138 results on '"Cardiomyopathy, Hypertrophic genetics"'
Search Results
2. Sarcomere gene mutations in hypertrophic cardiomyopathy: how to clinically manage this information?
- Author
-
Sanna GD and Finocchiaro G
- Subjects
- Humans, Disease Management, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Sarcomeres genetics, Mutation
- Abstract
Competing Interests: Declaration of competing interest The Authors have no conflicts of interest to declare related to this research.
- Published
- 2024
- Full Text
- View/download PDF
3. Comprehensive Proteomic Profiling of Human Myocardium Reveals Signaling Pathways Dysregulated in Hypertrophic Cardiomyopathy.
- Author
-
Lumish HS, Sherrid MV, Janssen PML, Ferrari G, Hasegawa K, Castillero E, Adlestein E, Swistel DG, Topkara VK, Maurer MS, Reilly MP, and Shimada YJ
- Subjects
- Humans, Male, Female, Middle Aged, Case-Control Studies, Adult, Gene Expression Profiling methods, Aged, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Proteomics methods, Signal Transduction, Myocardium metabolism
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiac disease. Signaling pathways that link genetic sequence variants to clinically overt HCM and progression to severe forms of HCM remain unknown., Objectives: The purpose of this study was to identify signaling pathways that are differentially regulated in HCM, using proteomic profiling of human myocardium, confirmed with transcriptomic profiling., Methods: In this multicenter case-control study, myocardial samples were obtained from cases with HCM and control subjects with nonfailing hearts. Proteomic profiling of 7,289 proteins from myocardial samples was performed using the SomaScan assay (SomaLogic). Pathway analysis of differentially expressed proteins was performed, using a false discovery rate <0.05. Pathway analysis of proteins whose concentrations correlated with clinical indicators of severe HCM (eg, reduced left ventricular ejection fraction, atrial fibrillation, and ventricular tachyarrhythmias) was also executed. Confirmatory analysis of differentially expressed genes was performed using myocardial transcriptomic profiling., Results: The study included 99 HCM cases and 15 control subjects. Pathway analysis of differentially expressed proteins revealed dysregulation of the Ras-mitogen-activated protein kinase, ubiquitin-mediated proteolysis, angiogenesis-related (eg, hypoxia-inducible factor-1, vascular endothelial growth factor), and Hippo pathways. Pathways known to be dysregulated in HCM, including metabolic, inflammatory, and extracellular matrix pathways, were also dysregulated. Pathway analysis of proteins associated with clinical indicators of severe HCM and of differentially expressed genes supported these findings., Conclusions: The present study represents the most comprehensive (>7,000 proteins) and largest-scale (n = 99 HCM cases) proteomic profiling of human HCM myocardium to date. Proteomic profiling and confirmatory transcriptomic profiling elucidate dysregulation of both newly recognized (eg, Ras-mitogen-activated protein kinase) and known pathways associated with pathogenesis and progression to severe forms of HCM., Competing Interests: Funding Support and Author Disclosures This work was supported by the National Institutes of Health (R01 HL157216 and R01 HL168382 to Dr Shimada, UL1 TR001873 to Dr Reilly, K24 HL107643 to Dr Reilly, K24 AG036778 to Dr Maurer, R01 HL170132 to Dr Topkara, R01 HL131872 to Dr Ferrari, and T32 HL007854 to Dr Lumish), the American Heart Association (2 National Clinical and Population Research Awards, 1 Career Development Award, and 1 Transformational Project Award to Dr Shimada), Korea Institute of Oriental Medicine (W22005 to Dr Shimada), Feldstein Medical Foundation (to Dr Shimada), Columbia University Irving Medical Center Precision Medicine Pilot Award (to Dr Shimada), and Columbia University Irving Medical Center Marjorie and Lewis Katz Cardiovascular Research Prize (to Dr Shimada). The funding organizations did not have any role in the study design, collection, analysis, or interpretation of data, in writing of the manuscript, or in the decision to submit the paper for publication. The researchers were independent from the funding organizations. Dr Maurer has received consulting income from Akcea, Alnylam, Eidos Therapeutics, Pfizer, Prothena, Novo Nordisk, and Intellia. Dr Shimada has received research funding from Bristol Myers Squibb; and has received consulting income from Bristol Myers Squibb and Moderna Japan. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose., (Copyright © 2024 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Carrying both the heterozygous Myh6-R453C and Tnnt2-R92W mutations aggravate the hypertrophic cardiomyopathy phenotype in mice.
- Author
-
Lu M, Li S, Han Z, Ma B, Wang L, Wan F, Lei S, Nie Y, and Wang J
- Subjects
- Animals, Humans, Male, Mice, Cardiac Myosins genetics, Disease Models, Animal, Mice, Inbred C57BL, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Heterozygote, Mutation, Myosin Heavy Chains genetics, Phenotype, Troponin T genetics
- Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disease of the heart muscle that is dominated by variations in eight genes encoding sarcomere proteins. Although there are clinical or basic research reports that carrying double mutations can lead to more severe HCM phenotypes, there are also research reports that after reanalyzing the reported mutations, the severity of clinical symptoms in patients with double mutations did not significantly increase compared to patients with only one mutation. To determine whether double pathogenic mutations can aggravate the phenotype of hypertrophic cardiomyopathy in mice, we constructed mice carrying single pathogenic heterozygous mutation Myh6-R453C or Tnnt2-R92W and mice carrying both pathogenic heterozygous mutations. Our results showed that mice with double heterozygous mutations exhibited significant hypertrophic cardiomyopathy phenotypes at 4 weeks of age, and the degree of hypertrophy was significantly higher than that of single heterozygous mutant mice of the same age. Our study suggests that carrying the two pathogenic heterozygous mutations simultaneously can aggravate the phenotype of HCM in mice, which provides experimental evidence for the genotype-phenotype relationship of double pathogenic mutations and provides reference significance for clinical risk stratification of HCM patients., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Causal relationship between leukocyte telomere length and two cardiomyopathies based on a bidirectional Mendelian randomization approach.
- Author
-
Li J, Hu L, and Huang X
- Subjects
- Humans, Telomere genetics, Cardiomyopathy, Dilated genetics, Cardiomyopathies genetics, Cardiomyopathy, Hypertrophic genetics, Genetic Predisposition to Disease, Mendelian Randomization Analysis, Leukocytes
- Abstract
This study aims to employ the Mendelian randomization (MR) approach to investigate the relationship between leukocyte telomere length (TL) and 2 prevalent forms of cardiomyopathies. Using R software (4.3.1) for MR study, independent genetic variants associated with leukocyte TL were extracted from the Integrative Epidemiology Unit database, while cardiomyopathies data were pooled from FinnGen and European Bioinformatics Institute databases. Analytical methodologies included inverse-variance weighting, MR-Egger regression, and weighted median methods. Further analyses involved MR-Egger intercept and MR-PRESSO for handling horizontal pleiotropy and Cochran Q test for study heterogeneity. Our forward Mendelian randomization study indicates a positive correlation between longer leukocyte TL and the risk of 2 forms of cardiomyopathies: the longer the leukocyte telomere, the higher is the risk of cardiomyopathies. Specifically, for hypertrophic obstructive cardiomyopathy the OR is 2.23 (95% CI: 1.19-4.14, P = .01), for hypertrophic cardiomyopathy the OR is 1.80 (95% CI: 1.14-2.85, P = .01), and for dilated cardiomyopathy the OR is 1.32 (95% CI: 1.01-1.71, P = .04). In contrast, our reverse Mendelian randomization showed that cardiomyopathies were not directly associated with TL, and the inverse-variance-weighted test was not statistically significant for any of the 3 (P > .05). The reliability tests for the forward Mendelian randomization, including both MR-Egger intercept and MR-PRESSO tests, show no evidence of horizontal pleiotropy, and Cochran Q test indicates no heterogeneity. The "leave-one-out" sensitivity analysis revealed no outlier genes. The reliability tests for the reverse Mendelian randomization, including both MR-Egger intercept and MR-PRESSO tests, also indicate no genetic pleiotropy. Despite the heterogeneity shown in our study between hypertrophic cardiomyopathy and leukocyte TL, the sensitivity analysis did not identify any anomalies. Our Mendelian randomization study suggests that longer leukocyte TL is associated with an increased risk of hypertrophic obstructive cardiomyopathy, hypertrophic cardiomyopathy, and dilated cardiomyopathy. However, the onset of these 2 kinds of disease does not directly lead to changes in leukocyte TL., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 the Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
6. Epigenetic Study of Cohort of Monozygotic Twins With Hypertrophic Cardiomyopathy Due to MYBPC3 (Cardiac Myosin-Binding Protein C).
- Author
-
Peñarroya A, Lorca R, Rodríguez Reguero JJ, Gómez J, Avanzas P, Tejedor JR, Fernandez AF, and Fraga MF
- Subjects
- Humans, Male, Female, Middle Aged, Cardiomyopathy, Hypertrophic genetics, Adult, Phenotype, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular physiopathology, Diseases in Twins genetics, Diseases in Twins blood, Genetic Predisposition to Disease, Severity of Illness Index, Twins, Monozygotic genetics, DNA Methylation, Epigenesis, Genetic, Carrier Proteins genetics
- Abstract
Background: Hypertrophic cardiomyopathy is an autosomal dominant cardiac disease. The mechanisms that determine its variable expressivity are poorly understood. Epigenetics could play a crucial role in bridging the gap between genotype and phenotype by orchestrating the interplay between the environment and the genome regulation. In this study we aimed to establish a possible correlation between the peripheral blood DNA methylation patterns and left ventricular hypertrophy severity in patients with hypertrophic cardiomyopathy, evaluating the potential impact of lifestyle variables and providing a biological context to the observed changes., Methods and Results: Methylation data were obtained from peripheral blood samples (Infinium MethylationEPIC BeadChip arrays). We employed multiple pair-matched models to extract genomic positions whose methylation correlates with the degree of left ventricular hypertrophy in 3 monozygotic twin pairs carrying the same founder pathogenic variant ( MYBPC3 p.Gly263Ter). This model enables the isolation of the environmental influence, beyond age, on DNA methylation changes by removing the genetic background. Our results revealed a more anxious personality among more severely affected individuals. We identified 56 differentially methylated positions that exhibited moderate, proportional changes in methylation associated with left ventricular hypertrophy. These differentially methylated positions were enriched in regions regulated by repressor histone marks and tended to cluster at genes involved in left ventricular hypertrophy development, such as HOXA5 , TRPC3 , UCN3 , or PLSCR2 , suggesting that changes in peripheral blood may reflect myocardial alterations., Conclusions: We present a unique pair-matched model, based on 3 monozygotic twin pairs carrying the same founder pathogenic variant and different phenotypes. This study provides further evidence of the pivotal role of epigenetics in hypertrophic cardiomyopathy variable expressivity.
- Published
- 2024
- Full Text
- View/download PDF
7. Family Screening in Hypertrophic Cardiomyopathy: Identification of Relatives With Low Yield From Systematic Follow-Up.
- Author
-
Silajdzija E, Rasmus Vissing C, Basse Christensen E, Lamiokor Mills H, Olivia Kock T, Andersen LJ, Snoer M, Thune JJ, Daniel Bartels E, Axelsson Raja A, Hørby Christensen A, and Bundgaard H
- Subjects
- Humans, Female, Male, Adult, Retrospective Studies, Follow-Up Studies, Middle Aged, Denmark epidemiology, Pedigree, Genetic Testing methods, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is a common inherited cardiac disease, and clinical and genetic family screening is recommended by guidelines., Objectives: This study sought to investigate the diagnostic yield of screening relatives of HCM patients and identify predictive factors for HCM development during long-term follow-up in relatives from gene-elusive families., Methods: This was a retrospective cohort study of families screened at clinics for inherited cardiomyopathies in Eastern Denmark, from 2006 to 2023., Results: We included 1,230 relatives (55% female; age: 42 ± 17 years) from 531 families. The combined clinical and genetic yield at baseline was 26% (n = 321). After 7 years (mean) of follow-up (6,762 person-years), 43 (4%) additional relatives developed HCM. The strongest predictors of developing HCM were carrying a likely pathogenic/pathogenic variant (HR: 4.58; 95% CI: 2.50-8.40; P < 0.001) and larger left ventricular maximum wall thickness (MWT) (HR: 2.21 per mm; 95% CI: 1.76-2.77 per mm; P < 0.001). In gene-elusive families, we found that an MWT of ≥10 mm represented the optimal classification threshold for developing HCM (area under the curve: 0.80), with only 2 (0.4%) relatives from gene-elusive families with an MWT of <10 mm developing HCM during follow-up., Conclusions: In HCM, the diagnostic yield of a single screening visit was 1 in 4, and the additional yield during 7 years of follow-up was 4%. Gene carriers and relatives from gene-elusive families with a baseline MWT of ≥10 mm were at the highest risk of developing HCM during follow-up. These findings may inform future recommendations on the management of relatives of HCM patients., Competing Interests: Funding Support and Author Disclosures The study was supported by Rigshospitalet Research Council. The funder had no part in the design of the study, the collection, analysis, or interpretation of data or publication. The authors have reported that they have no relationships relevant to the contents of this paper to disclose., (Copyright © 2024 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Klf9 is essential for cardiac mitochondrial homeostasis.
- Author
-
Zhang L, Zhang M, Huang J, Huang J, Zhang Y, Zhang Y, Chen H, Wang C, Xi X, Fan H, Wang J, Jiang D, Tian J, Zhang J, and Chang Y
- Subjects
- Animals, Humans, Disease Models, Animal, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Angiotensin II pharmacology, Angiotensin II metabolism, Cells, Cultured, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors metabolism, Kruppel-Like Transcription Factors deficiency, Myocytes, Cardiac metabolism, GTP Phosphohydrolases genetics, GTP Phosphohydrolases metabolism, Mitochondria, Heart metabolism, Mitochondria, Heart genetics, Mice, Knockout, Mitophagy, Energy Metabolism, Homeostasis, Mitochondrial Dynamics, Heart Failure metabolism, Heart Failure genetics
- Abstract
Mitochondrial dynamics and mitophagy are intimately linked physiological processes that are essential for cardiac homeostasis. Here we show that cardiac Krüppel-like factor 9 (Klf9) is dysregulated in human and rodent cardiomyopathy. Both global and cardiac-specific Klf9-deficient mice displayed hypertrophic cardiomyopathy. Klf9 knockout led to mitochondrial disarray and fragmentation, impairing mitochondrial respiratory function in cardiomyocytes. Furthermore, cardiac Klf9 deficiency inhibited mitophagy, thereby causing accumulation of dysfunctional mitochondria and acceleration of heart failure in response to angiotensin II treatment. In contrast, cardiac-specific Klf9 transgene improved cardiac systolic function. Mechanistically, Klf9 knockout decreased the expression of PGC-1α and its target genes involved in mitochondrial energy metabolism. Moreover, Klf9 controlled the expression of Mfn2, thereby regulating mitochondrial dynamics and mitophagy. Finally, adeno-associated virus-mediated Mfn2 rescue in Klf9-CKO hearts improved cardiac mitochondrial and systolic function. Thus, Klf9 integrates cardiac energy metabolism, mitochondrial dynamics and mitophagy. Modulating Klf9 activity may have therapeutic potential in the treatment of heart failure., Competing Interests: Competing interests All authors declare no competing interests. There are no conflicts of interest in the submission of this manuscript, which was approved by all authors for publication. We also confirm that this work is original. It has not been published elsewhere and is not currently under consideration for publication elsewhere., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
9. Integration analysis using bioinformatics and experimental validation on cellular signalling for sex differences of hypertrophic cardiomyopathy.
- Author
-
Kuang H, Xu Y, Liu G, Wu Y, Gong Z, and Yin Y
- Subjects
- Animals, Female, Humans, Male, Rats, Sex Characteristics, Gene Expression Profiling, Disease Models, Animal, Gene Ontology, Gene Expression Regulation, Gene Regulatory Networks, Sex Factors, Autophagy genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic pathology, Computational Biology methods, Signal Transduction, Rats, Inbred SHR
- Abstract
There is a paucity of research examining the molecular mechanisms underlying sex differences of clinical phenotypes and the prognosis in hypertrophic cardiomyopathy (HCM). The dataset GSE36961 was retrieved from Gene Expression Omnibus (GEO) database and comprehensive bioinformatics was employed to identify the core genes linked to sex differences in HCM patients. Additionally, gene set enrichment analysis (GSEA) was conducted to detect downstream signalling pathways. Furthermore, experimental validation was carried out using hearts from spontaneously hypertensive rats (SHRs). A comprehensive analysis revealed the identification of 208 differentially expressed genes (DEGs) in female patients with HCM with a notable downregulation of seven core genes. Notably, there were sex differences in the expression of ras dexamethasone-induced protein 1 (RASD1) and myosin 6 (MYH6) in HCM. Gene ontology (GO) analysis and GSEA demonstrated an enrichment of autophagy-related processes in disease progression in HCM females. Specifically, spearman's correlation analysis revealed a positive correlation between nicotinamide phosphoribosyl transferase (NAMPT) and RASD1 levels, particularly among female patients (R = 0.569, p < 0.001). Additionally, animal models confirmed that cardiac hypertrophy was more pronounced in SHR females compared to males. SHR females exhibited lower mRNA and protein expressions of RASD1 and NAMPT, which were associated with impaired autophagy. In this study, bioinformatics and validation using external data sets and animal models of left ventricular hypertrophy suggested that the RASD1/NAMPT axis is potentially a crucial mechanism underlying the elevated risk of cardiovascular disorders in HCM females, also pointing potentially prognostic biomarkers., (© 2024 The Author(s). Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
10. Twin Phenomena of Hypertrophic Cardiomyopathy: A Reported Case Series.
- Author
-
Zeng JT, Zhang YA, Ma TY, Huang K, Lu SJ, Zhong JH, and Li JJ
- Subjects
- Humans, Case Reports as Topic, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic genetics, Diseases in Twins genetics, Diseases in Twins diagnosis
- Abstract
Hypertrophic cardiomyopathy (HCM) is a prevalent genetic cardiovascular disease characterized by asymmetric thickening of the left ventricular wall, frequently occurring in families predisposed genetically. While HCM in twins is rare, it presents a unique opportunity to explore the disease's genetic and epigenetic underpinnings due to the phenotypic heterogeneity observed even among genetically identical individuals. This review collates and analyzes global clinical studies that focus on the twin phenomena in HCM. It explores the genetic foundations of HCM, examines the influence of environmental and epigenetic factors on disease expression, and emphasizes the crucial role of genetic screening in the early and differential diagnosis of HCM. By focusing on twin cases in HCM, this review aims to enhance our understanding of HCM's complex genetic background, which could lead to more personalized approaches in the management and treatment of this condition, thus drawing significant interest from researchers and clinicians alike.
- Published
- 2024
- Full Text
- View/download PDF
11. Evaluation of potential novel biomarkers for feline hypertrophic cardiomyopathy.
- Author
-
Chong A, Joshua J, Raheb S, Pires A, Colpitts M, Caswell JL, and Fonfara S
- Subjects
- Animals, Cats, Male, Female, Wnt-5a Protein genetics, Wnt-5a Protein blood, Interleukin-18 blood, Interleukin-18 genetics, Insulin-Like Growth Factor Binding Protein 2 blood, Insulin-Like Growth Factor Binding Protein 2 genetics, Enzyme-Linked Immunosorbent Assay veterinary, Wnt Proteins genetics, Glycogen Phosphorylase genetics, Glycogen Phosphorylase blood, Reverse Transcriptase Polymerase Chain Reaction veterinary, Cat Diseases blood, Cat Diseases diagnosis, Cat Diseases genetics, Cardiomyopathy, Hypertrophic veterinary, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic blood, Cardiomyopathy, Hypertrophic diagnosis, Biomarkers blood
- Abstract
Hypertrophic cardiomyopathy (HCM) is the most common cardiomyopathy in cats. The diagnosis can be difficult, requiring advanced echocardiographic skills. Additionally, circulating biomarkers (N-terminal pro-B type natriuretic peptide and cardiac troponin I) have several limitations when used for HCM screening. In previous work, we identified interleukin 18 (IL-18), insulin-like growth factor binding protein 2 (IGFBP-2), brain-type glycogen phosphorylase B (PYGB), and WNT Family Member 5 A (WNT5A) as myocardial genes that show significant differential expression between cats with HCM and healthy cats. The products of these genes are released into the circulation, and we hypothesized that IL-18, IGFBP-2, PYGB, and WNT5A serum RNA and protein concentrations differ between healthy cats, cats with subclinical HCM, and those with HCM and congestive heart failure (HCM + CHF). Reverse transcriptase quantitative polymerase chain reaction (RTqPCR) and enzyme-linked immunosorbent assay (ELISA) were applied to evaluate gene and protein expression, respectively, in the serum of eight healthy controls, eight cats with subclinical HCM, and six cats with HCM + CHF. Serum IGFBP-2 RNA concentrations were significantly different among groups and were highest in cats with subclinical HCM. Compared to healthy controls, serum IL-18 and WNT5A gene expression were significantly higher in cats with HCM + CHF, and WNT5A was higher in cats with subclinical HCM. No differences were observed for PYGB. These results indicate that further investigation via large scale clinical studies for IGFBP-2, WNT5A, and IL-18 may be valuable in diagnosing and staging feline HCM., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Crown Copyright © 2024. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Evaluation of potential links between phenotypic features and genetic variants in left ventricular outflow tract obstruction in hypertrophic cardiomyopathy using cardiovascular magnetic resonance imaging.
- Author
-
Güven B, Can TS, Deniz MF, Geçit MH, Geylan NA, Sinan ÜY, Oktay V, and Ersanlı MK
- Subjects
- Humans, Female, Male, Middle Aged, Cross-Sectional Studies, Aged, Adult, Genetic Association Studies, Risk Factors, Mitral Valve diagnostic imaging, Mitral Valve physiopathology, Ventricular Outflow Obstruction, Left, Ventricular Outflow Obstruction diagnostic imaging, Ventricular Outflow Obstruction genetics, Ventricular Outflow Obstruction physiopathology, Ventricular Outflow Obstruction etiology, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnostic imaging, Cardiomyopathy, Hypertrophic physiopathology, Phenotype, Predictive Value of Tests, Genetic Predisposition to Disease, Magnetic Resonance Imaging, Cine, Ventricular Function, Left
- Abstract
This study aimed to identify the phenotypic features contributing to the development of left ventricular outflow tract obstruction (LVOTO) in patients with hypertrophic cardiomyopathy (HCM) and to evaluate the genotype‒phenotype relationship. This cross-sectional study included 96 patients diagnosed with HCM (mean age: 56.9 ± 13.5 years, 32.3% female). The patients were divided into hypertrophic nonobstructive cardiomyopathy (HNCM; n = 60) and hypertrophic obstructive cardiomyopathy (HOCM; n = 36) groups. All patients underwent CMR. Patients (n = 77) who had previously provided formal approval underwent a genetic examination that included 18 genes. The anterior mitral leaflet (AML) length/LVOT diameter ratio, posterior mitral leaflet (PML) length/LVOT diameter ratio, and anterolateral papillary muscle (AL-PM) mobility were associated with LVOTO, independent of the basal IVS thickness, abnormal chordal attachment, and bifid PM. An AML length/LVOT diameter ratio of ≥ 2.30, a PML length/LVOT diameter ratio of ≥ 1.83, and an AL-PM mobility of ≥ 57.7% were predictors of LVOTO, with good sensitivity and specificity. Positive variants (VUS, LP, and P) were detected in 37.7% (29 of 77) of the patients who underwent genetic testing. The LP/P variant was detected in 20.8% (16 of 77) of patients. Three groups (variant-negative, VUS, and LP/P groups) had significant differences in the LVOT diameter (median 14, 12, and 10 mm, respectively; p = 0.021), AML length (mean 25.3, 26.5, and 27.5 mm, respectively; p = 0.029), AML length/LVOT diameter ratio (median 1.74, 2.33, and 2.85, respectively; p = 0.006), PML length/LVOT diameter ratio (median 1.29, 1.82, and 2.10, respectively; p = 0.045), and abnormal chordal attachment (6.3%, not observed, and 31.3%, respectively; p = 0.009). The AML length/LVOT diameter ratio, PML length/LVOT diameter ratio, and AL-PM mobility were associated with LVOTO. In addition, genetic testing results may provide information regarding the phenotypic expression of patients with HCM., Competing Interests: Declarations Ethical approval This study was approved by the Istanbul University-Cerrahpasa Clinical Research Ethics Committee (approval date: 04/07/2023, decision no: 257). Informed consent Written informed consent was obtained from all participants. Authors note This study is derived from the cardiology specialization thesis of the corresponding author Barış Güven. Conflict of interest The authors declare that there are no conflicts of interest regarding the publication of this article., (© 2024. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2024
- Full Text
- View/download PDF
13. A multi-omics atlas of sex-specific differences in obstructive hypertrophic cardiomyopathy.
- Author
-
Garmany R, Dasari S, Bos JM, Kim ET, Gluscevic M, Martinez KA, Tester DJ, Dos Remedios C, Maleszewski JJ, Dearani JA, Ommen SR, Geske JB, Giudicessi JR, and Ackerman MJ
- Subjects
- Humans, Male, Female, Middle Aged, Gene Expression Profiling, Phosphoproteins metabolism, Phosphoproteins genetics, Phosphorylation, Sex Factors, Gene Expression Regulation, Adult, Myocardium metabolism, Multiomics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Proteomics methods, Transcriptome genetics, Proteome metabolism, Sex Characteristics
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is a common genetic heart disease. Women with HCM tend to have a later onset but more severe disease course. However, the underlying pathobiological mechanisms for these differences remain unknown., Methods: Myectomy samples from 97 patients (53 males/44 females) with symptomatic obstructive HCM and 23 control cardiac tissues were included in this study. RNA-sequencing was performed on all samples. Mass spectrometry-based proteomics and phosphoproteomics was performed on a representative subset of samples., Results: The transcriptome, proteome, and phosphoproteome was similar between sexes and did not separate on PCA plotting. Overall, there were 482 differentially expressed genes (DEGs) between control females and control males while there were only 53 DEGs between HCM females and HCM males. There were 1983 DEGs between HCM females and control females compared to 1064 DEGs between HCM males and control males. Additionally, there was increased transcriptional downregulation of hypertrophy pathways in HCM females and in HCM males. HCM females had 119 differentially expressed proteins compared to control females while HCM males only had 27 compared to control males. Finally, the phosphoproteome showed females had 341 differentially phosphorylated proteins (DPPs) compared to controls while males only had 184. Interestingly, there was hypophosphorylation and inactivation of hypertrophy pathways in females but hyperphosphorylation and activation in males., Conclusion: There are subtle, but biologically relevant differences in the multi-omics profile of HCM. This study provides the most comprehensive atlas of sex-specific differences in the transcriptome, proteome, and phosphoproteome present at the time of surgical myectomy for obstructive HCM., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Integrative analysis of transcriptome, DNA methylome, and chromatin accessibility reveals candidate therapeutic targets in hypertrophic cardiomyopathy.
- Author
-
Gao J, Liu M, Lu M, Zheng Y, Wang Y, Yang J, Xue X, Liu Y, Tang F, Wang S, Song L, Wen L, and Wang J
- Subjects
- Humans, Animals, Mice, Early Growth Response Protein 1 genetics, Early Growth Response Protein 1 metabolism, Male, Epigenome, Nucleosomes metabolism, Nucleosomes genetics, Female, Middle Aged, Disease Models, Animal, Adult, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, DNA Methylation, Transcriptome, Chromatin metabolism, Chromatin genetics
- Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease and is characterized by primary left ventricular hypertrophy usually caused by mutations in sarcomere genes. The mechanism underlying cardiac remodeling in HCM remains incompletely understood. An investigation of HCM through integrative analysis at multi-omics levels will be helpful for treating HCM. DNA methylation and chromatin accessibility, as well as gene expression, were assessed by nucleosome occupancy and methylome sequencing (NOMe-seq) and RNA-seq, respectively, using the cardiac tissues of HCM patients. Compared with those of the controls, the transcriptome, DNA methylome, and chromatin accessibility of the HCM myocardium showed multifaceted differences. At the transcriptome level, HCM hearts returned to the fetal gene program through decreased sarcomeric and metabolic gene expression and increased extracellular matrix gene expression. In the DNA methylome, hypermethylated and hypomethylated differentially methylated regions were identified in HCM. At the chromatin accessibility level, HCM hearts showed changes in different genome elements. Several transcription factors, including SP1 and EGR1, exhibited a fetal-like pattern of binding motifs in nucleosome-depleted regions in HCM. In particular, the inhibition of SP1 or EGR1 in an HCM mouse model harboring sarcomere mutations markedly alleviated the HCM phenotype of the mutant mice and reversed fetal gene reprogramming. Overall, this study not only provides a high-precision multi-omics map of HCM heart tissue but also sheds light on the therapeutic strategy by intervening in the fetal gene reprogramming in HCM., (© The Author(s) 2024. Published by Oxford University Press on behalf of Higher Education Press.)
- Published
- 2024
- Full Text
- View/download PDF
15. Myocardial contractility characteristics of hypertrophic cardiomyopathy patients with and without sarcomere mutation.
- Author
-
Zhang J, Li J, Wang B, Wang J, Hu R, Shan B, Han Y, Zhao X, Zhang J, Zhang Y, Ta S, and Liu L
- Subjects
- Humans, Male, Female, Retrospective Studies, Middle Aged, Adult, Ventricular Function, Left physiology, Echocardiography, Three-Dimensional, Genotype, Sarcomeres genetics, Myocardial Contraction physiology, Myocardial Contraction genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic physiopathology, Cardiomyopathy, Hypertrophic diagnosis, Mutation
- Abstract
Hypertrophic cardiomyopathy (HCM) patients with sarcomere mutations have an increased risk of heart failure and left ventricular (LV) systolic dysfunction. We hypothesize that sarcomere mutation carriers have abnormal myocardial contractility before LV dysfunction. Therefore, we aimed to associate myocardial contractility with identified sarcomere mutations and predict genotyped HCM patients with sarcomere mutation by three-dimensional speckle tracking imaging (3D-STI). A retrospective analysis of 117 HCM patients identified 32 genotype-positive (G +) and 85 genotype-negative (G-) patients. Genotype-positive patients had higher globe circumferential strain (GCS), globe longitudinal strain (GLS), and globe radial strain (GRS) (p < 0.05), and multivariate logistic regression revealed that these variables were associated with a positive genetic status (p < 0.05). After the propensity matches other possible influencing factors, we developed three models, named Model GCS, Model GLS, and Model GRS, which could identified genotype-positive HCM patients with excellent performance (AUC of 0.855, 0.833, and 0.870 respectively, all p < 0.001). Genotype-positive HCM patients show a higher myocardial hyper-contractility status than patients without sarcomere mutations. When combined with clinical and echocardiographic markers, the 3D-STI parameters can effectively identify the likelihood of genotype-positive HCM., (© 2024. Springer Nature Japan KK, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
16. A rare form of LIM domain-binding protein 3 (LDB3) mutation causes hypertrophic cardiomyopathy and myofibrillar myopathy type 4.
- Author
-
Svaguša T, Sedlić F, Županić S, Manola Š, Bakoš M, Mirošević V, and Livun A
- Subjects
- Humans, Male, Adult, Female, Genetic Predisposition to Disease, Adaptor Proteins, Signal Transducing, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, LIM Domain Proteins genetics, Mutation, Myopathies, Structural, Congenital genetics, Myopathies, Structural, Congenital pathology
- Abstract
Polymorphisms in LDB3 gene can cause various forms of cardiomyopathy and myofibrillar myopathy 4 (MM4). Patient described in this study presented with a hypertrophic cardiomyopathy (HCM) and distal myopathy suggestive of myofibrillar myopathy 4. Genetic analysis using the TruSight Cardio Sequencing Kit (Illumina) revealed suspected LDB3 variant (c.1435G>A, p.(Gly479Arg)). This is the first case in which polymorphism in LDB3 gene is likely responsible for MM4 and HCM in the same patient., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
17. Novel association of LBX1 mutation with tetralogy of Fallot and hypertrophic cardiomyopathy: implications for cardiac development.
- Author
-
Cheng P, Wang G, Song Y, and An Y
- Subjects
- Child, Preschool, Female, Humans, Homeodomain Proteins genetics, Transcription Factors genetics, Cardiomyopathy, Hypertrophic genetics, Mutation, Tetralogy of Fallot genetics
- Abstract
Background: Tetralogy of Fallot (TOF) and hypertrophic cardiomyopathy (HCM) are common types of congenital heart disease with unique pathophysiologic features. Mutations in LBX1, a key regulator of muscle precursor cell migration, may disrupt these critical developmental processes, resulting in severe developmental abnormalities., Case Presentation: This case reports on a 4-year-old girl diagnosed with both TOF and HCM. Genetic analysis revealed iUPI-F (uniparental disomy) on the entire chromosome 10, with a c.808G > A (p. Glu270Lys) pure mutation in the LBX1., Conclusion: This patient exhibited both TOF and HCM with mutations in the LBX1 gene, suggesting a potentially novel genetic link. This case study emphasizes the need for further studies on the function of the LBX1 gene in cardiac development and its potential impact on TOF and HCM., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
18. Arg92Leu-cTnT Alters the cTnC-cTnI Interface Disrupting PKA-Mediated Relaxation.
- Author
-
Lynn ML, Jimenez J, Castillo RL, Vasquez C, Klass MM, Baldo AP, Kim A, Gibson C, Murphy AM, and Tardiff JC
- Subjects
- Animals, Humans, Troponin C genetics, Troponin C metabolism, Molecular Dynamics Simulation, Mutation, Mice, Male, Cyclic AMP-Dependent Protein Kinases metabolism, Troponin I genetics, Troponin I metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic physiopathology, Troponin T genetics, Troponin T metabolism
- Abstract
Background: Impaired left ventricular relaxation, high filling pressures, and dysregulation of Ca
2+ homeostasis are common findings contributing to diastolic dysfunction in hypertrophic cardiomyopathy (HCM). Studies have shown that impaired relaxation is an early observation in the sarcomere-gene-positive preclinical HCM cohort, which suggests the potential involvement of myofilament regulators in relaxation. A molecular-level understanding of mechanism(s) at the level of the myofilament is lacking. We hypothesized that mutation-specific, allosterically mediated, changes to the cTnC (cardiac troponin C)-cTnI (cardiac troponin I) interface can account for the development of early-onset diastolic dysfunction via decreased PKA accessibility to cTnI., Methods: HCM mutations R92L-cTnT (cardiac troponin T; Arg92Leu) and Δ160E-cTnT (Glu160 deletion) were studied in vivo, in vitro, and in silico via 2-dimensional echocardiography, Western blotting, ex vivo hemodynamics, stopped-flow kinetics, time-resolved fluorescence resonance energy transfer, and molecular dynamics simulations., Results: The HCM-causative mutations R92L-cTnT and Δ160E-cTnT result in different time-of-onset diastolic dysfunction. R92L-cTnT demonstrated early-onset diastolic dysfunction accompanied by a localized decrease in phosphorylation of cTnI. Constitutive phosphorylation of cTnI (cTnI-D23 D24 ) was sufficient to recover diastolic function to non-Tg levels only for R92L-cTnT. Mutation-specific changes in Ca2+ dissociation rates associated with R92L-cTnT reconstituted with cTnI-D23 D24 led us to investigate potential involvement of structural changes in the cTnC-cTnI interface as an explanation for these observations. We probed the interface via time-resolved fluorescence resonance energy transfer revealing a repositioning of the N-terminus of cTnI, closer to cTnC, and concomitant decreases in distance distributions at sites flanking the PKA consensus sequence. Implementing time-resolved fluorescence resonance energy transfer distances as constraints into our atomistic model identified additional electrostatic interactions at the consensus sequence., Conclusions: These data show that the early diastolic dysfunction observed in a subset of HCM is attributable to allosterically mediated structural changes at the cTnC-cTnI interface that impair accessibility of PKA, thereby blunting β-adrenergic responsiveness and identifying a potential molecular target for therapeutic intervention., Competing Interests: None.- Published
- 2024
- Full Text
- View/download PDF
19. The D75N and P161S Mutations in the C0-C2 Fragment of cMyBP-C Associated with Hypertrophic Cardiomyopathy Disturb the Thin Filament Activation, Nucleotide Exchange in Myosin, and Actin-Myosin Interaction.
- Author
-
Kochurova AM, Beldiia EA, Nefedova VV, Yampolskaya DS, Koubassova NA, Kleymenov SY, Antonets JY, Ryabkova NS, Katrukha IA, Bershitsky SY, Matyushenko AM, Kopylova GV, and Shchepkin DV
- Subjects
- Actin Cytoskeleton metabolism, Actin Cytoskeleton genetics, Humans, Mutation, Protein Binding, Animals, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Carrier Proteins chemistry, Actins metabolism, Actins genetics, Myosins metabolism, Myosins genetics, Myosins chemistry
- Abstract
About half of the mutations that lead to hypertrophic cardiomyopathy (HCM) occur in the MYBPC3 gene. However, the molecular mechanisms of pathogenicity of point mutations in cardiac myosin-binding protein C (cMyBP-C) remain poorly understood. In this study, we examined the effects of the D75N and P161S substitutions in the C0 and C1 domains of cMyBP-C on the structural and functional properties of the C0-C1-m-C2 fragment (C0-C2). Differential scanning calorimetry revealed that these mutations disorder the tertiary structure of the C0-C2 molecule. Functionally, the D75N mutation reduced the maximum sliding velocity of regulated thin filaments in an in vitro motility assay, while the P161S mutation increased it. Both mutations significantly reduced the calcium sensitivity of the actin-myosin interaction and impaired thin filament activation by cross-bridges. D75N and P161S C0-C2 fragments substantially decreased the sliding velocity of the F-actin-tropomyosin filament. ADP dose-dependently reduced filament sliding velocity in the presence of WT and P161S fragments, but the velocity remained unchanged with the D75N fragment. We suppose that the D75N mutation alters nucleotide exchange kinetics by decreasing ADP affinity to the ATPase pocket and slowing the myosin cycle. Our molecular dynamics simulations mean that the D75N mutation affects myosin S1 function. Both mutations impair cardiac contractility by disrupting thin filament activation. The results offer new insights into the HCM pathogenesis caused by missense mutations in N-terminal domains of cMyBP-C, highlighting the distinct effects of D75N and P161S mutations on cardiac contractile function.
- Published
- 2024
- Full Text
- View/download PDF
20. Hypertrophic cardiomyopathy-associated mutations drive stromal activation via EGFR-mediated paracrine signaling.
- Author
-
Ewoldt JK, Wang MC, McLellan MA, Cloonan PE, Chopra A, Gorham J, Li L, DeLaughter DM, Gao X, Lee JH, Willcox JAL, Layton O, Luu RJ, Toepfer CN, Eyckmans J, Seidman CE, Seidman JG, and Chen CS
- Subjects
- Humans, Fibroblasts metabolism, Myosin Heavy Chains metabolism, Myosin Heavy Chains genetics, Fibrosis, Stromal Cells metabolism, Carrier Proteins metabolism, Carrier Proteins genetics, Cell Proliferation, Cardiac Myosins, Paracrine Communication, ErbB Receptors metabolism, ErbB Receptors genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic pathology, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells cytology, Mutation, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology
- Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by thickening of the left ventricular wall, diastolic dysfunction, and fibrosis, and is associated with mutations in genes encoding sarcomere proteins. While in vitro studies have used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to study HCM, these models have not examined the multicellular interactions involved in fibrosis. Using engineered cardiac microtissues (CMTs) composed of HCM-causing MYH7 -variant hiPSC-CMs and wild-type fibroblasts, we observed cell-cell cross-talk leading to increased collagen deposition, tissue stiffening, and decreased contractility dependent on fibroblast proliferation. hiPSC-CM conditioned media and single-nucleus RNA sequencing data suggested that fibroblast proliferation is mediated by paracrine signals from MYH7 -variant cardiomyocytes. Furthermore, inhibiting epidermal growth factor receptor tyrosine kinase with erlotinib hydrochloride attenuated stromal activation. Last, HCM-causing MYBPC3 -variant CMTs also demonstrated increased stromal activation and reduced contractility, but with distinct characteristics. Together, these findings establish a paracrine-mediated cross-talk potentially responsible for fibrotic changes observed in HCM.
- Published
- 2024
- Full Text
- View/download PDF
21. Phenotypic expression, genotypic profiling and clinical outcomes of infantile hypertrophic cardiomyopathy: a retrospective study.
- Author
-
Ahamed H, Varghese S, Gutajahr G, Vaidyanathan B, Kappanayil M, Sasikumar N, Kumar S, Hari A, Krishnakumar M, and Kumar RK
- Subjects
- Humans, Male, Female, Infant, Retrospective Studies, Electrocardiography, India epidemiology, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Phenotype, Echocardiography
- Abstract
Background: Infantile hypertrophic cardiomyopathy (HCM) is a heterogeneous disorder. Apart from registries in high-income nations, there is a shortage of data on the aetiological basis of infantile HCM in low- and middle-income nations. This study attempts to characterise the phenotypic expression, genetic architecture and short-term clinical outcomes of infantile HCM from a South Asian tertiary referral centre., Methods: This study includes all infants from the Amrita HCM cohort between January 2011 and July 2021. Clinical history, ECG, echocardiographic data, and genetic analyses were evaluated., Results: 34 patients with infantile HCM were diagnosed at a median age of 3.7 months (IQR 1-6 months). Underlying aetiologies were RASopathy (n=13; 38%), non-syndromic (n=12; 35%) and inborn errors of metabolism (n=9; 27%). Genetic analysis was done in 20 patients (59%) with a yield of 90%. Clinical presentation included failure to thrive (n=29; 85%), dyspnoea on exertion (n=23; 68%) and clinical heart failure (n=24; 71%). Echo showed concentric left ventricular hypertrophy in 22 patients (65%), obstructive HCM in 11 patients (32%) and left ventricular systolic dysfunction in 6 patients (18%). The mortality rate was 10.0 deaths per 100 patient years over a median follow-up period of 3.1 years. The main risk markers for mortality were the age at diagnosis, gender and concentric Left ventricular hypertrophy., Conclusions: This cohort demonstrates the morphological, functional and genetical heterogeneity of infantile HCM, enunciating the need for integration of cardiology, metabolic and genetic services to achieve optimum outcomes in these patients., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
22. Chronic Activation of Tubulin Tyrosination Improves Heart Function.
- Author
-
Pietsch N, Chen CY, Kupsch S, Bacmeister L, Geertz B, Herrera-Rivero M, Siebels B, Voß H, Krämer E, Braren I, Westermann D, Schlüter H, Mearini G, Schlossarek S, van der Velden J, Caporizzo MA, Lindner D, Prosser BL, and Carrier L
- Subjects
- Animals, Humans, Mice, Tyrosine metabolism, Mice, Inbred C57BL, Mice, Transgenic, Cells, Cultured, Induced Pluripotent Stem Cells metabolism, Male, Myocardial Contraction, Tubulin metabolism, Myocytes, Cardiac metabolism, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic physiopathology, Cardiomyopathy, Hypertrophic pathology
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is the most common cardiac genetic disorder caused by sarcomeric gene variants and associated with left ventricular hypertrophy and diastolic dysfunction. The role of the microtubule network has recently gained interest with the findings that microtubule detyrosination (dTyr-MT) is markedly elevated in heart failure. Acute reduction of dTyr-MT by inhibition of the detyrosinase (VASH [vasohibin]/SVBP [small VASH-binding protein] complex) or activation of the tyrosinase (TTL [tubulin tyrosine ligase]) markedly improved contractility and reduced stiffness in human failing cardiomyocytes and thus posed a new perspective for HCM treatment. In this study, we tested the impact of chronic tubulin tyrosination in an HCM mouse model ( Mybpc3 knock-in), in human HCM cardiomyocytes, and in SVBP-deficient human engineered heart tissues (EHTs)., Methods: Adeno-associated virus serotype 9-mediated TTL transfer was applied in neonatal wild-type rodents, in 3-week-old knock-in mice, and in HCM human induced pluripotent stem cell-derived cardiomyocytes., Results: We show (1) TTL for 6 weeks dose dependently reduced dTyr-MT and improved contractility without affecting cytosolic calcium transients in wild-type cardiomyocytes; (2) TTL for 12 weeks reduced the abundance of dTyr-MT in the myocardium, improved diastolic filling, compliance, cardiac output, and stroke volume in knock-in mice; (3) TTL for 10 days normalized cell area in HCM human induced pluripotent stem cell-derived cardiomyocytes; (4) TTL overexpression activated transcription of tubulins and other cytoskeleton components but did not significantly impact the proteome in knock-in mice; (5) SVBP-deficient EHTs exhibited reduced dTyr-MT levels, higher force, and faster relaxation than TTL-deficient and wild-type EHTs. RNA sequencing and mass spectrometry analysis revealed distinct enrichment of cardiomyocyte components and pathways in SVBP-deficient versus TTL-deficient EHTs., Conclusions: This study provides the first proof of concept that chronic activation of tubulin tyrosination in HCM mice and in human EHTs improves heart function and holds promise for targeting the nonsarcomeric cytoskeleton in heart disease., Competing Interests: L. Carrier is a member of Scientific Advisory Board of and has shares in the company DiNAQOR AG (https://www.dinaqor.com/). B.L. Prosser is an inventor on a pending US patent application number 15/959181 for “Composition and Methods for Improving Heart Function and Treating Heart Failure.” The other authors report no conflicts.
- Published
- 2024
- Full Text
- View/download PDF
23. CRISPR/Cas9 gene editing in induced pluripotent stem cells to investigate the feline hypertrophic cardiomyopathy causing MYBPC3/R820W mutation.
- Author
-
Dutton LC, Dudhia J, Guest DJ, and Connolly DJ
- Subjects
- Animals, Cats, Humans, Cell Differentiation, Cat Diseases genetics, Cat Diseases pathology, Induced Pluripotent Stem Cells metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Cardiomyopathy, Hypertrophic veterinary, CRISPR-Cas Systems, Gene Editing, Carrier Proteins genetics, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Mutation
- Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in domestic cats, often leading to congestive heart failure and death, with current treatment strategies unable to reverse or prevent progression of the disease. The underlying pathological processes driving HCM remain unclear, which hinders novel drug discovery. The aim of this study was to generate a cellular model of the feline HCM-causing MYBPC3 mutation R820W. Using CRISPR/Cas9 gene editing we introduced the R820W mutation into a human induced pluripotent stem cell (iPSC) line. We differentiated both homozygous mutant clones and isogenic control clones to cardiomyocytes (iPSC-CMs). Protein quantification indicated that haploinsufficiency is not the disease mechanism of the mutation. Homozygous mutant iPSC-CMs had a larger cell area than isogenic controls, with the sarcomere structure and incorporation of cMyBP-C appearing similar between mutant and control iPSC-CMs. Contraction kinetic analysis indicated that homozygous iPSC-CMs have impaired relaxation and are hypocontractile compared to isogenic control iPSC-CMs. In summary, we demonstrate successful generation of an iPSC model of a feline MYBPC3 mutation, with the cellular model recapitulating aspects of HCM including cellular hypertrophy and impaired relaxation kinetics. We anticipate that further study of this model will lead to improved understanding of the disease-causing molecular mechanism, ultimately leading to novel drug discovery., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Dutton et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
24. Cardiac NAD + depletion in mice promotes hypertrophic cardiomyopathy and arrhythmias prior to impaired bioenergetics.
- Author
-
Doan KV, Luongo TS, Ts'olo TT, Lee WD, Frederick DW, Mukherjee S, Adzika GK, Perry CE, Gaspar RB, Walker N, Blair MC, Bye N, Davis JG, Holman CD, Chu Q, Wang L, Rabinowitz JD, Kelly DP, Cappola TP, Margulies KB, and Baur JA
- Subjects
- Animals, Disease Models, Animal, Cytokines metabolism, Mice, Knockout, Mice, Inbred C57BL, Pyridinium Compounds, Male, Death, Sudden, Cardiac etiology, Death, Sudden, Cardiac pathology, Mice, Niacinamide analogs & derivatives, Niacinamide pharmacology, Niacinamide therapeutic use, Niacinamide metabolism, Electrocardiography, Nicotinamide Phosphoribosyltransferase metabolism, Nicotinamide Phosphoribosyltransferase genetics, NAD metabolism, Energy Metabolism, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Arrhythmias, Cardiac metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology
- Abstract
Nicotinamide adenine dinucleotide (NAD
+ ) is an essential co-factor in metabolic reactions and co-substrate for signaling enzymes. Failing human hearts display decreased expression of the major NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (Nampt) and lower NAD+ levels, and supplementation with NAD+ precursors is protective in preclinical models. Here we show that Nampt loss in adult cardiomyocytes caused depletion of NAD+ along with marked metabolic derangements, hypertrophic remodeling and sudden cardiac deaths, despite unchanged ejection fraction, endurance and mitochondrial respiratory capacity. These effects were directly attributable to NAD+ loss as all were ameliorated by restoring cardiac NAD+ levels with the NAD+ precursor nicotinamide riboside (NR). Electrocardiograms revealed that loss of myocardial Nampt caused a shortening of QT intervals with spontaneous lethal arrhythmias causing sudden cardiac death. Thus, changes in NAD+ concentration can have a profound influence on cardiac physiology even at levels sufficient to maintain energetics., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
25. Identification of a novel likely pathogenic TPM1 variant linked to hypertrophic cardiomyopathy in a family with sudden cardiac death.
- Author
-
Azimi A, Soveizi M, Salmanipour A, Mozafarybazargany M, Ghaffari Jolfayi A, Maleki M, and Kalayinia S
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Exome Sequencing, Magnetic Resonance Imaging, Cine methods, Echocardiography, Phenotype, Electrocardiography, Iran epidemiology, Mutation, Missense, DNA genetics, Tropomyosin genetics, Death, Sudden, Cardiac etiology, Pedigree, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic complications, Cardiomyopathy, Hypertrophic diagnosis
- Abstract
Aims: Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic cardiac disorder characterized by unexplained left ventricular hypertrophy. It can cause a wide spectrum of clinical manifestations, ranging from asymptomatic to heart failure and sudden cardiac death (SCD). Approximately half of HCM cases are caused by variants in sarcomeric proteins, including α-tropomyosin (TPM1). In this study, we aimed to characterize the clinical and molecular phenotype of HCM in an Iranian pedigree with SCD., Methods and Results: The proband and available family members underwent comprehensive clinical evaluations, including echocardiography, cardiac magnetic resonance (CMR) imaging and electrocardiography (ECG). Whole-exome sequencing (WES) was performed in all available family members to identify the causal variant, which was validated, and segregation analysis was conducted via Sanger sequencing. WES identified a novel missense variant, c.761A>G:p.D254G (NM_001018005.2), in the TPM1 gene, in the proband, his father and one of his sisters. Bioinformatic analysis predicted it to be likely pathogenic. Clinical features in affected individuals were consistent with HCM., Conclusions: The identification of a novel TPM1 variant in a family with HCM and SCD underscores the critical role of genetic screening in at-risk families. Early detection of pathogenic variants can facilitate timely intervention and management, potentially reducing the risk of SCD in individuals with HCM., (© 2024 The Author(s). ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
26. Clonal haematopoiesis is associated with major adverse cardiovascular events in patients with hypertrophic cardiomyopathy.
- Author
-
Scolari FL, Brahmbhatt D, Abelson S, Lee D, Kim RH, Pedarzadeh A, Sakhnini A, Adler A, Chan RH, Dick JE, Rakowski H, and Billia F
- Subjects
- Humans, Male, Female, Middle Aged, DNA Methyltransferase 3A, Repressor Proteins genetics, Proto-Oncogene Proteins genetics, DNA (Cytosine-5-)-Methyltransferases genetics, DNA-Binding Proteins genetics, Troponin I blood, Aged, Echocardiography, Phenotype, Cytokines genetics, Natriuretic Peptide, Brain blood, Mutation, Adult, Risk Factors, Heart Arrest etiology, Dioxygenases, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic complications, Cardiomyopathy, Hypertrophic physiopathology, Clonal Hematopoiesis genetics
- Abstract
Aims: The heterogeneous phenotype of hypertrophic cardiomyopathy (HCM) is still not fully understood. Clonal haematopoiesis (CH) is emerging as a cardiovascular risk factor potentially associated with adverse clinical events. The prevalence, phenotype and outcomes related to CH in HCM patients were evaluated., Methods and Results: Patients with HCM and available biospecimens from the Peter Munk Cardiac Centre Cardiovascular Biobank were subjected to targeted sequencing for 35 myeloid genes associated with CH. CH prevalence, clinical characteristics, morphological phenotypes assessed by echocardiogram and cardiac magnetic resonance and outcomes were assessed. All patients were evaluated for a 71-plex cytokines/chemokines, troponin I and B-type natriuretic peptide analysis. Major adverse cardiovascular events (MACE) were defined as appropriate implantable cardioverter-defibrillator shock, stroke, cardiac arrest, orthotopic heart transplant and death. Among the 799 patients, CH was found in 183 (22.9%) HCM patients with sarcomeric germline mutations. HCM patients with CH were more symptomatic and with a higher burden of fibrosis than those without CH. CH was associated with MACE in those HCM patients with sarcomeric germline mutations (adjusted hazard ratio [HR] 6.89, 95% confidence interval [CI] 1.78-26.6; p = 0.005), with the highest risk among those that had DNMT3A, TET2 and ASXL1 mutations (adjusted HR 5.76, 95% CI 1.51-21.94; p = 0.010). Several cytokines (IL-1ra, IL-6, IL-17F, TGFα, CCL21, CCL1, CCL8, and CCL17), and troponin I were upregulated in gene-positive HCM patients with CH., Conclusions: These results indicate that CH in patients with HCM is associated with worse clinical outcomes. In the absence of CH, gene-positive patients with HCM have lower rates of MACE., (© 2024 The Author(s). European Journal of Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
27. Feline hypertrophic cardiomyopathy: Does the microRNA-mRNA regulatory network contribute to heart sarcomeric protein remodelling?
- Author
-
Guelfi G, Venanzi N, Capaccia C, Stefanetti V, Brachelente C, Sforna M, Porciello F, and Lepri E
- Subjects
- Animals, Cats, Myocardium metabolism, Myocardium pathology, Male, Gene Expression Regulation, Female, MicroRNAs genetics, MicroRNAs metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Sarcomeres metabolism, Sarcomeres genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Cat Diseases genetics, Cat Diseases metabolism, Gene Regulatory Networks
- Abstract
Feline primary hypertrophic cardiomyopathy (HCM) is an intrinsic myocardial disease characterized by concentric hypertrophy of the left ventricle. In the present study, we investigated the microRNA-mRNA regulatory network in feline myocardial tissue affected by primary (HCMI) and secondary HCM (HCMII). MRNA expression levels of sarcomeric genes, including, TNNT2, TNNI3, MYH7, MYBPC3, TPM1 and ACTC1 were assessed in the FFPE myocardial tissues. FFPE tissues from healthy cats were sequenced by the NGS, to explore, in the entire non-deposited miRNome, the expression level of microRNAs targeting the complementary sequences of selected sarcomeric mRNAs. The sarcomeric genes TNNT2, MYH7, MYBPC3 and TPM1 showed a statistically significant upregulation in HCMI compared to HCMII (p < .01), except ACTC1 which was downregulated (p < .01); TNNI3 showed no statistically significant difference. In HCMII miR-122-5p, miR-338-3p, miR-484, miR-370-3p, miR-92b-3p, miR-375 and miR-370-3p showed a significant upregulation (p < .01) compared to control. The exception was miR-30a-5p which showed downregulation. Worthy of note is the 4-fold higher expression of miR-370-3p, a key regulator of MYBPC3, in HMCI compared to HMCII. This research does not solve the aetiological mystery of HCM, but it may help to find a way to help diagnose and define the prognosis of HCM in cats., (© 2024 The Author(s). International Journal of Experimental Pathology published by John Wiley & Sons Ltd on behalf of Company of the International Journal of Experimental Pathology (CIJEP).)
- Published
- 2024
- Full Text
- View/download PDF
28. Reassessing the unifying hypothesis for hypercontractility caused by myosin mutations in hypertrophic cardiomyopathy.
- Author
-
Spudich JA, Nandwani N, Robert-Paganin J, Houdusse A, and Ruppel KM
- Subjects
- Humans, Myosins genetics, Myosins metabolism, Myocardial Contraction genetics, Animals, Cardiomyopathy, Hypertrophic genetics, Mutation
- Published
- 2024
- Full Text
- View/download PDF
29. Re-examining family history of sudden death as a risk marker in hypertrophic cardiomyopathy.
- Author
-
Siontis KC, Ommen SR, Maron MS, and Maron BJ
- Subjects
- Humans, Risk Factors, Medical History Taking, Genetic Predisposition to Disease, Death, Sudden, Cardiac etiology, Death, Sudden, Cardiac epidemiology, Death, Sudden, Cardiac prevention & control, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic complications, Cardiomyopathy, Hypertrophic physiopathology
- Published
- 2024
- Full Text
- View/download PDF
30. The W792R HCM missense mutation in the C6 domain of cardiac myosin binding protein-C increases contractility in neonatal mouse myocardium.
- Author
-
Mertens J, De Lange WJ, Farrell ET, Harbaugh EC, Gauchan A, Fitzsimons DP, Moss RL, and Ralphe JC
- Subjects
- Animals, Mice, Protein Domains, Sarcomeres metabolism, Calcium metabolism, Disease Models, Animal, Gene Knock-In Techniques, Mutation, Missense, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic physiopathology, Cardiomyopathy, Hypertrophic pathology, Myocardial Contraction genetics, Carrier Proteins genetics, Carrier Proteins metabolism, Animals, Newborn, Myocardium metabolism
- Abstract
Missense mutations in cardiac myosin binding protein C (cMyBP-C) are known to cause hypertrophic cardiomyopathy (HCM). The W792R mutation in the C6 domain of cMyBP-C causes severe, early onset HCM in humans, yet its impact on the function of cMyBP-C and the mechanism through which it causes disease remain unknown. To fully characterize the effect of the W792R mutation on cardiac morphology and function in vivo, we generated a murine knock-in model. We crossed heterozygous W792R
WR mice to produce homozygous mutant W792RRR , heterozygous W792RWR , and control W792RWW mice. W792RRR mice present with cardiac hypertrophy, myofibrillar disarray and fibrosis by postnatal day 10 (PND10), and do not survive past PND21. Full-length cMyBP-C is present at similar levels in W792RWW , W792RWR and W792RRR mice and is properly incorporated into the sarcomere. Heterozygous W792RWR mice displayed normal heart morphology and contractility. Permeabilized myocardium from PND10 W792RRR mice showed increased Ca2+ sensitivity, accelerated cross-bridge cycling kinetics, decreased cooperativity in the activation of force, and increased expression of hypertrophy-related genes. In silico modeling suggests that the W792R mutation destabilizes the fold of the C6 domain and increases torsion in the C5-C7 region, possibly impacting regulatory interactions of cMyBP-C with myosin and actin. Based on the data presented here, we propose a model in which mutant W792R cMyBP-C preferentially forms Ca2+ sensitizing interactions with actin, rather than inhibitory interactions with myosin. The W792R-cMyBP-C mouse model provides mechanistic insights into the pathology of this mutation and may provide a mechanism by which other central domain missense mutations in cMyBP-C may alter contractility, leading to HCM., (Copyright © 2024. Published by Elsevier Ltd.)- Published
- 2024
- Full Text
- View/download PDF
31. Deciphering metabolomics and lipidomics landscape in zebrafish hypertrophic cardiomyopathy model.
- Author
-
Jacob S, Abuarja T, Shaath R, Hasan W, Balayya S, Abdelrahman D, Almana K, Afreen H, Hani A, Nomikos M, Fakhro K, Elrayess MA, and Da'as SI
- Subjects
- Animals, Lipid Metabolism, Fatty Acids metabolism, Metabolic Networks and Pathways, Metabolome, Zebrafish metabolism, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Metabolomics methods, Lipidomics methods, Disease Models, Animal
- Abstract
To elucidate the lipidomic and metabolomic alterations associated with hypertrophic cardiomyopathy (HCM) pathogenesis, we utilized cmybpc3-/- zebrafish model. Fatty acid profiling revealed variability of 10 fatty acids profiles, with heterozygous (HT) and homozygous (HM) groups exhibiting distinct patterns. Hierarchical cluster analysis and multivariate analyses demonstrated a clear separation of HM from HT and control (CO) groups related to cardiac remodeling. Lipidomic profiling identified 257 annotated lipids, with two significantly dysregulated between CO and HT, and 59 between HM and CO. Acylcarnitines and phosphatidylcholines were identified as key contributors to group differentiation, suggesting a shift in energy source. Untargeted metabolomics revealed 110 and 53 significantly dysregulated metabolites. Pathway enrichment analysis highlighted perturbations in multiple metabolic pathways in the HM group, including nicotinate, nicotinamide, purine, glyoxylate, dicarboxylate, glycerophospholipid, pyrimidine, and amino acid metabolism. Our study provides comprehensive insights into the lipidomic and metabolomic unique signatures associated with cmybpc3-/- induced HCM in zebrafish. The identified biomarkers and dysregulated pathways shed light on the metabolic perturbations underlying HCM pathology, offering potential targets for further investigation and potential new therapeutic interventions., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
32. Clinical exome sequencing unravels the diverse spectrum of genetic heterogeneity and genotype-phenotype correlations in hypertrophic cardiomyopathy.
- Author
-
Harikrishnan S, Koshy L, Ganapathi S, Jeemon P, Ramya Das NK, Urulangodi M, Madhuma M, Vysakh Y, Subran A, and Lakshmikanth LR
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Genetic Heterogeneity, Genetic Association Studies methods, Mutation, Cohort Studies, Exome genetics, Genotype, Carrier Proteins, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Exome Sequencing methods
- Abstract
Background: Catalogues of pathogenic genetic mutations in hypertrophic cardiomyopathy (HCM) are disproportionately small when compared to that of the size of the population with South Asian ancestry and their collective increased risk of heart disease., Methods: We conducted clinical exome sequencing of 200 HCM patients to identified cardiomyopathy-associated genetic mutations. The clinical and echocardiographic characteristics of genotype-positive and genotype-negative patients were compared, and the likelihood of detecting a positive genetic test result was evaluated. Allelic burden analysis was done to compare the minor allele frequencies (MAF) of the pathogenic or likely pathogenic (P/LP) variants and variants of uncertain significance (VUSs) identified in the cohort against various population genomics databases., Results: The genetic yield was 40% for P/LP variants, with MYBPC3 and MYH7 as the predominant sarcomere genes. Younger age-at-diagnosis, family history of HCM, asymmetric hypertrophic (ASH) pattern, the ratio of the interventricular septum to posterior wall thickness (IVS/PW ratio), left atrial (LA) dimensions, severe mitral regurgitation grade (MR grade), late gadolinium enhancement (LGE) detected fibrosis and absence of hypertension were associated with an increased likelihood of HCM-associated variants. Patients who experienced ventricular tachycardia and premature cardiovascular death were significantly likely to carry MYBPC3 or loss-of-function variants. LA and interventricular septal (IVS) dimensions were associated with MYH7 variants. The rare variant burden for P/LP variants and VUSs was significantly enriched in HCM cases compared to population controls., Conclusion: Our study provides a comprehensive evaluation of HCM-associated genetic mutations from an Indian population. The identified genotype-phenotype associations could improve the yield of targeted genetic testing in HCM., Competing Interests: Declaration of competing interest The authors declare that there is no conflict of interest., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review.
- Author
-
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, and Piane M
- Subjects
- Humans, High-Throughput Nucleotide Sequencing methods, Mutation, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated diagnosis, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathies genetics, Cardiomyopathies diagnosis, Genetic Predisposition to Disease
- Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews , along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
- Published
- 2024
- Full Text
- View/download PDF
34. Biallelic TYR and TKFC variants in Egyptian patients with OCA1 and new expanded TKFC features.
- Author
-
Ashaat EA, Esmaiel NN, El-Saiedi SA, Ashaat NA, Hussen DF, Ramadan A, Al Kersh MA, AbdelHakim NS, Said I, Metwally AM, and Fayez A
- Subjects
- Humans, Male, Female, Egypt, Alleles, Infant, Newborn, Homozygote, Cardiomyopathy, Hypertrophic genetics, Mutation, Consanguinity, Pedigree
- Abstract
Background: Oculocutaneous albinism type1 (OCA1) is caused by the TYR gene's homozygous and compound heterozygous variants. TKFC gene variants cause triokinase & FMN cyclase deficiency syndrome with variable multisystemic disorders., Objectives: To determine the potential disease-causing variants in two deceased patients presenting atypical OCA1 features by demonstrating three generations for a single family. The two deceased neonates had severe skeletal abnormalities and fatal hypertrophic cardiomyopathy. We also explored the potential mechanisms for the causative relationship between TKFC and multisystem disorders., Patients and Methods: Due to the new emerging symptoms that weren't reported before with the TYR gene, the following methods were performed: Sanger sequencing for the TYR gene, followed by whole exome sequencing, co-segregation, and computational analyses., Results: Extensive parental consanguinity was found, and consequently an autosomal recessive mode of inheritance was prioritized. Upon performing sequencing and segregation data, the following has been confirmed: positive co-segregation of nonsense homozygous NM_000372.5:c.346C > T p.(Arg116*) variant in TYR gene and multisystem disease-missense homozygous NM_015533.4:c.598G > A p.(Val200Ile) variant in TKFC gene in the two affected index patients who deceased due to hypertrophic cardiomyopathy. Using computational analysis, we found that c.598G > A p.(Val200Ile) pathogenicity has led to the failure of L2-K1 active site closure due to the potential differential fluctuation between valine and isoleucine residues. Subsequently, disruption of endogenous DHA phosphorylation was found. Two potential mechanisms exploring the causative relationship between TKFC gene and multisystem disorders have been suggested., Conclusions: This study presented a first family with the co-existence of biallelic variants in TYR and TKFC genes associating severe skeletal abnormalities and lethal hypertrophic cardiomyopathy. Neither of these genes would have been pursued in the standard genetic counseling. Such discovery is paving the way for more efficient genetic counseling. Comparing TKFC results with literature data showed that our relevant expanded TKFC variant is the 3rd worldwide., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
35. Forskolin is an effective therapeutic small molecule for the treatment of hypertrophic cardiomyopathy through ADCY6/cAMP/PKA pathway.
- Author
-
Wang W, Xue Y, Li D, Shao C, Wu K, Sun N, and Chen Q
- Subjects
- Animals, Mice, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Disease Models, Animal, Male, Mice, Inbred C57BL, Humans, Cardiomyopathy, Hypertrophic drug therapy, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cyclic AMP metabolism, Colforsin pharmacology, Colforsin therapeutic use, Adenylyl Cyclases metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Signal Transduction drug effects
- Abstract
Hypertrophic cardiomyopathy (HCM) arises from a pathogenic variant in the gene responsible for encoding the myocardium-associated protein. Forskolin (FSK), a labdane diterpene isolated from Sphingomonas capillaris, exhibits diverse pharmacological effects, including bronchospasm relief, intraocular pressure reduction, and glaucoma treatment. However, whether FSK could regulate HCM and its associated mechanism remains unclear. Here, we discovered that FSK could mitigate cardiac hypertrophy in two HCM mouse models (Myh6
R404Q and Tnnt2R109Q ) in vivo. Additionally, FSK could prevent norepinephrine (NE)-induced cardiomyocyte hypertrophy in vitro. It reversed cardiac dysfunction, reduced enlarged cell size, and downregulated the expression of hypertrophy-related genes. We further demonstrated that FSK's mechanism in alleviating HCM relied on the activation of ADCY6. In conclusion, our findings demonstrate that FSK alleviates hypertrophic cardiomyopathy by modulating the ADCY6/cAMP/PKA pathway, suggesting that FSK holds promise as a therapeutic agent for HCM., Competing Interests: Declaration of competing interest No conflict of interest exits in the submission of this manuscript, and manuscript is approved by all authors for publication., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
36. Canadian Cardiovascular Society Clinical Practice Update on Contemporary Management of the Patient With Hypertrophic Cardiomyopathy.
- Author
-
Crean AM, Adler A, Arbour L, Chan J, Christian S, Cooper RM, Garceau P, Giraldeau G, Heydari B, Laksman Z, Mital S, Ong K, Overgaard C, Ruel M, Seifer CM, Ward MR, and Tadros R
- Subjects
- Humans, Canada, Cardiology methods, Cardiology standards, Death, Sudden, Cardiac prevention & control, Death, Sudden, Cardiac etiology, Disease Management, Genetic Testing methods, Genetic Testing standards, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic therapy, Societies, Medical standards
- Abstract
Numerous guidelines on the diagnosis and management of hypertrophic cardiomyopathy (HCM) have been published, by learned societies, over the past decade. Although helpful they are often long and less adapted to nonexperts. This writing panel was challenged to produce a document that grew as much from years of practical experience as it did from the peer-reviewed literature. As such, rather than produce yet another set of guidelines, we aim herein to deliver a concentrate of our own experiential learning and distill for the reader the essence of effective and appropriate HCM care. This Clinical Practice Update on HCM is therefore aimed at general cardiologists and other cardiovascular practitioners rather than for HCM specialists. We set the stage with a description of the condition and its clinical presentation, discuss the central importance of "obstruction" and how to look for it, review the role of cardiac magnetic resonance imaging, reflect on the appropriate use of genetic testing, review the treatment options for symptomatic HCM-crucially including cardiac myosin inhibitors, and deal concisely with practical issues surrounding risk assessment for sudden cardiac death, and management of the end-stage HCM patient. Uniquely, we have captured the pediatric experience on our panel to discuss appropriate differences in the management of younger patients with HCM. We ask the reader to remember that this document represents expert consensus opinion rather than dogma and to use their best judgement when dealing with the HCM patient in front of them., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
37. Unveiling MiRNA-124 as a biomarker in hypertrophic cardiomyopathy: An innovative approach using machine learning and intelligent data analysis.
- Author
-
Pisklova M and Osmak G
- Subjects
- Humans, Biomarkers metabolism, Gene Expression Profiling methods, Male, Female, MicroRNAs genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Machine Learning
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is a widespread hereditary cardiac pathology characterized by thickened heart walls and rearrangement of cardiomyocytes. Despite extensive research, the mechanisms underlying HCM development remain poorly understood, impeding the development of effective therapeutic and diagnostic strategies. Recent studies have suggested a polygenic nature of HCM development alongside monogenic forms. Transcriptomic profiling is a valuable tool for investigating such diseases. In this study, we propose a novel approach to study regulatory microRNAs (miRNAs) in the context of HCM, utilizing state-of-the-art data analysis tools., Methods and Results: Our method involves applying the Monte Carlo simulation and machine learning algorithm to transcriptomic data to generate high-capacity classifiers for HCM. From these classifiers, we extract key genes crucial for their performance, resulting in the identification of 16 key genes. Subsequently, we narrow down the pool of miRNAs by selecting those that may target the greatest number of key genes within the best models. We particularly focused on miR-124-3p, which we validated to have an association with HCM on an independent dataset. Subsequent investigation of its function revealed involvement of miR-124-3p in the RhoA signaling pathway., Conclusions: In this study we propose a new approach to analyze transcriptomic data to search for microRNAs associated with a disease. Using this approach for transcriptomic profiling data of patients with HCM, we identified miR-124-3p as a potential regulator of the RhoA signaling pathway in the pathogenesis of HCM., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interest., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
38. A Cautionary Tale of Hypertrophic Cardiomyopathy-From "Benign" Left Ventricular Hypertrophy to Stroke, Atrial Fibrillation, and Molecular Genetic Diagnostics: A Case Report and Review of Literature.
- Author
-
Gencheva D, Angelova P, Genova K, Atemin S, Sleptsova M, Todorov T, Nikolov F, Ruseva D, Mitev V, and Todorova A
- Subjects
- Humans, Male, Middle Aged, Stroke genetics, Stroke diagnosis, Echocardiography, NAV1.5 Voltage-Gated Sodium Channel genetics, Atrial Fibrillation genetics, Atrial Fibrillation diagnosis, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic complications, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular diagnosis
- Abstract
This case report concerns a 48-year-old man with a history of ischemic stroke at the age of 41 who reported cardiac hypertrophy, registered in his twenties when explained by increased physical activity. Family history was positive for a mother with permanent atrial fibrillation from her mid-thirties. At the age of 44, he had a first episode of persistent atrial fibrillation, accompanied by left atrial thrombosis while on a direct oral anticoagulant. He presented at our clinic at the age of 45 with another episode of persistent atrial fibrillation and decompensated heart failure. Echocardiography revealed a dilated left atrium, reduced left ventricular ejection fraction, and an asymmetric left ventricular hypertrophy. Cardiac magnetic resonance was positive for a cardiomyopathy with diffuse fibrosis, while slow-flow phenomenon was present on coronary angiography. Genetic testing by whole-exome sequencing revealed three variants in the patient, c.309C > A, p.His103Gln in the ACTC1 gene, c.116T > G, p.Leu39Ter in the PLN gene, and c.5827C > T, p.His1943Tyr in the SCN5A gene, the first two associated with hypertrophic cardiomyopathy and the latter possibly with familial atrial fibrillation. This case illustrates the need for advanced diagnostics in unexplained left ventricular hypertrophy, as hypertrophic cardiomyopathy is often overlooked, leading to potentially debilitating health consequences.
- Published
- 2024
- Full Text
- View/download PDF
39. Characteristics and outcomes associated with sarcomere mutations in patients with hypertrophic cardiomyopathy: A systematic review and meta-analysis.
- Author
-
Huang Z, Lin K, Huang J, Chen Y, Liu H, Zhang X, Luo W, and Xu Z
- Subjects
- Humans, Sarcomeres genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Mutation
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is an inherited heart disease that can lead to sudden cardiac death. Impact of genetic testing for the prognosis and treatment of patients with HCM needs to be improved. We conducted a systematic review and meta-analysis to investigate the characteristics and outcomes associated with sarcomere genotypes in index patients with HCM., Methods: A systematic search was conducted in Medline, Embase, and Cochrane Library up to Dec 31, 2023. Data on clinical characteristics, morphological and imaging features, outcomes and interventions were collected from published studies and pooled using a random-effects meta-analysis., Results: A total of 30 studies with 10,825 HCM index patients were included in the pooled analyses. The frequency of sarcomere genes in HCM patients was 41%. Sarcomere mutations were more frequent in women (p < 0.00001), and were associated with lower body mass index (26.1 ± 4.7 versus 27.5 ± 4.3; p = 0.003) and left ventricular ejection fraction (65.7% ± 10.1% vs. 67.1% ± 8.6%; p = 0.03), less apical hypertrophy (6.5% vs. 20.1%; p < 0.0001) and left ventricular outflow tract obstruction (29.1% vs. 33.2%; p = 0.03), greater left atrial volume index (43.6 ± 21.1 ml/m
2 vs. 37.3 ± 13.0 ml/m2 ; p = 0.02). Higher risks of ventricular tachycardia (23.4% vs. 14.1%; p < 0.0001), syncope (18.3% vs. 10.9%; p = 0.01) and heart failure (17.3% vs. 14.6%; p = 0.002) were also associated with sarcomere mutations., Conclusions: Sarcomere mutations are more frequent in women, and are associated with worse clinical characteristics and poor outcomes., Competing Interests: Declaration of competing interest The authors have no conflicts of interest to declare., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
40. Studying Pathogenetic Contribution of a Variant of Unknown Significance, p.M659I (c.1977G > A) in MYH7, to the Development of Hypertrophic Cardiomyopathy Using CRISPR/Cas9-Engineered Isogenic Induced Pluripotent Stem Cells.
- Author
-
Pavlova SV, Shulgina AE, Zakian SM, and Dementyeva EV
- Subjects
- Humans, Cell Differentiation genetics, Gene Editing methods, Mutation, Cell Line, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells cytology, CRISPR-Cas Systems, Cardiac Myosins genetics, Cardiac Myosins metabolism, Myosin Heavy Chains genetics, Myosin Heavy Chains metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Myocytes, Cardiac cytology
- Abstract
Hypertrophic cardiomyopathy (HCM) is a cardiovascular pathology that is caused by variants in genes encoding sarcomere-associated proteins. However, the clinical significance of numerous variants in HCM-associated genes is still unknown. CRISPR/Cas9 is a tool of nucleotide sequence editing that allows for the unraveling of different biological tasks. In this study, introducing a mutation with CRISPR/Cas9 into induced pluripotent stem cells (iPSCs) of a healthy donor and the directed differentiation of the isogenic iPSC lines into cardiomyocytes were used to assess the pathogenicity of a variant of unknown significance, p.M659I (c.1977G > A) in MYH7 , which was found previously in an HCM patient. Using two single-stranded donor oligonucleotides with and without the p.M659I (c.1977G > A) mutation, together with CRISPR/Cas9, an iPSC line heterozygous at the p.M659I (c.1977G > A) variant in MYH7 was generated. No CRISPR/Cas9 off-target activity was observed. The iPSC line with the introduced p.M659I (c.1977G > A) mutation in MYH7 retained its pluripotent state and normal karyotype. Compared to the isogenic control, cardiomyocytes derived from the iPSCs with the introduced p.M659I (c.1977G > A) mutation in MYH7 recapitulated known HCM features: enlarged size, elevated diastolic calcium level, changes in the expression of HCM-related genes, and disrupted energy metabolism. These findings indicate the pathogenicity of the variant.
- Published
- 2024
- Full Text
- View/download PDF
41. Genetics of hypertrophic cardiomyopathy: established and emerging implications for clinical practice.
- Author
-
Lopes LR, Ho CY, and Elliott PM
- Subjects
- Humans, Mutation, Genetic Predisposition to Disease genetics, Genetic Variation genetics, Genetic Testing methods, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Sarcomeres genetics
- Abstract
Pathogenic variation in genes encoding proteins of the cardiac sarcomere is responsible for 30%-40% of cases of hypertrophic cardiomyopathy. The main clinical utility of genetic testing is to provide diagnostic confirmation and facilitation of family screening. It also assists in the detection of aetiologies, which require distinct monitoring and treatment approaches. Other clinical applications, including the use of genetic information to inform risk prediction models, have been limited by the challenge of establishing robust genotype-phenotype correlations with actionable consequences, but new data on the interaction between rare and common genetic variation, as well as the emergence of therapies targeting disease-specific pathogenic mechanisms, herald a new era for genetic testing in routine practice., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
42. Cardiac Phenotype and Gene Mutations in RASopathies.
- Author
-
Faienza MF, Meliota G, Mentino D, Ficarella R, Gentile M, Vairo U, and D'amato G
- Subjects
- Humans, Phenotype, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, ras Proteins genetics, ras Proteins metabolism, MAP Kinase Signaling System genetics, Pulmonary Valve Stenosis genetics, Pulmonary Valve Stenosis pathology, Genetic Association Studies, Heart Defects, Congenital genetics, Heart Defects, Congenital pathology, Mutation, Noonan Syndrome genetics, Noonan Syndrome pathology
- Abstract
Cardiac involvement is a major feature of RASopathies, a group of phenotypically overlapping syndromes caused by germline mutations in genes encoding components of the RAS/MAPK (mitogen-activated protein kinase) signaling pathway. In particular, Noonan syndrome (NS) is associated with a wide spectrum of cardiac pathologies ranging from congenital heart disease (CHD), present in approximately 80% of patients, to hypertrophic cardiomyopathy (HCM), observed in approximately 20% of patients. Genotype-cardiac phenotype correlations are frequently described, and they are useful indicators in predicting the prognosis concerning cardiac disease over the lifetime. The aim of this review is to clarify the molecular mechanisms underlying the development of cardiac diseases associated particularly with NS, and to discuss the main morphological and clinical characteristics of the two most frequent cardiac disorders, namely pulmonary valve stenosis (PVS) and HCM. We will also report the genotype-phenotype correlation and its implications for prognosis and treatment. Knowing the molecular mechanisms responsible for the genotype-phenotype correlation is key to developing possible targeted therapies. We will briefly address the first experiences of targeted HCM treatment using RAS/MAPK pathway inhibitors.
- Published
- 2024
- Full Text
- View/download PDF
43. COQ7 defect causes prenatal onset of mitochondrial CoQ 10 deficiency with cardiomyopathy and gastrointestinal obstruction.
- Author
-
Pettenuzzo I, Carli S, Sánchez-Cuesta A, Isidori F, Montanari F, Grippa M, Lanzoni G, Ambrosetti I, Di Pisa V, Cordelli DM, Mondardini MC, Pippucci T, Ragni L, Cenacchi G, Costa R, Lima M, Capristo MA, Tropeano CV, Caporali L, Carelli V, Brunelli E, Maffei M, Ahmed Sheikhmaye H, Fetta A, Brea-Calvo G, and Garone C
- Subjects
- Female, Humans, Infant, Male, Ataxia genetics, Ataxia pathology, Ataxia diagnosis, Cardiomyopathies genetics, Cardiomyopathies pathology, Cardiomyopathies diagnosis, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Cardiomyopathy, Hypertrophic diagnosis, Muscle Weakness genetics, Muscle Weakness pathology, Mutation, Ophthalmoplegia genetics, Ophthalmoplegia pathology, Ophthalmoplegia diagnosis, Pedigree, Mixed Function Oxygenases genetics, Mixed Function Oxygenases metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Mitochondrial Diseases genetics, Mitochondrial Diseases pathology, Mitochondrial Diseases diagnosis, Ubiquinone analogs & derivatives, Ubiquinone deficiency, Ubiquinone genetics
- Abstract
COQ7 pathogenetic variants cause primary CoQ
10 deficiency and a clinical phenotype of encephalopathy, peripheral neuropathy, or multisystemic disorder. Early diagnosis is essential for promptly starting CoQ10 supplementation. Here, we report novel compound heterozygous variants in the COQ7 gene responsible for a prenatal onset (20 weeks of gestation) of hypertrophic cardiomyopathy and intestinal dysmotility in a Bangladesh consanguineous family with two affected siblings. The main clinical findings were dysmorphisms, recurrent intestinal occlusions that required ileostomy, left ventricular non-compaction cardiomyopathy, ascending aorta dilation, arterial hypertension, renal dysfunction, diffuse skin desquamation, axial hypotonia, neurodevelopmental delay, and growth retardation. Exome sequencing revealed compound heterozygous rare variants in the COQ7 gene, c.613_617delGCCGGinsCAT (p.Ala205HisfsTer48) and c.403A>G (p.Met135Val). In silico analysis and functional in vitro studies confirmed the pathogenicity of the variants responsible for abolished activities of complexes I + III and II + III in muscle homogenate, severe decrease of CoQ10 levels, and reduced basal and maximal respiration in patients' fibroblasts. The first proband deceased at 14 months of age, whereas supplementation with a high dose of CoQ10 (30 mg/kg/day) since the first days of life modified the clinical course in the second child, showing a recovery of milestones acquirement at the last follow-up (18 months of age). Our study expands the clinical spectrum of primary CoQ10 deficiency due to COQ7 gene defects and highlights the essential role of multidisciplinary and combined approaches for a timely diagnosis., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
44. Hypertrophic Cardiomyopathy.
- Author
-
Dungu JN, Hardy-Wallace A, Dimarco AD, and Savage HO
- Subjects
- Humans, Death, Sudden, Cardiac prevention & control, Death, Sudden, Cardiac etiology, Genetic Testing methods, Cardiomyopathy, Hypertrophic therapy, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic genetics
- Abstract
Purpose of Review: Hypertrophic cardiomyopathy (HCM) is a common inherited cardiac condition with potential for severe complications including sudden cardiac death. Early diagnosis allows appropriate risk stratification and prompt intervention to minimise the potential for adverse outcomes. The implications of poorly coordinated screening are significant, either missing relatives at high-risk or burdening low-risk individuals with a diagnosis associated with reduced life expectancy. We aim to guide clinicians through the diagnostic pathway through to novel treatment options. Several conditions mimic the condition, and we discuss the phenocopies and how to differentiate from HCM., Recent Findings: We summarise the latest developments informing clinical decision making in the modern era of myosin inhibitors and future gene editing therapies. Early identification will enable prompt referral to specialist centres. A diagnostic flowchart is included, to guide the general cardiology and heart failure clinician in important decision making regarding the care of the HCM patient and importantly their relatives at risk. We have highlighted the importance of screening because genotype-positive/phenotype-negative patients are likely to have the most to gain from novel therapies., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
45. Machine learning-driven diagnostic signature provides new insights in clinical management of hypertrophic cardiomyopathy.
- Author
-
Liu S, Yuan P, Zheng Y, Guo C, Ren Y, Weng S, Zhang Y, Liu L, Xing Z, Wang L, and Han X
- Subjects
- Humans, Gene Expression Profiling methods, Disease Management, Male, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Machine Learning
- Abstract
Aims: In an era of evolving diagnostic possibilities, existing diagnostic systems are not fully sufficient to promptly recognize patients with early-stage hypertrophic cardiomyopathy (HCM) without symptomatic and instrumental features. Considering the sudden death of HCM, developing a novel diagnostic model to clarify the patients with early-stage HCM and the immunological characteristics can avoid misdiagnosis and attenuate disease progression., Methods and Results: Three hundred eighty-five samples from four independent cohorts were systematically retrieved. The weighted gene co-expression network analysis, differential expression analysis (|log2(foldchange)| > 0.5 and adjusted P < 0.05), and protein-protein interaction network were sequentially performed to identify HCM-related hub genes. With a machine learning algorithm, the least absolute shrinkage and selection operator regression algorithm, a stable diagnostic model was developed. The immune-cell infiltration and biological functions of HCM were also explored to characterize its underlying pathogenic mechanisms and the immune signature. Two key modules were screened based on weighted gene co-expression network analysis. Pathogenic mechanisms relevant to extracellular matrix and immune pathways have been discovered. Twenty-seven co-regulated genes were recognized as HCM-related hub genes. Based on the least absolute shrinkage and selection operator algorithm, a stable HCM diagnostic model was constructed, which was further validated in the remaining three cohorts (n = 385). Considering the tight association between HCM and immune-related functions, we assessed the infiltrating abundance of various immune cells and stromal cells based on the xCell algorithm, and certain immune cells were significantly different between high-risk and low-risk groups., Conclusions: Our study revealed a number of hub genes and novel pathways to provide potential targets for the treatment of HCM. A stable model was developed, providing an efficient tool for the diagnosis of HCM., (© 2024 The Authors. ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
46. Phenotypic Spectrum of Subclinical Sarcomere-Related Hypertrophic Cardiomyopathy and Transition to Overt Disease.
- Author
-
Topriceanu CC, Moon JC, Axelsson Raja A, Captur G, and Ho CY
- Subjects
- Humans, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular diagnostic imaging, Disease Progression, Sarcomeres genetics, Sarcomeres metabolism, Sarcomeres pathology, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Phenotype
- Abstract
Genetic hypertrophic cardiomyopathy (HCM) is classically caused by pathogenic/likely pathogenic variants in sarcomere genes (G+). Currently, HCM is diagnosed if there is unexplained left ventricular (LV) hypertrophy with LV wall thickness ≥15 mm in probands or ≥13 mm in at-risk relatives. Although LV hypertrophy is a key feature, this binary metric does not encompass the full constellation of phenotypic features, particularly in the subclinical stage of the disease. Subtle phenotypic manifestations can be identified in sarcomere variant carriers with normal LV wall thickness, before diagnosis with HCM (G+/LV hypertrophy-; subclinical HCM). We conducted a systematic review to summarize current knowledge about the phenotypic spectrum of subclinical HCM and factors influencing penetrance and expressivity. Although the mechanisms driving the development of LV hypertrophy are yet to be elucidated, activation of profibrotic pathways, impaired relaxation, abnormal Ca
2+ signaling, altered myocardial energetics, and microvascular dysfunction have all been identified in subclinical HCM. Progression from subclinical to clinically overt HCM may be more likely if early phenotypic manifestations are present, including ECG abnormalities, longer mitral valve leaflets, lower global E' velocities on Doppler echocardiography, and higher serum N-terminal propeptide of B-type natriuretic peptide. Longitudinal studies of variant carriers are critically needed to improve our understanding of penetrance, characterize the transition to disease, identify risk predictors of phenotypic evolution, and guide the development of novel treatment strategies aimed at influencing disease trajectory., Competing Interests: The views expressed in this article are those of the authors who declare that they have no conflict of interest, except for Dr Moon who is the chief executive officer of MyCardium AI and has served on advisory boards for Genzyme and Sanofi.- Published
- 2024
- Full Text
- View/download PDF
47. Clinical and Immunologic Features of a Patient With Homozygous FNIP1 Variant.
- Author
-
Ulaş S, Naiboğlu S, Özyilmaz İ, Öztürk Demir AG, Turan I, Yuzkan S, Ayaz A, and Çeliksoy MH
- Subjects
- Humans, Male, Infant, Agammaglobulinemia genetics, Carrier Proteins genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Neutropenia genetics, Neutropenia congenital, Neutropenia immunology, Homozygote
- Abstract
Agammaglobulinemia represents the most profound primary antibody deficiency, stemming from early cessation of B-cell development. Deficiency in folliculin-interacting protein 1 (FNIP1) is a novel inborn error of immunity characterized by a severe defect in B-cell development, agammaglobulinemia, variable neutropenia, and hypertrophic cardiomyopathy. FNIP1 plays a critical role in B-cell development and metabolic homeostasis, establishing a metabolic checkpoint that ensures pre-B cells possess sufficient metabolic capacity to undergo division while concurrently limiting lymphogenesis due to abnormal growth. Disruption of FNIP1 functionality affects the fundamental metabolic regulators adenosine monophosphate-activated protein kinase and mTOR, culminating in a severe B-cell deficiency alongside hypogammaglobulinemia, hypertrophic cardiomyopathy, preexcitation syndrome, and intermittent neutropenia. This case report presents an 11-month-old male patient with FNIP1 deficiency who, in addition to classical features, exhibited posterior cerebellar hypoplasia., Competing Interests: The authors declare no conflict of interest., (Copyright © 2024 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
48. Different clinical presentation, cardiac morphology and gene mutations in two sisters with hypertrophic cardiomyopathy-A case report.
- Author
-
Tayier B, Lv J, Ma L, Guan L, and Mu Y
- Subjects
- Humans, Female, Echocardiography, DNA Mutational Analysis, Magnetic Resonance Imaging, Cine methods, Myocardium pathology, Pedigree, Adult, DNA genetics, Mutation, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic diagnosis, Siblings
- Published
- 2024
- Full Text
- View/download PDF
49. Survival analysis and gender differences in hypertrophic cardiomyopathy proband patients referred for genetic testing.
- Author
-
Lorca R, Salgado M, Álvarez-Velasco R, Reguro JR, Alonso V, Gómez J, Coto E, Cuesta-Llavona E, Lopez-Negrete E, Pascual I, Avanzas P, and Tome M
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, Survival Analysis, Cohort Studies, Follow-Up Studies, Survival Rate trends, Referral and Consultation, Spain epidemiology, Sex Factors, Sex Characteristics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic mortality, Cardiomyopathy, Hypertrophic diagnosis, Genetic Testing methods
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is believed to have low overall mortality rate, that could be influenced by gender, particularly among probands. We aimed to evaluate the survival rates and possible gender differences in a homogeneous cohort of HCM proband patients, referred for genetic testing, from the same geographical area, without differences in medical care access nor clinical referral pathways., Methods: we compared the mortality rates of a cohort of consecutive HCM probands referred for genetic testing (2000-2022), from a Spanish region (xxx1) with a centralized genetic testing pathway, with its control reference population by Ederer II method. Gender differences were analyzed., Results: Among the 649 HCM probands included in this study, there were significantly more men than women (61.3% vs 38.7, p < 0.05), with an earlier diagnosis (53.5 vs 61.1 years old, p < 0.05). Clinical evolution or arrhythmogenic HCM profile did no show no significant gender differences. Mean follow up was 9,8 years ±6,6 SD (9,9 ± 7 vs 9,6 ± 6,1, p = 0.59). No statistically significant differences in observed mortality, expected survival and excess mortality were found in the general HCM proband cohort. However, we found a significant excess mortality in female probands with HCM. No additional differences in analysis by genetic status were identified., Conclusion: Expected survival in our HCM probands did not differ from its reference population. However, despite no gender differences in phenotype severity were identified, proband HCM women did present a diagnosis delay and worse mortality outcomes., Competing Interests: Declaration of competing interest There are no conflicts of interest. This research was funded by Instituto de Salud Carlos III (ISCIII) (grant number PI22/00705). All patients signed the informed consent (local Ethical Committee approval CEImPA 2022.254)., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
50. Machine learning and experimental validation of novel biomarkers for hypertrophic cardiomyopathy and cancers.
- Author
-
Dai H, Liu Y, Zhu M, Tao S, Hu C, Luo P, Jiang A, and Zhang G
- Subjects
- Humans, Male, Female, Prognosis, Biomarkers, Tumor metabolism, Biomarkers, Tumor genetics, Middle Aged, Nomograms, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic genetics, Machine Learning, Neoplasms metabolism, Neoplasms diagnosis, Neoplasms genetics, Neoplasms pathology, Biomarkers metabolism
- Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary cardiac disorder marked by anomalous thickening of the myocardium, representing a significant contributor to mortality. While the involvement of immune inflammation in the development of cardiac ailments is well-documented, its specific impact on HCM pathogenesis remains uncertain. Five distinct machine learning algorithms, namely LASSO, SVM, RF, Boruta and XGBoost, were utilized to discover new biomarkers associated with HCM. A unique nomogram was developed using two newly identified biomarkers and subsequently validated. Furthermore, samples of HCM and normal heart tissues were gathered from our institution to confirm the variance in expression levels and prognostic significance of GATM and MGST1. Five novel biomarkers (DARS2, GATM, MGST1, SDSL and ARG2) associated with HCM were identified. Subsequent validation revealed that GATM and MGST1 exhibited significant diagnostic utility for HCM in both the training and test cohorts, with all AUC values exceeding 0.8. Furthermore, a novel risk assessment model for HCM patients based on the expression levels of GATM and MGST1 demonstrated favourable performance in both the training (AUC = 0.88) and test cohorts (AUC = 0.9). Furthermore, our study revealed that GATM and MGST1 exhibited elevated expression levels in HCM tissues, demonstrating strong discriminatory ability between HCM and normal cardiac tissues (AUC of GATM = 0.79; MGST1 = 0.86). Our findings suggest that two specific cell types, monocytes and multipotent progenitors (MPP), may play crucial roles in the pathogenesis of HCM. Notably, GATM and MGST1 were found to be highly expressed in various tumours and showed significant prognostic implications. Functionally, GATM and MGST1 are likely involved in xenobiotic metabolism and epithelial mesenchymal transition in a wide range of cancer types. GATM and MGST1 have been identified as novel biomarkers implicated in the progression of both HCM and cancer. Additionally, monocytes and MPP may also play a role in facilitating the progression of HCM., (© 2024 The Author(s). Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.