliott PM, Kaski JP. Yield of Clinical Screening for Hypertrophic Cardiomyopathy in Child First-Degree Relatives. Circulation. 2019;140:184-192.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 44]  [Cited by in RCA: 69]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
27.  Elliott PM, Gimeno Blanes JR, Mahon NG, Poloniecki JD, McKenna WJ. Relation between severity of left-ventricular hypertrophy and prognosis in patients with hypertrophic cardiomyopathy. Lancet. 2001;357:420-424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 327]  [Cited by in RCA: 307]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
28.  Dainis AM, Ashley EA. Cardiovascular Precision Medicine in the Genomics Era. JACC Basic Transl Sci. 2018;3:313-326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
29.  Green EM, Wakimoto H, Anderson RL, Evanchik MJ, Gorham JM, Harrison BC, Henze M, Kawas R, Oslob JD, Rodriguez HM, Song Y, Wan W, Leinwand LA, Spudich JA, McDowell RS, Seidman JG, Seidman CE. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science. 2016;351:617-621.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 340]  [Cited by in RCA: 529]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
30.  Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, Koski A, Ji D, Hayama T, Ahmed R, Darby H, Van Dyken C, Li Y, Kang E, Park AR, Kim D, Kim ST, Gong J, Gu Y, Xu X, Battaglia D, Krieg SA, Lee DM, Wu DH, Wolf DP, Heitner SB, Belmonte JCI, Amato P, Kim JS, Kaul S, Mitalipov S. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548:413-419.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 588]  [Cited by in RCA: 591]  [Article Influence: 73.9]  [Reference Citation Analysis (0)]
31.  Mearini G, Stimpel D, Geertz B, Weinberger F, Krämer E, Schlossarek S, Mourot-Filiatre J, Stoehr A, Dutsch A, Wijnker PJ, Braren I, Katus HA, Müller OJ, Voit T, Eschenhagen T, Carrier L. Mybpc3 gene therapy for neonatal cardiomyopathy enables long-term disease prevention in mice. Nat Commun. 2014;5:5515.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 97]  [Cited by in RCA: 135]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
32.  Harper AR, Goel A, Grace C, Thomson KL, Petersen SE, Xu X, Waring A, Ormondroyd E, Kramer CM, Ho CY, Neubauer S; HCMR Investigators, Tadros R, Ware JS, Bezzina CR, Farrall M, Watkins H. Common genetic variants and modifiable risk factors underpin hypertrophic cardiomyopathy susceptibility and expressivity. Nat Genet. 2021;53:135-142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 231]  [Cited by in RCA: 201]  [Article Influence: 50.3]  [Reference Citation Analysis (0)]
33.  Chan RH, Maron BJ, Olivotto I, Pencina MJ, Assenza GE, Haas T, Lesser JR, Gruner C, Crean AM, Rakowski H, Udelson JE, Rowin E, Lombardi M, Cecchi F, Tomberli B, Spirito P, Formisano F, Biagini E, Rapezzi C, De Cecco CN, Autore C, Cook EF, Hong SN, Gibson CM, Manning WJ, Appelbaum E, Maron MS. Prognostic value of quantitative contrast-enhanced cardiovascular magnetic resonance for the evaluation of sudden death risk in patients with hypertrophic cardiomyopathy. Circulation. 2014;130:484-495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 592]  [Cited by in RCA: 755]  [Article Influence: 68.6]  [Reference Citation Analysis (1)]
34.  Mentias A, Raeisi-Giglou P, Smedira NG, Feng K, Sato K, Wazni O, Kanj M, Flamm SD, Thamilarasan M, Popovic ZB, Lever HM, Desai MY. Late Gadolinium Enhancement in Patients With Hypertrophic Cardiomyopathy and Preserved Systolic Function. J Am Coll Cardiol. 2018;72:857-870.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 98]  [Cited by in RCA: 168]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
35.  Neubauer S, Kolm P, Ho CY, Kwong RY, Desai MY, Dolman SF, Appelbaum E, Desvigne-Nickens P, DiMarco JP, Friedrich MG, Geller N, Harper AR, Jarolim P, Jerosch-Herold M, Kim DY, Maron MS, Schulz-Menger J, Piechnik SK, Thomson K, Zhang C, Watkins H, Weintraub WS, Kramer CM; HCMR Investigators. Distinct Subgroups in Hypertrophic Cardiomyopathy in the NHLBI HCM Registry. J Am Coll Cardiol. 2019;74:2333-2345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 84]  [Cited by in RCA: 211]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
36.  Chen CJ, Su MM, Liao YC, Chang FL, Wu CK, Lin LY, Chen YS, Lin YH, Hwang JJ, Yu SL, Kao HL, Chen WJ, Lu TP, Shih CY, Yeh SS, Yang DH, Lai LP, Juang JJ. Long-term outcomes and left ventricular diastolic function of sarcomere mutation-positive and mutation-negative patients with hypertrophic cardiomyopathy: a prospective cohort study. Eur Heart J Cardiovasc Imaging. 2020;jeaa317.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
37.  Wang J, Yang S, Ma X, Zhao K, Yang K, Yu S, Yin G, Dong Z, Song Y, Cui C, Li J, Wang C, Hao J, Lu M, Chen X, Zhao S. Assessment of late gadolinium enhancement in hypertrophic cardiomyopathy improves risk stratification based on current guidelines. Eur Heart J. 2023;44:4781-4792.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 26]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
38.  Chung H, Kim Y, Park CH, Kim JY, Min PK, Yoon YW, Kim TH, Lee BK, Hong BK, Rim SJ, Kwon HM, Lee KA, Choi EY. Effect of sarcomere and mitochondria-related mutations on myocardial fibrosis in patients with hypertrophic cardiomyopathy. J Cardiovasc Magn Reson. 2021;23:18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
39.  Zhou N, Weng H, Zhao W, Tang L, Ge Z, Tian F, Meng F, Pan C, Shu X. Gene-echocardiography: refining genotype-phenotype correlations in hypertrophic cardiomyopathy. Eur Heart J Cardiovasc Imaging. 2023;25:127-135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
40.  Ho CY, Day SM, Ashley EA, Michels M, Pereira AC, Jacoby D, Cirino AL, Fox JC, Lakdawala NK, Ware JS, Caleshu CA, Helms AS, Colan SD, Girolami F, Cecchi F, Seidman CE, Sajeev G, Signorovitch J, Green EM, Olivotto I. Genotype and Lifetime Burden of Disease in Hypertrophic Cardiomyopathy: Insights from the Sarcomeric Human Cardiomyopathy Registry (SHaRe). Circulation. 2018;138:1387-1398.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 504]  [Cited by in RCA: 563]  [Article Influence: 80.4]  [Reference Citation Analysis (0)]
41.  Olivotto I, Girolami F, Sciagrà R, Ackerman MJ, Sotgia B, Bos JM, Nistri S, Sgalambro A, Grifoni C, Torricelli F, Camici PG, Cecchi F. Microvascular function is selectively impaired in patients with hypertrophic cardiomyopathy and sarcomere myofilament gene mutations. J Am Coll Cardiol. 2011;58:839-848.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 111]  [Cited by in RCA: 123]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
42.  Maron BJ, Spirito P, Green KJ, Wesley YE, Bonow RO, Arce J. Noninvasive assessment of left ventricular diastolic function by pulsed Doppler echocardiography in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol. 1987;10:733-742.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 295]  [Cited by in RCA: 280]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
43.  Briguori C, Betocchi S, Romano M, Manganelli F, Angela Losi M, Ciampi Q, Gottilla R, Lombardi R, Condorelli M, Chiariello M. Exercise capacity in hypertrophic cardiomyopathy depends on left ventricular diastolic function. Am J Cardiol. 1999;84:309-315.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 59]  [Cited by in RCA: 55]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
44.  Harris KM, Spirito P, Maron MS, Zenovich AG, Formisano F, Lesser JR, Mackey-Bojack S, Manning WJ, Udelson JE, Maron BJ. Prevalence, clinical profile, and significance of left ventricular remodeling in the end-stage phase of hypertrophic cardiomyopathy. Circulation. 2006;114:216-225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 450]  [Cited by in RCA: 474]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
45.  Ichida M, Nishimura Y, Kario K. Clinical significance of left ventricular apical aneurysms in hypertrophic cardiomyopathy patients: the role of diagnostic electrocardiography. J Cardiol. 2014;64: 265-272.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
46.  Vriesendorp PA, Schinkel AF, Van Cleemput J, Willems R, Jordaens LJ, Theuns DA, van Slegtenhorst MA, de Ravel TJ, ten Cate FJ, Michels M. Implantable cardioverter-defibrillators in hypertrophic cardiomyopathy: patient outcomes, rate of appropriate and inappropriate interventions, and complications. Am Heart J. 2013;166:496-502.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 63]  [Cited by in RCA: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
47.  Olivotto I, Maron MS, Adabag AS, Casey SA, Vargiu D, Link MS, Udelson JE, Cecchi F, Maron BJ. Gender-related differences in the clinical presentation and outcome of hypertrophic cardiomyopathy. J Am Coll Cardiol. 2005;46:480-487.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 269]  [Cited by in RCA: 299]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
48.  Decker JA, Rossano JW, Smith EO, Cannon B, Clunie SK, Gates C, Jefferies JL, Kim JJ, Price JF, Dreyer WJ, Towbin JA, Denfield SW. Risk factors and mode of death in isolated hypertrophic cardiomyopathy in children. J Am Coll Cardiol. 2009;54:250-254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 95]  [Cited by in RCA: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
49.  Coppini R, Ho CY, Ashley E, Day S, Ferrantini C, Girolami F, Tomberli B, Bardi S, Torricelli F, Cecchi F, Mugelli A, Poggesi C, Tardiff J, Olivotto I. Clinical phenotype and outcome of hypertrophic cardiomyopathy associated with thin-filament gene mutations. J Am Coll Cardiol. 2014;64:2589-2600.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 84]  [Cited by in RCA: 129]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
50.  Tardiff JC. Thin filament mutations: developing an integrative approach to a complex disorder. Circ Res. 2011;108:765-782.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 128]  [Cited by in RCA: 125]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
51.  Tardiff JC. Sarcomeric proteins and familial hypertrophic cardiomyopathy: linking mutations in structural proteins to complex cardiovascular phenotypes. Heart Fail Rev. 2005;10:237-248.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 141]  [Cited by in RCA: 139]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
52.  Doolan A, Tebo M, Ingles J, Nguyen L, Tsoutsman T, Lam L, Chiu C, Chung J, Weintraub RG, Semsarian C. Cardiac troponin I mutations in Australian families with hypertrophic cardiomyopathy: clinical, genetic and functional consequences. J Mol Cell Cardiol. 2005;38:387-393.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 34]  [Cited by in RCA: 34]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
53.  Pasquale F, Syrris P, Kaski JP, Mogensen J, McKenna WJ, Elliott P. Long-term outcomes in hypertrophic cardiomyopathy caused by mutations in the cardiac troponin T gene. Circ Cardiovasc Genet. 2012;5:10-17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 76]  [Cited by in RCA: 91]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
54.  Maron MS, Maron BJ, Harrigan C, Buros J, Gibson CM, Olivotto I, Biller L, Lesser JR, Udelson JE, Manning WJ, Appelbaum E. Hypertrophic cardiomyopathy phenotype revisited after 50 years with cardiovascular magnetic resonance. J Am Coll Cardiol. 2009;54:220-228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 319]  [Cited by in RCA: 330]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
55.  Spudich JA. Hypertrophic and dilated cardiomyopathy: four decades of basic research on muscle lead to potential therapeutic approaches to these devastating genetic diseases. Biophys J. 2014;106:1236-1249.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 188]  [Cited by in RCA: 212]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
56.  Adhikari AS, Trivedi DV, Sarkar SS, Song D, Kooiker KB, Bernstein D, Spudich JA, Ruppel KM. β-Cardiac myosin hypertrophic cardiomyopathy mutations release sequestered heads and increase enzymatic activity. Nat Commun. 2019;10:2685.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 34]  [Cited by in RCA: 58]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
57.  Toepfer CN, Wakimoto H, Garfinkel AC, McDonough B, Liao D, Jiang J, Tai AC, Gorham JM, Lunde IG, Lun M, Lynch TL 4th, McNamara JW, Sadayappan S, Redwood CS, Watkins HC, Seidman JG, Seidman CE. Hypertrophic cardiomyopathy mutations in MYBPC3 dysregulate myosin. Sci Transl Med. 2019;11:eaat1199.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 136]  [Cited by in RCA: 143]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
58.  Homburger JR, Green EM, Caleshu C, Sunitha MS, Taylor RE, Ruppel KM, Metpally RP, Colan SD, Michels M, Day SM, Olivotto I, Bustamante CD, Dewey FE, Ho CY, Spudich JA, Ashley EA. Multidimensional structure-function relationships in human β-cardiac myosin from population-scale genetic variation. Proc Natl Acad Sci U S A. 2016;113:6701-6706.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 92]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
59.  Towe EC, Bos JM, Ommen SR, Gersh BJ, Ackerman MJ. Genotype-Phenotype Correlations in Apical Variant Hypertrophic Cardiomyopathy. Congenit Heart Dis. 2015;10:E139-E145.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 31]  [Cited by in RCA: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
60.  Debl K, Djavidani B, Buchner S, Poschenrieder F, Feuerbach S, Riegger G, Luchner A. Triphasic mitral inflow pattern and regional triphasic mitral annulus velocity in hypertrophic cardiomyopathy. J Am Soc Echocardiogr. 2008;21:407.e5-407.e6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
61.  Michels M, Soliman OI, Phefferkorn J, Hoedemaekers YM, Kofflard MJ, Dooijes D, Majoor-Krakauer D, Ten Cate FJ. Disease penetrance and risk stratification for sudden cardiac death in asymptomatic hypertrophic cardiomyopathy mutation carriers. Eur Heart J. 2009;30:2593-2598.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 69]  [Cited by in RCA: 72]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
62.  Bruder O, Wagner A, Jensen CJ, Schneider S, Ong P, Kispert EM, Nassenstein K, Schlosser T, Sabin GV, Sechtem U, Mahrholdt H. Myocardial scar visualized by cardiovascular magnetic resonance imaging predicts major adverse events in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol. 2010;56:875-887.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 425]  [Cited by in RCA: 446]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
63.  Park J, Lucas AM, Zhang X, Chaudhary K, Cho JH, Nadkarni G, Dobbyn A, Chittoor G, Josyula NS, Katz N, Breeyear JH, Ahmadmehrabi S, Drivas TG, Chavali VRM, Fasolino M, Sawada H, Daugherty A, Li Y, Zhang C, Bradford Y, Weaver J, Verma A, Judy RL, Kember RL, Overton JD, Reid JG, Ferreira MAR, Li AH, Baras A, LeMaire SA, Shen YH, Naji A, Kaestner KH, Vahedi G, Edwards TL, Chen J, Damrauer SM, Justice AE, Do R, Ritchie MD, Rader DJ. Exome-wide evaluation of rare coding variants using electronic health records identifies new gene-phenotype associations. Nat Med. 2021;27:66-72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 42]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
64.  Park J, Levin MG, Haggerty CM, Hartzel DN, Judy R, Kember RL, Reza N; Regeneron Genetics Center, Ritchie MD, Owens AT, Damrauer SM, Rader DJ. A genome-first approach to aggregating rare genetic variants in LMNA for association with electronic health record phenotypes. Genet Med. 2020;22:102-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
65.  Cirino AL, Seidman CE, Ho CY. Genetic Testing and Counseling for Hypertrophic Cardiomyopathy. Cardiol Clin. 2019;37:35-43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
66.  Bagnall RD, Ingles J, Dinger ME, Cowley MJ, Ross SB, Minoche AE, Lal S, Turner C, Colley A, Rajagopalan S, Berman Y, Ronan A, Fatkin D, Semsarian C. Whole Genome Sequencing Improves Outcomes of Genetic Testing in Patients With Hypertrophic Cardiomyopathy. J Am Coll Cardiol. 2018;72:419-429.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 106]  [Cited by in RCA: 145]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]