51
|
Hermida A, Ader F, Millat G, Jedraszak G, Maury P, Cador R, Catalan PA, Clerici G, Combes N, De Groote P, Dupin-Deguine D, Eschalier R, Faivre L, Garcia P, Guillon B, Janin A, Kugener B, Lackmy M, Laredo M, Le Guillou X, Lesaffre F, Lucron H, Milhem A, Nadeau G, Nguyen K, Palmyre A, Perdreau E, Picard F, Rebotier N, Richard P, Rooryck C, Seitz J, Verloes A, Vernier A, Winum P, Yabeta GAD, Bouchot O, Chevalier P, Charron P, Gandjbakhch E. NEXN Gene in Cardiomyopathies and Sudden Cardiac Deaths: Prevalence, Phenotypic Expression, and Prognosis. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004285. [PMID: 38059363 DOI: 10.1161/circgen.123.004285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/05/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Few clinical data are available on NEXN mutation carriers, and the gene's involvement in cardiomyopathies or sudden death has not been fully established. Our objectives were to assess the prevalence of putative pathogenic variants in NEXN and to describe the phenotype and prognosis of patients carrying the variants. METHODS DNA samples from consecutive patients with cardiomyopathy or sudden cardiac death/sudden infant death syndrome/idiopathic ventricular fibrillation were sequenced with a custom panel of genes. Index cases carrying at least one putative pathogenic variant in the NEXN gene were selected. RESULTS Of the 9516 index patients sequenced, 31 were carriers of a putative pathogenic variant in NEXN only, including 2 with double variants and 29 with a single variant. Of the 29 unrelated probands with a single variant (16 males; median age at diagnosis, 32.0 [26.0-49.0] years), 21 presented with dilated cardiomyopathy (prevalence, 0.33%), and 3 presented with hypertrophic cardiomyopathy (prevalence, 0.14%). Three patients had idiopathic ventricular fibrillation, and there were 2 cases of sudden infant death syndrome (prevalence, 0.46%). For patients with dilated cardiomyopathy, the median left ventricle ejection fraction was 37.5% (26.25-50.0) at diagnosis and improved with treatment in 13 (61.9%). Over a median follow-up period of 6.0 years, we recorded 3 severe arrhythmic events and 2 severe hemodynamic events. CONCLUSIONS Putative pathogenic NEXN variants were mainly associated with dilated cardiomyopathy; in these individuals, the prognosis appeared to be relatively good. However, severe and early onset phenotypes were also observed-especially in patients with double NEXN variants. We also detected NEXN variants in patients with hypertrophic cardiomyopathy and sudden infant death syndrome/idiopathic ventricular fibrillation, although a causal link could not be established.
Collapse
Affiliation(s)
- Alexis Hermida
- Cardiology, Arrhythmia, and Cardiac Stimulation Service (A.H.), Amiens-Picardie University Hospital
- EA4666 HEMATIM, University of Picardie-Jules Verne, Amiens (A.H., G.J.)
- Institute of Cardiology and ICAN Institute for Cardiometabolism and Nutrition (A.H., M. Laredo, P. Charron, E.G.)
- Department of Genetics, Department of Cardiology, and Referral center for hereditary cardiac diseases, APHP, Pitié-Salpêtrière Hospital (A.H., P. Charron, E.G.)
| | - Flavie Ader
- Unité Pédagogique de Biochimie, Département des Sciences Biologiques et Médicales, UFR de Pharmacie-Faculté de Santé, Université Paris Cité (F.A.)
- Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, DMU Biogem, Service de Biochimie Métabolique, AP-HP-Sorbonne Université, Pitié-Salpêtrière -Charles Foix (F.A., P.R.)
- Sorbonne Université, INSERM 1166, Paris (F.A., M. Laredo, P.R., P. Charron, E.G.)
| | - Gilles Millat
- Service de Génétique Moléculaire, Hospices Civils de Lyon (G.M., A.J.)
| | - Guillaume Jedraszak
- Molecular Genetics Laboratory (G.J.), Amiens-Picardie University Hospital
- EA4666 HEMATIM, University of Picardie-Jules Verne, Amiens (A.H., G.J.)
| | | | - Romain Cador
- Service de Cardiologie, Hôpital Saint Joseph, Paris (R.C.)
| | | | - Gaël Clerici
- Service de Cardiologie, Centre hospitalier universitaire, Saint Pierre, La Réunion (G.C.)
| | - Nicolas Combes
- Service de Cardiologie, Clinique Pasteur, Toulouse (N.C.)
| | - Pascal De Groote
- France CHU Lille, Service de Cardiologie & Inserm U1167, Institut Pasteur de Lille (P.D.G.)
| | | | | | | | - Patricia Garcia
- Unité Mort Inattendue du Nourrisson, Hôpital de la Conception, APHM, Marseille (P.G.)
| | | | - Alexandre Janin
- Service de Génétique Moléculaire, Hospices Civils de Lyon (G.M., A.J.)
| | | | - Marylin Lackmy
- Unité de Génétique Clinique, CHU de Guadeloupe, Pointe à Pitre (M. Lackmy)
| | - Mikael Laredo
- Institute of Cardiology and ICAN Institute for Cardiometabolism and Nutrition (A.H., M. Laredo, P. Charron, E.G.)
- Sorbonne Université, INSERM 1166, Paris (F.A., M. Laredo, P.R., P. Charron, E.G.)
| | | | | | - Hugues Lucron
- Service de Cardiologie pédiatrique, CHU Martinique, Fort-de-France (H.L.)
| | | | - Gwenaël Nadeau
- Service de génétique clinique CH Métropole Savoie, Chambéry (G.N.)
| | | | - Aurélien Palmyre
- APHP, Ambroise Paré Hospital, Department of Genetics and Referral center for cardiac hereditary cardiac diseases, Boulogne-Billancourt (A.P., P. Charron)
| | - Elodie Perdreau
- Département médico chirurgical de cardiologie pédiatrique (E.P.), Hôpital Louis Pradel, HCL, Lyon
| | - François Picard
- Service de Cardiologie, Hôpital Cardiologique Haut Leveque, Bordeaux (F.P.)
| | | | - Pascale Richard
- Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, DMU Biogem, Service de Biochimie Métabolique, AP-HP-Sorbonne Université, Pitié-Salpêtrière -Charles Foix (F.A., P.R.)
- Sorbonne Université, INSERM 1166, Paris (F.A., M. Laredo, P.R., P. Charron, E.G.)
| | | | - Julien Seitz
- Service de Cardiologie, Hôpital Saint Joseph, Marseille (J.S.)
| | - Alain Verloes
- Departement de génétique, Hôpital Robert Debré, APHP (A. Verloes)
| | | | | | - Grace-A-Dieu Yabeta
- Service de Cardiologie, CH Ouest Guyane, Saint-Laurent-du-Maroni (G.-A.-D.Y.)
| | - Océane Bouchot
- Service de Cardiologie, CH Annecy Genevois, Annecy, France (O.B.)
| | | | - Philippe Charron
- Institute of Cardiology and ICAN Institute for Cardiometabolism and Nutrition (A.H., M. Laredo, P. Charron, E.G.)
- Department of Genetics, Department of Cardiology, and Referral center for hereditary cardiac diseases, APHP, Pitié-Salpêtrière Hospital (A.H., P. Charron, E.G.)
- Sorbonne Université, INSERM 1166, Paris (F.A., M. Laredo, P.R., P. Charron, E.G.)
- APHP, Ambroise Paré Hospital, Department of Genetics and Referral center for cardiac hereditary cardiac diseases, Boulogne-Billancourt (A.P., P. Charron)
| | - Estelle Gandjbakhch
- Institute of Cardiology and ICAN Institute for Cardiometabolism and Nutrition (A.H., M. Laredo, P. Charron, E.G.)
- Department of Genetics, Department of Cardiology, and Referral center for hereditary cardiac diseases, APHP, Pitié-Salpêtrière Hospital (A.H., P. Charron, E.G.)
- Sorbonne Université, INSERM 1166, Paris (F.A., M. Laredo, P.R., P. Charron, E.G.)
| |
Collapse
|
52
|
Afana AS, Vasiliu L, Sascău R, Adam RD, Rădulescu C, Onciul S, Cinteză E, Chirita-Emandi A, Jurcuț R. Phospholamban p.Leu39* Cardiomyopathy Compared with Other Sarcomeric Cardiomyopathies: Age-Matched Patient Cohorts and Literature Review. J Cardiovasc Dev Dis 2024; 11:41. [PMID: 38392255 PMCID: PMC10889724 DOI: 10.3390/jcdd11020041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heterogeneous genetic disorder, most often caused by sarcomeric gene mutations, with a small proportion due to variants in non-sarcomeric loci. Phospholamban (PLN) is a phosphoprotein associated with the cardiac sarcoplasmic reticulum, a major determinant of cardiac contractility and relaxation. We conducted a retrospective study to determine the prevalence, phenotypical spectrum and clinical course of patients carrying the PLN p.Leu39* variant. A cohort including 11 PLN patients was identified among all patients with HCM (9/189, 4.8%) and DCM (2/62, 3.2%) who underwent genetic testing from two tertiary centers and five more were detected through cascade screening. Complete phenotyping was performed. PLN p.Leu39* variant-driven cardiomyopathy presented mostly as hypertrophic, with frequent progression to end-stage dilated HCM. We proceeded to compare these results to a similar analysis of a control cohort consisting of age-matched individuals that inherited pathogenic or likely pathogenic variants in common sarcomeric genes (MYBPC3/MYH7). Overall, the clinical characteristics and examination findings of patients carrying PLN p.Leu39* were not different from patients with cardiomyopathy related to sarcomeric mutations except for the presence of pathological Q waves and the incidence of non-sustained ventricular arrhythmias, which were higher in PLN patients than in those with MYBPC3/MYH7-related diseases.
Collapse
Affiliation(s)
- Andreea Sorina Afana
- Expert Center for Genetic Cardiovascular Diseases, Emergency Institute for Cardiovascular Diseases, 258 Fundeni Street, 022328 Bucharest, Romania
- Emergency Clinical County Hospital Craiova, 1 Tabaci Street, 200642 Craiova, Romania
- Cardiology Department, University of Medicine and Pharmacy Craiova, 2 Petru Rares Street, 200349 Craiova, Romania
| | - Laura Vasiliu
- Institute of Cardiovascular Diseases "Prof. Dr. George I.M. Georgescu", 700503 Iași, Romania
- Cardiology Department, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iași, Romania
| | - Radu Sascău
- Institute of Cardiovascular Diseases "Prof. Dr. George I.M. Georgescu", 700503 Iași, Romania
- Cardiology Department, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iași, Romania
| | - Robert Daniel Adam
- Expert Center for Genetic Cardiovascular Diseases, Emergency Institute for Cardiovascular Diseases, 258 Fundeni Street, 022328 Bucharest, Romania
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila", 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Cristina Rădulescu
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila", 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
- Emerald Medical Center, 75 Nicolae G. Caramfil Street, 077190 Bucharest, Romania
| | - Sebastian Onciul
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila", 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
- Emerald Medical Center, 75 Nicolae G. Caramfil Street, 077190 Bucharest, Romania
- Emergency Clinical Hospital Floreasca, 8 Calea Floreasca, 014461 Bucharest, Romania
| | - Eliza Cinteză
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila", 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
- Department of Pediatric Cardiology, "Marie Curie" Emergency Children's Hospital, 41451 Bucharest, Romania
| | - Adela Chirita-Emandi
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy "Victor Babeș" Timișoara, 2 Piaţa Eftimie Murgu Street, 300041 Timişoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children "Louis Țurcanu" Timișoara, 2 Doctor Iosif Nemoianu Street, 300011 Timișoara, Romania
| | - Ruxandra Jurcuț
- Expert Center for Genetic Cardiovascular Diseases, Emergency Institute for Cardiovascular Diseases, 258 Fundeni Street, 022328 Bucharest, Romania
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila", 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| |
Collapse
|
53
|
Ananthamohan K, Stelzer JE, Sadayappan S. Hypertrophic cardiomyopathy in MYBPC3 carriers in aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:9. [PMID: 38406555 PMCID: PMC10883298 DOI: 10.20517/jca.2023.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by abnormal thickening of the myocardium, leading to arrhythmias, heart failure, and elevated risk of sudden cardiac death, particularly among the young. This inherited disease is predominantly caused by mutations in sarcomeric genes, among which those in the cardiac myosin binding protein-C3 (MYBPC3) gene are major contributors. HCM associated with MYBPC3 mutations usually presents in the elderly and ranges from asymptomatic to symptomatic forms, affecting numerous cardiac functions and presenting significant health risks with a spectrum of clinical manifestations. Regulation of MYBPC3 expression involves various transcriptional and translational mechanisms, yet the destiny of mutant MYBPC3 mRNA and protein in late-onset HCM remains unclear. Pathogenesis related to MYBPC3 mutations includes nonsense-mediated decay, alternative splicing, and ubiquitin-proteasome system events, leading to allelic imbalance and haploinsufficiency. Aging further exacerbates the severity of HCM in carriers of MYBPC3 mutations. Advancements in high-throughput omics techniques have identified crucial molecular events and regulatory disruptions in cardiomyocytes expressing MYBPC3 variants. This review assesses the pathogenic mechanisms that promote late-onset HCM through the lens of transcriptional, post-transcriptional, and post-translational modulation of MYBPC3, underscoring its significance in HCM across carriers. The review also evaluates the influence of aging on these processes and MYBPC3 levels during HCM pathogenesis in the elderly. While pinpointing targets for novel medical interventions to conserve cardiac function remains challenging, the emergence of personalized omics offers promising avenues for future HCM treatments, particularly for late-onset cases.
Collapse
Affiliation(s)
- Kalyani Ananthamohan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
54
|
Kumar P, Paramasivam G, Devasia T, Prabhu M, Rai MK, Prakashini K, Mallya S, Reghunathan D, Megha A, Nayak K, Moka R. A Novel TPM1 Mutation Causes Familial Hypertrophic Cardiomyopathy in an Indian Family: Genetic and Clinical Correlation. Indian J Clin Biochem 2024; 39:142-145. [PMID: 38223010 PMCID: PMC10784234 DOI: 10.1007/s12291-022-01036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited cardiac disorder characterised by unexplained left ventricular hypertrophy in the absence of abnormal loading conditions. The global prevalence of HCM is estimated to be 1 in 250 in the general population. It is caused due to mutations in genes coding for sarcomeric proteins. α-tropomyosin (TPM1) is an important protein in the sarcomeric thin filament which regulates sarcomere contraction. Mutations in TPM1 are known to cause hypertrophic cardiomyopathy, dilated cardiomyopathy and left ventricular non-compaction. Mutations in TPM1 causing hypertrophic cardiomyopathy are < 1%. However, some high-risk mutations causing sudden cardiac death are also known in this gene. We present a case of a novel heterozygous TPM1 mutation, NM_001018005.2:c.203A>G, p.Gln68Arg; co-segregating in an Indian family with hypertrophic cardiomyopathy. Our report expands the mutational spectrum of HCM due to TPM1 and provides the correlated cardiac phenotype.
Collapse
Affiliation(s)
- Prabodh Kumar
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| | - Ganesh Paramasivam
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Tom Devasia
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Mukund Prabhu
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Maneesh K. Rai
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Mangalore, 575001 Karnataka India
| | - K. Prakashini
- Department of Radiodiagnosis and Imaging, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Dinesh Reghunathan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| | - A. Megha
- Department of Cardiovascular Technology, Manipal College of Health Profession, Manipal Academy of Higher Education (Manipal), Manipal, 576104 Karnataka India
| | - Krishnananda Nayak
- Department of Cardiovascular Technology, Manipal College of Health Profession, Manipal Academy of Higher Education (Manipal), Manipal, 576104 Karnataka India
| | - Rajasekhar Moka
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| |
Collapse
|
55
|
Hosseini SY, Mallick R, Mäkinen P, Ylä-Herttuala S. Navigating the prime editing strategy to treat cardiovascular genetic disorders in transforming heart health. Expert Rev Cardiovasc Ther 2024; 22:75-89. [PMID: 38494784 DOI: 10.1080/14779072.2024.2328642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/06/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION After understanding the genetic basis of cardiovascular disorders, the discovery of prime editing (PE), has opened new horizons for finding their cures. PE strategy is the most versatile editing tool to change cardiac genetic background for therapeutic interventions. The optimization of elements, prediction of efficiency, and discovery of the involved genes regulating the process have not been completed. The large size of the cargo and multi-elementary structure makes the in vivo heart delivery challenging. AREAS COVERED Updated from recent published studies, the fundamentals of the PEs, their application in cardiology, potentials, shortcomings, and the future perspectives for the treatment of cardiac-related genetic disorders will be discussed. EXPERT OPINION The ideal PE for the heart should be tissue-specific, regulatable, less immunogenic, high transducing, and safe. However, low efficiency, sup-optimal PE architecture, the large size of required elements, the unclear role of transcriptomics on the process, unpredictable off-target effects, and its context-dependency are subjects that need to be considered. It is also of great importance to see how beneficial or detrimental cell cycle or epigenomic modifier is to bring changes into cardiac cells. The PE delivery is challenging due to the size, multi-component properties of the editors and liver sink.
Collapse
Affiliation(s)
- Seyed Younes Hosseini
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Bacteriology and Virology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
56
|
Martinez-Martin I, Crousilles A, Ochoa JP, Velazquez-Carreras D, Mortensen SA, Herrero-Galan E, Delgado J, Dominguez F, Garcia-Pavia P, de Sancho D, Wilmanns M, Alegre-Cebollada J. Titin domains with reduced core hydrophobicity cause dilated cardiomyopathy. Cell Rep 2023; 42:113490. [PMID: 38052212 DOI: 10.1016/j.celrep.2023.113490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The underlying genetic defect in most cases of dilated cardiomyopathy (DCM), a common inherited heart disease, remains unknown. Intriguingly, many patients carry single missense variants of uncertain pathogenicity targeting the giant protein titin, a fundamental sarcomere component. To explore the deleterious potential of these variants, we first solved the wild-type and mutant crystal structures of I21, the titin domain targeted by pathogenic variant p.C3575S. Although both structures are remarkably similar, the reduced hydrophobicity of deeply buried position 3575 strongly destabilizes the mutant domain, a scenario supported by molecular dynamics simulations and by biochemical assays that show no disulfide involving C3575. Prompted by these observations, we have found that thousands of similar hydrophobicity-reducing variants associate specifically with DCM. Hence, our results imply that titin domain destabilization causes DCM, a conceptual framework that not only informs pathogenicity assessment of gene variants but also points to therapeutic strategies counterbalancing protein destabilization.
Collapse
Affiliation(s)
- Ines Martinez-Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| | - Audrey Crousilles
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, 22607 Hamburg, Germany
| | - Juan Pablo Ochoa
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHIM, CIBERCV, 28222 Madrid, Spain; Health in Code, 15008 A Coruña, Spain
| | | | - Simon A Mortensen
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, 22607 Hamburg, Germany
| | - Elias Herrero-Galan
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Javier Delgado
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Fernando Dominguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHIM, CIBERCV, 28222 Madrid, Spain
| | - Pablo Garcia-Pavia
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHIM, CIBERCV, 28222 Madrid, Spain
| | - David de Sancho
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU, 20018 Donostia-San Sebastian, Euskadi, Spain; Donostia International Physics Center (DIPC), 20018 Donostia-San Sebastian, Euskadi, Spain
| | - Matthias Wilmanns
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, 22607 Hamburg, Germany
| | | |
Collapse
|
57
|
Allouba M, Walsh R, Afify A, Hosny M, Halawa S, Galal A, Fathy M, Theotokis PI, Boraey A, Ellithy A, Buchan R, Govind R, Whiffin N, Anwer S, ElGuindy A, Ware JS, Barton PJR, Yacoub M, Aguib Y. Ethnicity, consanguinity, and genetic architecture of hypertrophic cardiomyopathy. Eur Heart J 2023; 44:5146-5158. [PMID: 37431535 PMCID: PMC10733735 DOI: 10.1093/eurheartj/ehad372] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/28/2023] [Accepted: 05/24/2023] [Indexed: 07/12/2023] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity that is partly explained by the diversity of genetic variants contributing to disease. Accurate interpretation of these variants constitutes a major challenge for diagnosis and implementing precision medicine, especially in understudied populations. The aim is to define the genetic architecture of HCM in North African cohorts with high consanguinity using ancestry-matched cases and controls. METHODS AND RESULTS Prospective Egyptian patients (n = 514) and controls (n = 400) underwent clinical phenotyping and genetic testing. Rare variants in 13 validated HCM genes were classified according to standard clinical guidelines and compared with a prospective HCM cohort of majority European ancestry (n = 684). A higher prevalence of homozygous variants was observed in Egyptian patients (4.1% vs. 0.1%, P = 2 × 10-7), with variants in the minor HCM genes MYL2, MYL3, and CSRP3 more likely to present in homozygosity than the major genes, suggesting these variants are less penetrant in heterozygosity. Biallelic variants in the recessive HCM gene TRIM63 were detected in 2.1% of patients (five-fold greater than European patients), highlighting the importance of recessive inheritance in consanguineous populations. Finally, rare variants in Egyptian HCM patients were less likely to be classified as (likely) pathogenic compared with Europeans (40.8% vs. 61.6%, P = 1.6 × 10-5) due to the underrepresentation of Middle Eastern populations in current reference resources. This proportion increased to 53.3% after incorporating methods that leverage new ancestry-matched controls presented here. CONCLUSION Studying consanguineous populations reveals novel insights with relevance to genetic testing and our understanding of the genetic architecture of HCM.
Collapse
Affiliation(s)
- Mona Allouba
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Roddy Walsh
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Alaa Afify
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mohammed Hosny
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Sarah Halawa
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Aya Galal
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mariam Fathy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Pantazis I Theotokis
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Ahmed Boraey
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Amany Ellithy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Rachel Buchan
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
| | - Risha Govind
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
- Present affiliation: National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Nicola Whiffin
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, UK
| | - Shehab Anwer
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Ahmed ElGuindy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Paul J R Barton
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Magdi Yacoub
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Harefield Heart Science Centre, Hill End Rd, Harefield, Uxbridge UB9 6JH, UK
| | - Yasmine Aguib
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| |
Collapse
|
58
|
Tarnovskaya SI, Kostareva AA, Zhorov BS. In silico analysis of TRPM4 variants of unknown clinical significance. PLoS One 2023; 18:e0295974. [PMID: 38100498 PMCID: PMC10723691 DOI: 10.1371/journal.pone.0295974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND TRPM4 is a calcium-activated channel that selectively permeates monovalent cations. Genetic variants of the channel in cardiomyocytes are associated with various heart disorders, such as progressive familial heart block and Brugada syndrome. About97% of all known TRPM4 missense variants are classified as variants of unknown clinical significance (VUSs). The very large number of VUSs is a serious problem in diagnostics and treatment of inherited heart diseases. METHODS AND RESULTS We collected 233 benign or pathogenic missense variants in the superfamily of TRP channels from databases ClinVar, Humsavar and Ensembl Variation to compare performance of 22 algorithms that predict damaging variants. We found that ClinPred is the best-performing tool for TRP channels. We also used the paralogue annotation method to identify disease variants across the TRP family. In the set of 565 VUSs of hTRPM4, ClinPred predicted pathogenicity of 299 variants. Among these, 12 variants are also categorized as LP/P variants in at least one paralogue of hTRPM4. We further used the cryo-EM structure of hTRPM4 to find scores of contact pairs between parental (wild type) residues of VUSs for which ClinPred predicts a high probability of pathogenicity of variants for both contact partners. We propose that 68 respective missense VUSs are also likely pathogenic variants. CONCLUSIONS ClinPred outperformed other in-silico tools in predicting damaging variants of TRP channels. ClinPred, the paralogue annotation method, and analysis of residue contacts the hTRPM4 cryo-EM structure collectively suggest pathogenicity of 80 TRPM4 VUSs.
Collapse
Affiliation(s)
- Svetlana I. Tarnovskaya
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Anna A. Kostareva
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Department of Women’s and Children’s Health and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Boris S. Zhorov
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| |
Collapse
|
59
|
Walsh R. The Trouble with Trabeculation: How Genetics Can Help to Unravel a Complex and Controversial Phenotype. J Cardiovasc Transl Res 2023; 16:1310-1324. [PMID: 38019448 DOI: 10.1007/s12265-023-10459-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Excessive trabeculation of the cardiac left ventricular wall is a complex phenotypic substrate associated with various physiological and pathological processes. There has been considerable conjecture as to whether hypertrabeculation contributes to disease and whether left ventricular non-compaction (LVNC) cardiomyopathy is a distinct pathology. Building on recent insights into the genetic basis of LVNC cardiomyopathy, in particular three meta-analysis studies exploring genotype-phenotype associations using different methodologies, this review examines how genetic research can advance our understanding of trabeculation. Three groups of genes implicated in LVNC are described-those associated with other cardiomyopathies, other cardiac/syndromic conditions and putatively with isolated LVNC cardiomyopathy-demonstrating how these findings can inform the underlying pathologies in LVNC patients and aid differential diagnosis and management in clinical practice despite the limited utility suggested for LVNC genetic testing in recent guidelines. The outstanding questions and future research priorities for exploring the genetics of hypertrabeculation are discussed.
Collapse
Affiliation(s)
- Roddy Walsh
- Department of Experimental Cardiology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
60
|
Bradford WH, Zhang J, Gutierrez-Lara EJ, Liang Y, Do A, Wang TM, Nguyen L, Mataraarachchi N, Wang J, Gu Y, McCulloch A, Peterson KL, Sheikh F. Plakophilin 2 gene therapy prevents and rescues arrhythmogenic right ventricular cardiomyopathy in a mouse model harboring patient genetics. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1246-1261. [PMID: 39196150 PMCID: PMC11357983 DOI: 10.1038/s44161-023-00370-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/16/2023] [Indexed: 08/29/2024]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a fatal genetic heart disease characterized by cardiac arrhythmias, in which fibrofatty deposition leads to heart failure, with no effective treatments. Plakophilin 2 (PKP2) is the most frequently mutated gene in ARVC, and although altered RNA splicing has been implicated, there are no models to study its effect and therapeutics. Here, we generate a mouse model harboring a PKP2 mutation (IVS10-1G>C) affecting RNA splicing, recapitulating ARVC features and sudden death starting at 4 weeks. Administering AAV-PKP2 gene therapy (adeno-associated viral therapy to drive cardiac expression of PKP2) to neonatal mice restored PKP2 protein levels, completely preventing cardiac desmosomal and pathological deficits associated with ARVC, ensuring 100% survival of mice up to 6 months. Late-stage AAV-PKP2 administration rescued desmosomal protein deficits and reduced pathological deficits including improved cardiac function in adult mice, resulting in 100% survival up to 4 months. We suggest that AAV-PKP2 gene therapy holds promise for circumventing ARVC associated with PKP2 mutations, including splice site mutations.
Collapse
Affiliation(s)
- William H Bradford
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Yan Liang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Aryanne Do
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tsui-Min Wang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Lena Nguyen
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Jie Wang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Kirk L Peterson
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
61
|
Zahavich L, Akilen R, George K, Mital S. Heart Failure with Recovered Ejection Fraction in Patients with Vinculin Loss-of-function Variants. J Cardiovasc Transl Res 2023; 16:1303-1309. [PMID: 37548861 PMCID: PMC10721703 DOI: 10.1007/s12265-023-10421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023]
Abstract
Predictors of myocardial recovery in heart failure (HF) are poorly understood. We explored if vinculin (VCL) variants are associated with myocardial recovery in dilated cardiomyopathy (DCM). Six infants with DCM with a VCL loss-of-function (LOF) variant were identified. Median age at diagnosis was 2 months, median LV ejection fraction was 24%, and median LV end-diastolic diameter z-score was 10.8. All patients received HF medications. Five patients (83%) showed normalization of LV function at a median age of 2.7 years. One patient progressed to end-stage HF requiring heart transplant. This case series identified a unique phenotype of HF with reduced ejection fraction at presentation that evolved to HF with recovered EF in over 80% of infant DCM cases with LOF VCL variants. These findings have prognostic implications for counseling and management of VCL-associated DCM and highlight a possible genetic basis for HF with recovered ejection fraction.
Collapse
Affiliation(s)
- Laura Zahavich
- Department of Genetic Counselling, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Labatt Family Heart Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rajadurai Akilen
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kristen George
- Labatt Family Heart Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Seema Mital
- Labatt Family Heart Centre, Hospital for Sick Children, Toronto, Ontario, Canada.
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.
| |
Collapse
|
62
|
Costa M, García S A, Pastor O. The consequences of data dispersion in genomics: a comparative analysis of data sources for precision medicine. BMC Med Inform Decis Mak 2023; 23:256. [PMID: 37946154 PMCID: PMC10636939 DOI: 10.1186/s12911-023-02342-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Genomics-based clinical diagnosis has emerged as a novel medical approach to improve diagnosis and treatment. However, advances in sequencing techniques have increased the generation of genomics data dramatically. This has led to several data management problems, one of which is data dispersion (i.e., genomics data is scattered across hundreds of data repositories). In this context, geneticists try to remediate the above-mentioned problem by limiting the scope of their work to a single data source they know and trust. This work has studied the consequences of focusing on a single data source rather than considering the many different existing genomics data sources. METHODS The analysis is based on the data associated with two groups of disorders (i.e., oncology and cardiology) accessible from six well-known genomic data sources (i.e., ClinVar, Ensembl, GWAS Catalog, LOVD, CIViC, and CardioDB). Two dimensions have been considered in this analysis, namely, completeness and concordance. Completeness has been evaluated at two levels. First, by analyzing the information provided by each data source with regard to a conceptual schema data model (i.e., the schema level). Second, by analyzing the DNA variations provided by each data source as related to any of the disorders selected (i.e., the data level). Concordance has been evaluated by comparing the consensus among the data sources regarding the clinical relevance of each variation and disorder. RESULTS The data sources with the highest completeness at the schema level are ClinVar, Ensembl, and CIViC. ClinVar has the highest completeness at the data level data source for the oncology and cardiology disorders. However, there are clinically relevant variations that are exclusive to other data sources, and they must be considered in order to provide the best clinical diagnosis. Although the information available in the data sources is predominantly concordant, discordance among the analyzed data exist. This can lead to inaccurate diagnoses. CONCLUSION Precision medicine analyses using a single genomics data source leads to incomplete results. Also, there are concordance problems that threaten the correctness of the genomics-based diagnosis results.
Collapse
Affiliation(s)
- Mireia Costa
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain.
| | - Alberto García S
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain
| | - Oscar Pastor
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain
| |
Collapse
|
63
|
Chumakova OS, Baklanova TN, Milovanova NV, Zateyshchikov DA. Hypertrophic Cardiomyopathy in Underrepresented Populations: Clinical and Genetic Landscape Based on a Russian Single-Center Cohort Study. Genes (Basel) 2023; 14:2042. [PMID: 38002985 PMCID: PMC10671745 DOI: 10.3390/genes14112042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited cardiac disorder characterized by marked clinical and genetic heterogeneity. Ethnic groups underrepresented in studies may have distinctive characteristics. We sought to evaluate the clinical and genetic landscape of Russian HCM patients. A total of 193 patients (52% male; 95% Eastern Slavic origin; median age 56 years) were clinically evaluated, including genetic testing, and prospectively followed to document outcomes. As a result, 48% had obstructive HCM, 25% had HCM in family, 21% were asymptomatic, and 68% had comorbidities. During 2.8 years of follow-up, the all-cause mortality rate was 2.86%/year. A total of 5.7% received an implantable cardioverter-defibrillator (ICD), and 21% had septal reduction therapy. A sequencing analysis of 176 probands identified 64 causative variants in 66 patients (38%); recurrent variants were MYBPC3 p.Q1233* (8), MYBPC3 p.R346H (2), MYH7 p.A729P (2), TPM1 p.Q210R (3), and FLNC p.H1834Y (2); 10 were multiple variant carriers (5.7%); 5 had non-sarcomeric HCM, ALPK3, TRIM63, and FLNC. Thin filament variant carriers had a worse prognosis for heart failure (HR = 7.9, p = 0.007). In conclusion, in the Russian HCM population, the low use of ICD and relatively high mortality should be noted by clinicians; some distinct recurrent variants are suspected to have a founder effect; and family studies on some rare variants enriched worldwide knowledge in HCM.
Collapse
Affiliation(s)
- Olga S. Chumakova
- Moscow Healthcare Department, City Clinical Hospital 17, 119620 Moscow, Russia; (T.N.B.); (D.A.Z.)
- E.I. Chazov National Medical Research Center for Cardiology, 121552 Moscow, Russia
| | - Tatiana N. Baklanova
- Moscow Healthcare Department, City Clinical Hospital 17, 119620 Moscow, Russia; (T.N.B.); (D.A.Z.)
| | | | - Dmitry A. Zateyshchikov
- Moscow Healthcare Department, City Clinical Hospital 17, 119620 Moscow, Russia; (T.N.B.); (D.A.Z.)
- E.I. Chazov National Medical Research Center for Cardiology, 121552 Moscow, Russia
| |
Collapse
|
64
|
García-Vielma C, Lazalde-Córdova LG, Arzola-Hernández JC, González-Aceves EN, López-Zertuche H, Guzmán-Delgado NE, González-Salazar F. Identification of variants in genes associated with hypertrophic cardiomyopathy in Mexican patients. Mol Genet Genomics 2023; 298:1289-1299. [PMID: 37498360 PMCID: PMC10657276 DOI: 10.1007/s00438-023-02048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
The objective of this work was to identify genetic variants in Mexican patients diagnosed with hypertrophic cardiomyopathy (HCM). According to world literature, the genes mainly involved are MHY7 and MYBPC3, although variants have been found in more than 50 genes related to heart disease and sudden death, and to our knowledge there are no studies in the Mexican population. These variants are reported and classified in the ClinVar (PubMed) database and only some of them are recognized in the Online Mendelian Information in Men (OMIM). The present study included 37 patients, with 14 sporadic cases and 6 familial cases, with a total of 21 index cases. Next-generation sequencing was performed on a predesigned panel of 168 genes associated with heart disease and sudden death. The sequencing analysis revealed twelve (57%) pathogenic or probably pathogenic variants, 9 of them were familial cases, managing to identify pathogenic variants in relatives without symptoms of the disease. At the molecular level, nine of the 12 variants (75%) were single nucleotide changes, 2 (17%) deletions, and 1 (8%) splice site alteration. The genes involved were MYH7 (25%), MYBPC3 (25%) and ACADVL, KCNE1, TNNI3, TPM1, SLC22A5, TNNT2 (8%). In conclusion; we found five variants that were not previously reported in public databases. It is important to follow up on the reclassification of variants, especially those of uncertain significance in patients with symptoms of the condition. All patients included in the study and their relatives received family genetic counseling.
Collapse
Affiliation(s)
- Catalina García-Vielma
- Centro de Investigación Biomédica del Noreste, Departamento de Citogenética, Instituto Mexicano del Seguro Social, Monterrey, NL, México.
| | - Luis Gerardo Lazalde-Córdova
- Centro de Investigación Biomédica del Noreste, Departamento de Citogenética, Instituto Mexicano del Seguro Social, Monterrey, NL, México
| | - José Cruz Arzola-Hernández
- Departamento de Electrofisiología, Instituto Mexicano del Seguro Social, Unidad Médica de Alta Especialidad. Hospital de cardiología No. 34 "Dr. Alfonso J. Treviño Treviño" del Centro Médico Nacional del Noreste, Monterrey, NL, México
| | - Erick Noel González-Aceves
- Departamento de Electrofisiología, Instituto Mexicano del Seguro Social, Unidad Médica de Alta Especialidad. Hospital de cardiología No. 34 "Dr. Alfonso J. Treviño Treviño" del Centro Médico Nacional del Noreste, Monterrey, NL, México
| | | | - Nancy Elena Guzmán-Delgado
- Departamento de Electrofisiología, Instituto Mexicano del Seguro Social, Unidad Médica de Alta Especialidad. Hospital de cardiología No. 34 "Dr. Alfonso J. Treviño Treviño" del Centro Médico Nacional del Noreste, Monterrey, NL, México.
| | - Francisco González-Salazar
- Centro de Investigación Biomédica del Noreste, Departamento de Citogenética, Instituto Mexicano del Seguro Social, Monterrey, NL, México
| |
Collapse
|
65
|
Kim MJ, Cha S, Baek JS, Yu JJ, Seo GH, Kang M, Do HS, Lee SE, Lee BH. Genetic heterogeneity of cardiomyopathy and its correlation with patient care. BMC Med Genomics 2023; 16:270. [PMID: 37904158 PMCID: PMC10614404 DOI: 10.1186/s12920-023-01639-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/21/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Cardiomyopathy, which is a genetically and phenotypically heterogeneous pathological condition, is associated with increased morbidity and mortality. Genetic diagnosis of cardiomyopathy enables accurate phenotypic classification and optimum patient management and counseling. This study investigated the genetic spectrum of cardiomyopathy and its correlation with the clinical course of the disease. METHODS The samples of 72 Korean patients with cardiomyopathy (43 males and 29 females) were subjected to whole-exome sequencing (WES). The familial information and clinical characteristics of the patients were reviewed and analyzed according to their genotypes. RESULTS Dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), left ventricular non-compaction cardiomyopathy, and restrictive cardiomyopathy was detected in 41 (56.9%), 25 (34.7%), 4 (5.6%), and 2 (2.8%) patients, respectively. WES analysis revealed positive results in 37 (51.4%) patients. Subsequent familial testing identified ten additional familial cases. Among DCM cases, 19 (46.3%) patients exhibited positive results, with TTN variants being the most common alteration, followed by LMNA and MYH7 variants. Meanwhile, among HCM cases, 15 (60%) patients exhibited positive results with MYH7 variants being the most common alteration. In six patients with positive results, extracardiac surveillance was warranted based on disease information. The incidence of worse outcomes, such as mortality and life-threatening arrhythmic events, in patients with DCM harboring LMNA variants, was higher than that in patients with DCM harboring TTN or MYH7 variants. CONCLUSIONS Diverse genotypes were identified in a substantial proportion of patients with cardiomyopathy. Genetic diagnosis enables personalized disease surveillance and management.
Collapse
Affiliation(s)
- Mi Jin Kim
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seulgi Cha
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Suk Baek
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Jin Yu
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | - Minji Kang
- Genome Research Center for Birth Defects and Genetic Diseases, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Hyo-Sang Do
- Genome Research Center for Birth Defects and Genetic Diseases, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Sang Eun Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Beom Hee Lee
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicines, Seoul, Korea.
| |
Collapse
|
66
|
Bhaskara GB, Haque T, Bonnette JE, Napier JD, Bauer D, Schmutz J, Juenger TE. Evolutionary Analyses of Gene Expression Divergence in Panicum hallii: Exploring Constitutive and Plastic Responses Using Reciprocal Transplants. Mol Biol Evol 2023; 40:msad210. [PMID: 37738160 PMCID: PMC10556983 DOI: 10.1093/molbev/msad210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
The evolution of gene expression is thought to be an important mechanism of local adaptation and ecological speciation. Gene expression divergence occurs through the evolution of cis- polymorphisms and through more widespread effects driven by trans-regulatory factors. Here, we explore expression and sequence divergence in a large sample of Panicum hallii accessions encompassing the species range using a reciprocal transplantation experiment. We observed widespread genotype and transplant site drivers of expression divergence, with a limited number of genes exhibiting genotype-by-site interactions. We used a modified FST-QST outlier approach (QPC analysis) to detect local adaptation. We identified 514 genes with constitutive expression divergence above and beyond the levels expected under neutral processes. However, no plastic expression responses met our multiple testing correction as QPC outliers. Constitutive QPC outlier genes were involved in a number of developmental processes and responses to abiotic environments. Leveraging earlier expression quantitative trait loci results, we found a strong enrichment of expression divergence, including for QPC outliers, in genes previously identified with cis and cis-environment interactions but found no patterns related to trans-factors. Population genetic analyses detected elevated sequence divergence of promoters and coding sequence of constitutive expression outliers but little evidence for positive selection on these proteins. Our results are consistent with a hypothesis of cis-regulatory divergence as a primary driver of expression divergence in P. hallii.
Collapse
Affiliation(s)
| | - Taslima Haque
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Jason E Bonnette
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Joseph D Napier
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Diane Bauer
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Jeremy Schmutz
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Genome Sequencing Center, HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Thomas E Juenger
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
67
|
Asatryan B, Bleijendaal H, Wilde AAM. Toward advanced diagnosis and management of inherited arrhythmia syndromes: Harnessing the capabilities of artificial intelligence and machine learning. Heart Rhythm 2023; 20:1399-1407. [PMID: 37442407 DOI: 10.1016/j.hrthm.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
The use of advanced computational technologies, such as artificial intelligence (AI), is now exerting a significant influence on various aspects of life, including health care and science. AI has garnered remarkable public notice with the release of deep learning models that can model anything from artwork to academic papers with minimal human intervention. Machine learning, a method that uses algorithms to extract information from raw data and represent it in a model, and deep learning, a method that uses multiple layers to progressively extract higher-level features from the raw input with minimal human intervention, are increasingly leveraged to tackle problems in the health sector, including utilization for clinical decision support in cardiovascular medicine. Inherited arrhythmia syndromes are a clinical domain where multiple unanswered questions remain despite unprecedented progress over the past 2 decades with the introduction of large panel genetic testing and the first steps in precision medicine. In particular, AI tools can help address gaps in clinical diagnosis by identifying individuals with concealed or transient phenotypes; enhance risk stratification by elevating recognition of underlying risk burden beyond widely recognized risk factors; improve prediction of response to therapy, and further prognostication. In this contemporary review, we provide a summary of the AI models developed to solve challenges in inherited arrhythmia syndromes and also outline gaps that can be filled with the development of intelligent AI models.
Collapse
Affiliation(s)
- Babken Asatryan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Hidde Bleijendaal
- University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands; Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur A M Wilde
- University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands; Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, The Netherlands; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart)
| |
Collapse
|
68
|
Kim Y, Gunnarsdóttir OB, Viveiros A, Reichart D, Quiat D, Willcox JAL, Zhang H, Chen H, Curran JJ, Kim DH, Urschel S, McDonough B, Gorham J, DePalma SR, Seidman JG, Seidman CE, Oudit GY. Genetic Contribution to End-Stage Cardiomyopathy Requiring Heart Transplantation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:452-461. [PMID: 37767697 DOI: 10.1161/circgen.123.004062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Many cardiovascular disorders propel the development of advanced heart failure that necessitates cardiac transplantation. When treatable causes are excluded, studies to define causes are often abandoned, resulting in a diagnosis of end-stage idiopathic cardiomyopathy. We studied whether DNA sequence analyses could identify unrecognized causes of end-stage nonischemic cardiomyopathy requiring heart transplantation and whether the prevalence of genetic causes differed from ambulatory cardiomyopathy cases. METHODS We performed whole exome and genome sequencing of 122 explanted hearts from 101 adult and 21 pediatric patients with idiopathic cardiomyopathy from a single center. Data were analyzed for pathogenic/likely pathogenic variants in nuclear and mitochondrial genomes and assessed for nonhuman microbial sequences. The frequency of damaging genetic variants was compared among cardiomyopathy cohorts with different clinical severity. RESULTS Fifty-four samples (44.3%) had pathogenic/likely pathogenic cardiomyopathy gene variants. The frequency of pathogenic variants was similar in pediatric (42.9%) and adult (43.6%) samples, but the distribution of mutated genes differed (P=8.30×10-4). The prevalence of causal genetic variants was significantly higher in end-stage than in previously reported ambulatory adult dilated cardiomyopathy cases (P<0.001). Among remaining samples with unexplained causes, no damaging mitochondrial variants were identified, but 28 samples contained parvovirus genome sequences, including 2 samples with 6- to 9-fold higher levels than the overall mean levels in other samples. CONCLUSIONS Pathogenic variants and viral myocarditis were identified in 45.9% of patients with unexplained end-stage cardiomyopathy. Damaging gene variants are significantly more frequent among transplant compared with patients with ambulatory cardiomyopathy. Genetic analyses can help define cause of end-stage cardiomyopathy to guide management and risk stratification of patients and family members.
Collapse
Affiliation(s)
- Yuri Kim
- Division of Cardiovascular Medicine, Brigham and Women's Hospital (Y.K., B.M., C.E.S.)
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Oddný Brattberg Gunnarsdóttir
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Anissa Viveiros
- Department of Medicine (A.V., H.Z., H.C., D.H.K., G.Y.O.), University of Alberta
- Mazankowski Alberta Heart Institute, Edmonton, Canada (A.V., H.Z., H.C., D.H.K., G.Y.O.)
| | - Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
- Department of Medicine I, University Hospital, Ludwig Maximilian University of Munich, Germany (D.R.)
| | - Daniel Quiat
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
- Department of Cardiology, Boston Children's Hospital, MA (D.Q.)
| | - Jon A L Willcox
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Hao Zhang
- Department of Medicine (A.V., H.Z., H.C., D.H.K., G.Y.O.), University of Alberta
- Mazankowski Alberta Heart Institute, Edmonton, Canada (A.V., H.Z., H.C., D.H.K., G.Y.O.)
| | - Huachen Chen
- Department of Medicine (A.V., H.Z., H.C., D.H.K., G.Y.O.), University of Alberta
- Mazankowski Alberta Heart Institute, Edmonton, Canada (A.V., H.Z., H.C., D.H.K., G.Y.O.)
| | - Justin J Curran
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Daniel H Kim
- Department of Medicine (A.V., H.Z., H.C., D.H.K., G.Y.O.), University of Alberta
- Mazankowski Alberta Heart Institute, Edmonton, Canada (A.V., H.Z., H.C., D.H.K., G.Y.O.)
| | - Simon Urschel
- Department of Pediatrics (S.U.), University of Alberta
- Stollery Children's Hospital, Edmonton, Alberta, Canada (S.U.)
| | - Barbara McDonough
- Division of Cardiovascular Medicine, Brigham and Women's Hospital (Y.K., B.M., C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (B.M., S.R.D., C.E.S.)
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Steven R DePalma
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (B.M., S.R.D., C.E.S.)
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
| | - Christine E Seidman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital (Y.K., B.M., C.E.S.)
- Department of Genetics, Harvard Medical School, Boston, MA (Y.K., O.B.G., D.R., D.Q., J.A.L.W., J.J.C., J.G., S.R.D., J.G.S., C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (B.M., S.R.D., C.E.S.)
| | - Gavin Y Oudit
- Department of Medicine (A.V., H.Z., H.C., D.H.K., G.Y.O.), University of Alberta
- Mazankowski Alberta Heart Institute, Edmonton, Canada (A.V., H.Z., H.C., D.H.K., G.Y.O.)
| |
Collapse
|
69
|
Al-Ghamdi BS, Alhadeq F, Alqahtani A, Alruwaili N, Rababh M, Alghamdi S, Almanea W, Alhassnan Z. Clinical and Genetic Characteristics of Arrhythmogenic Right Ventricular Cardiomyopathy Patients: A Single-Center Experience. Cardiol Res 2023; 14:379-386. [PMID: 37936624 PMCID: PMC10627368 DOI: 10.14740/cr1531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/22/2023] [Indexed: 11/09/2023] Open
Abstract
Background Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited progressive cardiomyopathy. We aimed to define the long-term clinical outcome and genetic characteristics of patients and family members with positive genetic tests for ARVC in a single tertiary care cardiac center in Saudi Arabia. Methods We enrolled 46 subjects in the study, including 23 index-patients (probands) with ARVC based on the revised 2010 ARVC Task Force Criteria (TFC) and 23 family members who underwent a genetic test for the ARVC between 2016 and 2020. Results Of the probands, 17 (73.9%) were males with a mean age at presentation of 24.95 ± 13.9 years (7 to 55 years). Predominant symptoms were palpitations in 14 patients (60.9%), and syncope in 10 patients (43.47%). Sustained ventricular tachycardia (VT) was documented in 12 patients (52.2%). The mean left ventricular ejection fraction (LVEF) by echocardiogram was 52.81±6.311% (30-55%), and the mean right ventricular ejection fraction (RVEF) by cardiac MRI was 41.3±11.37% (23-64%). Implantable cardioverter-defibrillator (ICD) implantation was performed in 17 patients (73.9%), and over a mean follow-up of 13.65 ± 6.83 years, appropriate ICD therapy was noted in 12 patients (52.2%). Genetic variants were identified in 33 subjects (71.7%), 16 patients and 17 family members, with the most common variant of plakophilin 2 (PKP2) in 27 subjects (81.8%). Conclusions ARVC occurs during early adulthood in Saudi patients. It is associated with a significant arrhythmia burden in these patients. The PKP2 gene is the most common gene defect in Saudi patients, consistent with what is observed in other nations. We reported in this study two novel variants in PKP2 and desmocollin 2 (DSC2) genes. Genetic counseling is needed to include all first-degree family members for early diagnosis and management of the disease in our country.
Collapse
Affiliation(s)
- Bandar Saeed Al-Ghamdi
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Faten Alhadeq
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| | - Aisha Alqahtani
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| | - Nadiah Alruwaili
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
| | | | | | - Waleed Almanea
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
- Pediatric Cardiology, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Zuhair Alhassnan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| |
Collapse
|
70
|
Tan K, Foo R, Loh M. Cardiomyopathy in Asian Cohorts: Genetic and Epigenetic Insights. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:496-506. [PMID: 37589150 DOI: 10.1161/circgen.123.004079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Previous studies on cardiomyopathies have been particularly valuable for clarifying pathological mechanisms in heart failure, an etiologically heterogeneous disease. In this review, we specifically focus on cardiomyopathies in Asia, where heart failure is particularly pertinent. There has been an increase in prevalence of cardiomyopathies in Asia, in sharp contrast with the decline observed in Western countries. Indeed, important disparities in cardiomyopathy incidence, clinical characteristics, and prognosis have been reported in Asian versus White cohorts. These have been accompanied by emerging descriptions of a distinct rare and common genetic basis for disease among Asian cardiomyopathy patients marked by an increased burden of variants with uncertain significance, reclassification of variants deemed pathogenic based on evidence from predominantly White cohorts, and the discovery of Asian-specific cardiomyopathy-associated loci with underappreciated pathogenicity under conventional classification criteria. Findings from epigenetic studies of heart failure, particularly DNA methylation studies, have complemented genetic findings in accounting for the phenotypic variability in cardiomyopathy. Though extremely limited, findings from Asian ancestry-focused DNA methylation studies of cardiomyopathy have shown potential to contribute to general understanding of cardiomyopathy pathophysiology by proposing disease and cause-relevant pathophysiological mechanisms. We discuss the value of multiomics study designs incorporating genetic, methylation, and transcriptomic information for future DNA methylation studies in Asian cardiomyopathy cohorts to yield Asian ancestry-specific insights that will improve risk stratification in the Asian population.
Collapse
Affiliation(s)
- Konstanze Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore (R.F.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore (R.F.)
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
- Genome Institute of Singapore, Singapore (GIS), Agency for Science, Technology and Research (A*STAR) (M.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (M.L.)
- National Skin Centre, Singapore (M.L.)
| |
Collapse
|
71
|
Alaei Z, Zamani N, Rabbani B, Mahdieh N. TCAP gene is not a common cause of cardiomyopathy in Iranian patients. Eur J Med Res 2023; 28:376. [PMID: 37752589 PMCID: PMC10523715 DOI: 10.1186/s40001-023-01019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most frequent cardiomyopathies that cause acute heart failure and sudden cardiac death. Previous genetic reports have shown that pathogenic variants of genes encoding Z-disc components such as telethonin protein (TCAP) are the primary cause of DCM and HCM. METHODS This study was the first investigation on the TCAP gene among the Iranian cardiomyopathies population wherein the TCAP gene was analyzed in 40 unrelated patients (17 females and 23 males) who were clinically diagnosed with HCM and DCM. In addition, we conducted a thorough review of all published articles and the databases that were the first to report novel pathogenic or likely pathogenic variants the in TCAP gene. RESULTS In the cohort of this study, we identified only one intronic variant c.111-42G > A in one of the HCM patients that were predicted as polymorphism by in-silico analysis. Moreover, a total of 44 variants were reported for the TCAP gene in the literature where a majority of mutations were found to be missense. Pathogenic mutations in TCAP may cause diseases including limb-girdle muscular dystrophy 2G (LGMD-2G), DCM, HCM, intestinal pseudo-obstruction, and telethonin deficiency. However, a large number of affected patients were clinically diagnosed with limb-girdle 2G compared to other presenting phenotypes. DISCUSSION These findings suggest that the TCAP gene pathogenic mutations might not be a common cause of cardiomyopathies among Iranian patients. These gene disease-causing mutations may cause various manifestations, but it has a high prevalence among LGMD-2G, HCM, and DCM patients.
Collapse
Affiliation(s)
- Zahra Alaei
- Faculty of Basic Sciences, Islamic Azad University, East Tehran Branch, Tehran, Iran
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Zamani
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Rabbani
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nejat Mahdieh
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Genetics Laboratory, Rajaie Cardiovascular Medical and Research Center, Vali-E-Asr Avenue, Tehran, 1996911151, Iran.
| |
Collapse
|
72
|
Hayesmoore JB, Bhuiyan ZA, Coviello DA, du Sart D, Edwards M, Iascone M, Morris-Rosendahl DJ, Sheils K, van Slegtenhorst M, Thomson KL. EMQN: Recommendations for genetic testing in inherited cardiomyopathies and arrhythmias. Eur J Hum Genet 2023; 31:1003-1009. [PMID: 37443332 PMCID: PMC10474043 DOI: 10.1038/s41431-023-01421-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Inherited cardiomyopathies and arrhythmias (ICAs) are a prevalent and clinically heterogeneous group of genetic disorders that are associated with increased risk of sudden cardiac death and heart failure. Making a genetic diagnosis can inform the management of patients and their at-risk relatives and, as such, molecular genetic testing is now considered an integral component of the clinical care pathway. However, ICAs are characterised by high genetic and allelic heterogeneity, incomplete / age-related penetrance, and variable expressivity. Therefore, despite our improved understanding of the genetic basis of these conditions, and significant technological advances over the past two decades, identifying and recognising the causative genotype remains challenging. As clinical genetic testing for ICAs becomes more widely available, it is increasingly important for clinical laboratories to consolidate existing knowledge and experience to inform and improve future practice. These recommendations have been compiled to help clinical laboratories navigate the challenges of ICAs and thereby facilitate best practice and consistency in genetic test provision for this group of disorders. General recommendations on internal and external quality control, referral, analysis, result interpretation, and reporting are described. Also included are appendices that provide specific information pertinent to genetic testing for hypertrophic, dilated, and arrhythmogenic right ventricular cardiomyopathies, long QT syndrome, Brugada syndrome, and catecholaminergic polymorphic ventricular tachycardia.
Collapse
Affiliation(s)
- Jesse B Hayesmoore
- Oxford Regional Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Zahurul A Bhuiyan
- Division of Genetic Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Desirée du Sart
- Biological Sciences and Genomics, Monash University, Melbourne, VIC, Australia
| | - Matthew Edwards
- Clinical Genetics and Genomics Laboratory, Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Maria Iascone
- Laboratorio di Genetica Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics Laboratory, Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | | | - Kate L Thomson
- Oxford Regional Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
73
|
Thompson AD, Wagner MJ, Rodriguez J, Malhotra A, Vander Roest S, Lilienthal U, Shao H, Vignesh M, Weber K, Yob JM, Prosser BL, Helms AS, Gestwicki JE, Ginsburg D, Day SM. An Unbiased Screen Identified the Hsp70-BAG3 Complex as a Regulator of Myosin-Binding Protein C3. JACC Basic Transl Sci 2023; 8:1198-1211. [PMID: 37791314 PMCID: PMC10544073 DOI: 10.1016/j.jacbts.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 10/05/2023]
Abstract
Variants in the gene myosin-binding protein C3 (MYBPC3) account for approximately 50% of familial hypertrophic cardiomyopathy (HCM), leading to reduced levels of myosin-binding protein C3 (MyBP-C), the protein product made by gene MYBPC3. Elucidation of the pathways that regulate MyBP-C protein homeostasis could uncover new therapeutic strategies. Toward this goal, we screened a library of 2,426 bioactive compounds and identified JG98, an allosteric modulator of heat shock protein 70 that inhibits interaction with Bcl-2-associated athanogene (BAG) domain co-chaperones. JG98 reduces MyBP-C protein levels. Furthermore, genetic reduction of BAG3 phenocopies treatment with JG-98 by reducing MYBP-C protein levels.. Thus, an unbiased compound screen identified the heat shock protein 70-BAG3 complex as a regulator of MyBP-C stability.
Collapse
Affiliation(s)
- Andrea D. Thompson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Marcus J. Wagner
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juliani Rodriguez
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alok Malhotra
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Steve Vander Roest
- Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Ulla Lilienthal
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Mathav Vignesh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Keely Weber
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jaime M. Yob
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin L. Prosser
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam S. Helms
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason E. Gestwicki
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - David Ginsburg
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
- The Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharlene M. Day
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
74
|
Foreman J, Perrett D, Mazaika E, Hunt SE, Ware JS, Firth HV. DECIPHER: Improving Genetic Diagnosis Through Dynamic Integration of Genomic and Clinical Data. Annu Rev Genomics Hum Genet 2023; 24:151-176. [PMID: 37285546 PMCID: PMC7615097 DOI: 10.1146/annurev-genom-102822-100509] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
DECIPHER (Database of Genomic Variation and Phenotype in Humans Using Ensembl Resources) shares candidate diagnostic variants and phenotypic data from patients with genetic disorders to facilitate research and improve the diagnosis, management, and therapy of rare diseases. The platform sits at the boundary between genomic research and the clinical community. DECIPHER aims to ensure that the most up-to-date data are made rapidly available within its interpretation interfaces to improve clinical care. Newly integrated cardiac case-control data that provide evidence of gene-disease associations and inform variant interpretation exemplify this mission. New research resources are presented in a format optimized for use by a broad range of professionals supporting the delivery of genomic medicine. The interfaces within DECIPHER integrate and contextualize variant and phenotypic data, helping to determine a robust clinico-molecular diagnosis for rare-disease patients, which combines both variant classification and clinical fit. DECIPHER supports discovery research, connecting individuals within the rare-disease community to pursue hypothesis-driven research.
Collapse
Affiliation(s)
- Julia Foreman
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom; ,
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Daniel Perrett
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom; ,
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Erica Mazaika
- National Heart and Lung Institute and MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom; ,
| | - Sarah E Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom; ,
| | - James S Ware
- National Heart and Lung Institute and MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom; ,
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Helen V Firth
- Wellcome Sanger Institute, Hinxton, United Kingdom
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom;
| |
Collapse
|
75
|
Koshy L, Ganapathi S, Jeemon P, Madhuma M, Vysakh Y, Lakshmikanth L, Harikrishnan S. Sarcomeric gene variants among Indians with hypertrophic cardiomyopathy: A scoping review. Indian J Med Res 2023; 158:119-135. [PMID: 37787257 PMCID: PMC10645028 DOI: 10.4103/ijmr.ijmr_3567_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 10/04/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic heart muscle disease that frequently causes sudden cardiac death (SCD) among young adults. Several pathogenic mutations in genes encoding the cardiac sarcomere have been identified as diagnostic factors for HCM and proposed as prognostic markers for SCD. The objective of this review was to determine the scope of available literature on the variants encoding sarcomere proteins associated with SCD reported among Indian patients with HCM. The eligibility criteria for the scoping review included full text articles that reported the results of genetic screening for sarcomeric gene mutations in HCM patients of Indian south Asian ancestry. We systematically reviewed studies from the databases of Medline, Scopus, Web of Science core collection and Google Scholar. The electronic search strategy included a combination of generic terms related to genetics, disease and population. The protocol of the study was registered with Open Science Framework (https://osf.io/53gde/). A total of 19 articles were identified that reported pathogenic or likely pathogenic (P/LP) variants within MYH7, MYBPC3, TNNT2, TNNI3 and TPM1 genes, that included 16 singletons, one de novo and one digenic mutation (MYH7/ TPM1) associated with SCD among Indian patients. Evidence from functional studies and familial segregation implied a plausible mechanistic role of these P/LP variants in HCM pathology. This scoping review has compiled all the P/LP variants reported to-date among Indian patients and summarized their association with SCD. Single homozygous, de novo and digenic mutations were observed to be associated with severe phenotypes compared to single heterozygous mutations. The abstracted genetic information was updated with reference sequence ID (rsIDs) and compiled into freely accessible HCMvar database, available at https://hcmvar.heartfailure.org.in/. This can be used as a population specific genetic database for reference by clinicians and researchers involved in the identification of diagnostic and prognostic markers for HCM.
Collapse
Affiliation(s)
- Linda Koshy
- Centre for Advance Research & Excellence in Heart Failure, Thiruvananthapuram, Kerala, India
| | - Sanjay Ganapathi
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Panniyammakal Jeemon
- Achutha Menon Centre for Health Science Studies, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - M. Madhuma
- Centre for Advance Research & Excellence in Heart Failure, Thiruvananthapuram, Kerala, India
| | - Y. Vysakh
- Centre for Advance Research & Excellence in Heart Failure, Thiruvananthapuram, Kerala, India
| | - L.R. Lakshmikanth
- Centre for Advance Research & Excellence in Heart Failure, Thiruvananthapuram, Kerala, India
| | - Sivadasanpillai Harikrishnan
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
76
|
Chumakova OS, Baulina NM. Advanced searching for hypertrophic cardiomyopathy heritability in real practice tomorrow. Front Cardiovasc Med 2023; 10:1236539. [PMID: 37583586 PMCID: PMC10425241 DOI: 10.3389/fcvm.2023.1236539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease associated with morbidity and mortality at any age. As studies in recent decades have shown, the genetic architecture of HCM is quite complex both in the entire population and in each patient. In the rapidly advancing era of gene therapy, we have to provide a detailed molecular diagnosis to our patients to give them the chance for better and more personalized treatment. In addition to emphasizing the importance of genetic testing in routine practice, this review aims to discuss the possibility to go a step further and create an expanded genetic panel that contains not only variants in core genes but also new candidate genes, including those located in deep intron regions, as well as structural variations. It also highlights the benefits of calculating polygenic risk scores based on a combination of rare and common genetic variants for each patient and of using non-genetic HCM markers, such as microRNAs that can enhance stratification of risk for HCM in unselected populations alongside rare genetic variants and clinical factors. While this review is focusing on HCM, the discussed issues are relevant to other cardiomyopathies.
Collapse
Affiliation(s)
- Olga S. Chumakova
- Laboratory of Functional Genomics of Cardiovascular Diseases, National Medical Research Centre of Cardiology Named After E.I. Chazov, Moscow, Russia
| | | |
Collapse
|
77
|
Ostrominski JW, Guo R, Elliott PM, Ho CY. Cardiac Myosin Inhibitors for Managing Obstructive Hypertrophic Cardiomyopathy: JACC: Heart Failure State-of-the-Art Review. JACC. HEART FAILURE 2023; 11:735-748. [PMID: 37407153 DOI: 10.1016/j.jchf.2023.04.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 07/07/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is frequently caused by pathogenic variants in genes encoding sarcomere proteins and is characterized by left ventricular (LV) hypertrophy, hypercontractility, and-in many cases-left ventricular outflow tract (LVOT) obstruction. Despite standard management, obstructive HCM (oHCM) can still cause substantial morbidity, highlighting the critical need for more effective disease-specific therapeutic approaches. Over the past decade, improved understanding of the molecular pathobiology of HCM has culminated in development of cardiac myosin inhibitors (CMIs), a novel drug class that in recent randomized clinical trials has been shown to decrease LVOT obstruction, improve exercise capacity, and ameliorate symptom burden in patients with oHCM. Although promising, areas of uncertainty remain, including the long-term safety and efficacy of CMIs and whether they have the potential to modify progression of disease. Herein, we review key milestones in the clinical development of CMIs, contextualize CMIs with established oHCM therapies, and discuss future challenges and opportunities for the use of CMIs across the HCM spectrum.
Collapse
Affiliation(s)
- John W Ostrominski
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ruby Guo
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Perry M Elliott
- Centre for Heart Muscle Disease, Institute of Cardiological Sciences, University College London and St Bartholomew's Hospital, London, United Kingdom
| | - Carolyn Y Ho
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
78
|
Papadopoulou E, Bouzarelou D, Tsaousis G, Papathanasiou A, Vogiatzi G, Vlachopoulos C, Miliou A, Papachristou P, Prappa E, Servos G, Ritsatos K, Seretis A, Frogoudaki A, Nasioulas G. Application of next generation sequencing in cardiology: current and future precision medicine implications. Front Cardiovasc Med 2023; 10:1202381. [PMID: 37424920 PMCID: PMC10327645 DOI: 10.3389/fcvm.2023.1202381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Inherited cardiovascular diseases are highly heterogeneous conditions with multiple genetic loci involved. The application of advanced molecular tools, such as Next Generation Sequencing, has facilitated the genetic analysis of these disorders. Accurate analysis and variant identification are required to maximize the quality of the sequencing data. Therefore, the application of NGS for clinical purposes should be limited to laboratories with a high level of technological expertise and resources. In addition, appropriate gene selection and variant interpretation can result in the highest possible diagnostic yield. Implementation of genetics in cardiology is imperative for the accurate diagnosis, prognosis and management of several inherited disorders and could eventually lead to the realization of precision medicine in this field. However, genetic testing should also be accompanied by an appropriate genetic counseling procedure that clarifies the significance of the genetic analysis results for the proband and his family. In this regard, a multidisciplinary collaboration among physicians, geneticists, and bioinformaticians is imperative. In the present review, we address the current state of knowledge regarding genetic analysis strategies employed in the field of cardiogenetics. Variant interpretation and reporting guidelines are explored. Additionally, gene selection procedures are accessed, with a particular emphasis on information concerning gene-disease associations collected from international alliances such as the Gene Curation Coalition (GenCC). In this context, a novel approach to gene categorization is proposed. Moreover, a sub-analysis is conducted on the 1,502,769 variation records with submitted interpretations in the Clinical Variation (ClinVar) database, focusing on cardiology-related genes. Finally, the most recent information on genetic analysis's clinical utility is reviewed.
Collapse
Affiliation(s)
| | | | | | | | - Georgia Vogiatzi
- Third Department of Cardiology, Sotiria Hospital, Athens, Greece
| | - Charalambos Vlachopoulos
- Unit of Inherited Cardiac Conditions and Sports Cardiology, First Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Miliou
- Unit of Inherited Cardiac Conditions and Sports Cardiology, First Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Efstathia Prappa
- Second Department of Cardiology, Arrhythmia Unit, Evangelismos General Hospital of Athens, Athens, Greece
| | - Georgios Servos
- Pediatric Cardiology Unit, “P. & A. Kyriakou” Children’s Hospital, Athens, Greece
| | - Konstantinos Ritsatos
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, Athens, Greece
| | - Aristeidis Seretis
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, Athens, Greece
| | - Alexandra Frogoudaki
- Second Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
79
|
Pannone L, Bisignani A, Osei R, Gauthey A, Sorgente A, Vergara P, Monaco C, Della Rocca DG, Del Monte A, Strazdas A, Mojica J, Al Housari M, Miraglia V, Mouram S, Paparella G, Ramak R, Overeinder I, Bala G, Almorad A, Ströker E, Pappaert G, Sieira J, de Ravel T, La Meir M, Brugada P, Chierchia GB, Van Dooren S, de Asmundis C. Genetic testing in children with Brugada syndrome: results from a large prospective registry. Europace 2023; 25:euad079. [PMID: 37061847 PMCID: PMC10227762 DOI: 10.1093/europace/euad079] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/07/2023] [Indexed: 04/17/2023] Open
Abstract
AIMS A pathogenic/likely pathogenic (P/LP) variant in SCN5A is found in 20-25% of patients with Brugada syndrome (BrS). However, the diagnostic yield and prognosis of gene panel testing in paediatric BrS is unclear. The aim of this study is to define the diagnostic yield and outcomes of SCN5A gene testing with ACMG variant classification in paediatric BrS patients compared with adults. METHODS AND RESULTS All consecutive patients diagnosed with BrS, between 1992 and 2022, were prospectively enrolled in the UZ Brussel BrS registry. Inclusion criteria were: (i) BrS diagnosis; (ii) genetic analysis performed with a large gene panel; and (iii) classification of gene variants following ACMG guidelines. Paediatric patients were defined as ≤16 years of age. The primary endpoint was ventricular arrhythmias (VAs). A total of 500 BrS patients were included, with 63 paediatric patients and 437 adult patients. Among children with BrS, 29 patients (46%) had a P/LP variant (P+) in SCN5A and no variants were found in 34 (54%) patients (P-). After a mean follow-up of 125.9 months, 8 children (12.7%) experienced a VA, treated with implanted cardioverter defibrillator shock. At survival analysis, P- paediatric patients had higher VA-free survival during the follow-up, compared with P+ paediatric patients. P+ status was an independent predictor of VA. There was no difference in VA-free survival between paediatric and adult BrS patients for both P- and P+. CONCLUSION In a large BrS cohort, the diagnostic yield for P/LP variants in the paediatric population is 46%. P+ children with BrS have a worse arrhythmic prognosis.
Collapse
Affiliation(s)
- Luigi Pannone
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Antonio Bisignani
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Randy Osei
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Brussels, Belgium
| | - Anaïs Gauthey
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Antonio Sorgente
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Pasquale Vergara
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Cinzia Monaco
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Domenico Giovanni Della Rocca
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Alvise Del Monte
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Antanas Strazdas
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Joerelle Mojica
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Maysam Al Housari
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Vincenzo Miraglia
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Sahar Mouram
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Gaetano Paparella
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Robbert Ramak
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Ingrid Overeinder
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Gezim Bala
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Alexandre Almorad
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Erwin Ströker
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Gudrun Pappaert
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Juan Sieira
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Thomy de Ravel
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Brussels, Belgium
| | - Mark La Meir
- Cardiac Surgery Department, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, Brussels, Belgium
| | - Pedro Brugada
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Gian Battista Chierchia
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| | - Sonia Van Dooren
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Reproduction and Genetics, Brussels Interuniversity Genomics High Throughput Core (BRIGHTcore), Brussels, Belgium
| | - Carlo de Asmundis
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan, 101 1090 Brussels, Belgium
| |
Collapse
|
80
|
de la Guía-Galipienso F, Ugedo-Alzaga K, Grazioli G, Quesada-Ocete FJ, Feliu-Rey E, Perez MV, Quesada-Dorador A, Sanchis-Gomar F. Arrhythmogenic Cardiomyopathy and Athletes - A Dangerous Relationship. Curr Probl Cardiol 2023:101799. [PMID: 37172878 DOI: 10.1016/j.cpcardiol.2023.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a disease characterized by a progressive replacement of myocardium by fibro-adipose material, predisposing to ventricular arrhythmias (VA) and sudden cardiac death (SCD). Its prevalence is estimated at 1:2000 to 1:5000, with a higher incidence in males, and clinical onset is usually between the 2nd and 4th decade of life. The prevalence of ACM in SCD victims is relatively high, making it one of the most common etiologies in young patients with SCD, especially if they are athletes. Cardiac events occur more frequently in individuals with ACM who participate in competitive sports and/or high-intensity training. In effect, exercise activity can worsen RV function in cases of hereditary ACM. Estimating the incidence of SCD caused by ACM in athletes remains challenging, being reported frequency ranging from 3-20%. Here, we review the potential implications of exercising on the clinical course of the classical genetic form of ACM, as well as the diagnostic tools, risk stratification, and the different therapeutic tools available for managing ACM.
Collapse
Affiliation(s)
- Fernando de la Guía-Galipienso
- From the Glorieta Policlinic, Denia, Alicante, Spain; REMA-Sports Cardiology Clinic, Denia, Alicante, Spain; Cardiology Service, Hospital HCB Benidorm, Alicante, Spain; School of Medicine, Catholic University of Valencia San Vicente Mártir, Valencia, Spain.
| | | | | | - Francisco Javier Quesada-Ocete
- School of Medicine, Catholic University of Valencia San Vicente Mártir, Valencia, Spain; Arrhythmia Unit, Cardiology Service, General University Hospital Consortium of Valencia, Valencia, Spain
| | - Eloísa Feliu-Rey
- Magnetic Resonance Unit, Inscanner, General University Hospital of Alicante, Alicante, Spain
| | - Marco V Perez
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
| | - Aurelio Quesada-Dorador
- School of Medicine, Catholic University of Valencia San Vicente Mártir, Valencia, Spain; Arrhythmia Unit, Cardiology Service, General University Hospital Consortium of Valencia, Valencia, Spain
| | - Fabian Sanchis-Gomar
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA..
| |
Collapse
|
81
|
Rayani K, Davies B, Cheung M, Comber D, Roberts JD, Tadros R, Green MS, Healey JS, Simpson CS, Sanatani S, Steinberg C, MacIntyre C, Angaran P, Duff H, Hamilton R, Arbour L, Leather R, Seifer C, Fournier A, Atallah J, Kimber S, Makanjee B, Alqarawi W, Cadrin-Tourigny J, Joza J, Gardner M, Talajic M, Bagnall RD, Krahn AD, Laksman ZWM. Identification and in-silico characterization of splice-site variants from a large cardiogenetic national registry. Eur J Hum Genet 2023; 31:512-520. [PMID: 36138163 PMCID: PMC10172209 DOI: 10.1038/s41431-022-01193-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/23/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Splice-site variants in cardiac genes may predispose carriers to potentially lethal arrhythmias. To investigate, we screened 1315 probands and first-degree relatives enrolled in the Canadian Hearts in Rhythm Organization (HiRO) registry. 10% (134/1315) of patients in the HiRO registry carry variants within 10 base-pairs of the intron-exon boundary with 78% (104/134) otherwise genotype negative. These 134 probands were carriers of 57 unique variants. For each variant, American College of Medical Genetics and Genomics (ACMG) classification was revisited based on consensus between nine in silico tools. Due in part to the in silico algorithms, seven variants were reclassified from the original report, with the majority (6/7) downgraded. Our analyses predicted 53% (30/57) of variants to be likely/pathogenic. For the 57 variants, an average of 9 tools were able to score variants within splice sites, while 6.5 tools responded for variants outside these sites. With likely/pathogenic classification considered a positive outcome, the ACMG classification was used to calculate sensitivity/specificity of each tool. Among these, Combined Annotation Dependent Depletion (CADD) had good sensitivity (93%) and the highest response rate (131/134, 98%), dbscSNV was also sensitive (97%), and SpliceAI was the most specific (64%) tool. Splice variants remain an important consideration in gene elusive inherited arrhythmia syndromes. Screening for intronic variants, even when restricted to the ±10 positions as performed here may improve genetic testing yield. We compare 9 freely available in silico tools and provide recommendations regarding their predictive capabilities. Moreover, we highlight several novel cardiomyopathy-associated variants which merit further study.
Collapse
Affiliation(s)
- Kaveh Rayani
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brianna Davies
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Cheung
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Drake Comber
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada
| | - Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
| | - Martin S Green
- Heart Institute, University of Ottawa, Ottawa, ON, Canada
| | | | | | | | - Christian Steinberg
- Institut Universitaire de Cardiologie et Pneumologie de Quebec, Laval University, Quebec City, QC, Canada
| | - Ciorsti MacIntyre
- Division of Cardiology, QEII Health Sciences Center, Halifax, NS, Canada
| | - Paul Angaran
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Henry Duff
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Robert Hamilton
- Division of Cardiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
| | - Laura Arbour
- Division of Medical Genetics, Island Health, Victoria, BC, Canada
| | | | - Colette Seifer
- Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Anne Fournier
- Division of Pediatric Cardiology, CHU Sainte-Justine, Universite de Montreal, Montreal, QC, Canada
| | - Joseph Atallah
- Division of Pediatric Cardiology, University of Alberta Stollery Children's Hospital, Edmonton, AB, Canada
| | - Shane Kimber
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Bhavanesh Makanjee
- Heart Health Institute, Scarborough Health Network, Scarborough, ON, Canada
| | - Wael Alqarawi
- Heart Institute, University of Ottawa, Ottawa, ON, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Center, Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
| | - Jacqueline Joza
- Division of Cardiology, McGill University Health Centre, Montreal, QC, Canada
| | - Martin Gardner
- Division of Cardiology, QEII Health Sciences Center, Halifax, NS, Canada
| | - Mario Talajic
- Cardiovascular Genetics Center, Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
| | - Richard D Bagnall
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Andrew D Krahn
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Zachary W M Laksman
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
82
|
Arrigo AB, Zhu W, Williams KA, Guzman-Moreno C, Lo C, Lin JHI. Contribution of LRP1 in Human Congenital Heart Disease Correlates with Its Roles in the Outflow Tract and Atrioventricular Cushion Development. Genes (Basel) 2023; 14:genes14040947. [PMID: 37107705 PMCID: PMC10137934 DOI: 10.3390/genes14040947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Due to the prevalence of congenital heart disease in the human population, determining the role of variants in congenital heart disease (CHD) can give a better understanding of the cause of the disorder. A homozygous missense mutation in the LDL receptor-related protein 1 (Lrp1) in mice was shown to cause congenital heart defects, including atrioventricular septal defect (AVSD) and double outlet right ventricle (DORV). Integrative analysis of publicly available single-cell RNA sequencing (scRNA-seq) datasets and spatial transcriptomics of human and mouse hearts indicated that LRP1 is predominantly expressed in mesenchymal cells and mainly located in the developing outflow tract and atrioventricular cushion. Gene burden analysis of 1922 CHD individuals versus 2602 controls with whole-exome sequencing showed a significant excess of rare damaging LRP1 mutations in CHD (odds ratio (OR) = 2.22, p = 1.92 × 10-4), especially in conotruncal defect with OR of 2.37 (p = 1.77 × 10-3) and atrioventricular septal defect with OR of 3.14 (p = 0.0194). Interestingly, there is a significant relationship between those variants that have an allele frequency below 0.01% and atrioventricular septal defect, which is the phenotype observed previously in a homozygous N-ethyl-N-nitrosourea (ENU)-induced Lrp1 mutant mouse line.
Collapse
Affiliation(s)
- Angelo B Arrigo
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Wenjuan Zhu
- Centre for Cardiovascular Genomics and Medicine, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Kylia A Williams
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Carla Guzman-Moreno
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Cecilia Lo
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Jiuann-Huey I Lin
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| |
Collapse
|
83
|
Nelson S, Beck-Previs S, Sadayappan S, Tong C, Warshaw DM. Myosin-binding protein C stabilizes, but is not the sole determinant of SRX myosin in cardiac muscle. J Gen Physiol 2023; 155:e202213276. [PMID: 36688870 PMCID: PMC9884578 DOI: 10.1085/jgp.202213276] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/08/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
The myosin super-relaxed (SRX) state is central to striated muscle metabolic and functional regulation. In skeletal muscle, SRX myosin are predominantly colocalized with myosin-binding protein C (MyBP-C) in the sarcomere C-zone. To define how cardiac MyBP-C (cMyBP-C) and its specific domains contribute to stabilizing the SRX state in cardiac muscle, we took advantage of transgenic cMyBP-C null mice and those expressing cMyBP-C with a 271-residue N-terminal truncation. Utilizing super-resolution microscopy, we determined the lifetime and subsarcomeric location of individual fluorescent-ATP turnover events within isolated cardiac myofibrils. The proportion of SRX myosin demonstrated a gradient along the half-thick filament, highest in the P- and C-zones (72 ± 9% and 71 ± 6%, respectively) and lower in the D-zone (45 ± 10%), which lies farther from the sarcomere center and lacks cMyBP-C, suggesting a possible role for cMyBP-C in stabilizing the SRX. However, myofibrils from cMyBP-C null mice demonstrated an ∼40% SRX reduction, not only within the now cMyBP-C-free C-zone (49 ± 9% SRX), but also within the D-zone (22 ± 5% SRX). These data suggest that the influence of cMyBP-C on the SRX state is not limited to the C-zone but extends along the thick filament. Interestingly, myofibrils with N-terminal truncated cMyBP-C had an SRX content and spatial gradient similar to the cMyBP-C null, indicating that the N terminus of cMyBP-C is necessary for cMyBP-C's role in enhancing the SRX gradient along the entire thick filament. Given that SRX myosin exist as a gradient along the thick filament that is highest in the C-zone, even in the absence of cMyBP-C or its N-terminus, an inherent bias must exist in the structure of the thick filament to stabilize the SRX state.
Collapse
Affiliation(s)
- Shane Nelson
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Samantha Beck-Previs
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carl Tong
- Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
84
|
Sebastian SA, Panthangi V, Singh K, Rayaroth S, Gupta A, Shantharam D, Rasool BQ, Padda I, Co EL, Johal G. Hypertrophic Cardiomyopathy: Current Treatment and Future Options. Curr Probl Cardiol 2023; 48:101552. [PMID: 36529236 DOI: 10.1016/j.cpcardiol.2022.101552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a disease involving the cardiac sarcomere. It is associated with various disease-causing gene mutations and phenotypic expressions, managed with different therapies with variable prognoses. The heterogeneity of the disease is evident in the fact that it burdens patients of all ages. HCM is the most prevalent cause of sudden death in athletes. However, several technological advancements and therapeutic options have reduced mortality in patients with HCM to 0.5% per year. In addition, rapid advances in our knowledge of the molecular defects accountable for HCM have strengthened our awareness of the disorder and recommended new approaches to the assessment of prognosis. Despite all these evolutions, a small subgroup of patients with HCM will experience sudden cardiac death, and risk stratification remains a critical challenge. This review provides a practical guide to the updated recommendations for patients with HCM, including clinical updates for diagnosis, family screening, clinical imaging, risk stratification, and management.
Collapse
Affiliation(s)
| | | | - Karanbir Singh
- Department of Internal Medicine, Government Medical College, Amritsar, Punjab, India
| | - Swetha Rayaroth
- Department of Internal Medicine, JSS Medical College, Mysuru, Karnataka, India
| | - Aditi Gupta
- Department of Internal Medicine, Jawaharlal Nehru Medical College, Belgaum, Karnataka, India
| | - Darshan Shantharam
- Department of Internal Medicine, Yenepoya Medical college, Mangalore, India
| | | | - Inderbir Padda
- Department of Internal Medicine, Richmond University Medical Center, Staten Island, New York
| | - Edzel Lorraine Co
- Department of Internal Medicine, University of Santo Tomas, Manila, Philippines
| | - Gurpreet Johal
- Department of Cardiology, Valley Medical Center, University of Washington, Seattle, Washington
| |
Collapse
|
85
|
Girolami F, Gozzini A, Pálinkás ED, Ballerini A, Tomberli A, Baldini K, Marchi A, Zampieri M, Passantino S, Porcedda G, Calabri GB, Bennati E, Spaziani G, Crotti L, Cecchi F, Favilli S, Olivotto I. Genetic Testing and Counselling in Hypertrophic Cardiomyopathy: Frequently Asked Questions. J Clin Med 2023; 12:jcm12072489. [PMID: 37048573 PMCID: PMC10095452 DOI: 10.3390/jcm12072489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/13/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Genetic counselling and genetic testing in hypertrophic cardiomyopathy (HCM) represent an integral part of the diagnostic algorithm to confirm the diagnosis, distinguish it from phenocopies, and suggest tailored therapeutic intervention strategies. Additionally, they enable cascade genetic testing in the family. With the implementation of Next Generation Sequencing technologies (NGS), the interpretation of genetic data has become more complex. In this regard, cardiologists play a central role, aiding geneticists to correctly evaluate the pathogenicity of the identified genetic alterations. In the ideal setting, geneticists and cardiologists must work side by side to diagnose HCM as well as convey the correct information to patients in response to their many questions and concerns. After a brief overview of the role of genetics in the diagnosis of HCM, we present and discuss the frequently asked questions by HCM patients throughout our 20-year genetic counselling experience. Appropriate communication between the team and the families is key to the goal of delivering the full potential of genetic testing to our patients.
Collapse
Affiliation(s)
- Francesca Girolami
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
- Correspondence:
| | - Alessia Gozzini
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Eszter Dalma Pálinkás
- Doctoral School of Clinical Medicine, University of Szeged, 6720 Szeged, Hungary
- Cardiomyopathy Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Adelaide Ballerini
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Alessia Tomberli
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Katia Baldini
- Cardiomyopathy Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Alberto Marchi
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Mattia Zampieri
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Silvia Passantino
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Giulio Porcedda
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | | | - Elena Bennati
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Gaia Spaziani
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Lia Crotti
- Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, 20100 Milan, Italy
- Department of Medicine and Surgery, University Milano Bicocca, 20126 Milan, Italy
| | - Franco Cecchi
- Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, 20100 Milan, Italy
| | - Silvia Favilli
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
| | - Iacopo Olivotto
- Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
| |
Collapse
|
86
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
87
|
Halder SS, Rynkiewicz MJ, Creso JG, Sewanan LR, Howland L, Moore JR, Lehman W, Campbell SG. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TPM1 variant S215L. PNAS NEXUS 2023; 2:pgad011. [PMID: 36896133 PMCID: PMC9991458 DOI: 10.1093/pnasnexus/pgad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/12/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disorder often caused by mutations to sarcomeric genes. Many different HCM-associated TPM1 mutations have been identified but they vary in their degrees of severity, prevalence, and rate of disease progression. The pathogenicity of many TPM1 variants detected in the clinical population remains unknown. Our objective was to employ a computational modeling pipeline to assess pathogenicity of one such variant of unknown significance, TPM1 S215L, and validate predictions using experimental methods. Molecular dynamic simulations of tropomyosin on actin suggest that the S215L significantly destabilizes the blocked regulatory state while increasing flexibility of the tropomyosin chain. These changes were quantitatively represented in a Markov model of thin-filament activation to infer the impacts of S215L on myofilament function. Simulations of in vitro motility and isometric twitch force predicted that the mutation would increase Ca2+ sensitivity and twitch force while slowing twitch relaxation. In vitro motility experiments with thin filaments containing TPM1 S215L revealed higher Ca2+ sensitivity compared with wild type. Three-dimensional genetically engineered heart tissues expressing TPM1 S215L exhibited hypercontractility, upregulation of hypertrophic gene markers, and diastolic dysfunction. These data form a mechanistic description of TPM1 S215L pathogenicity that starts with disruption of the mechanical and regulatory properties of tropomyosin, leading thereafter to hypercontractility and finally induction of a hypertrophic phenotype. These simulations and experiments support the classification of S215L as a pathogenic mutation and support the hypothesis that an inability to adequately inhibit actomyosin interactions is the mechanism whereby thin-filament mutations cause HCM.
Collapse
Affiliation(s)
- Saiti S Halder
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | | | - Jenette G Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | - Lorenzo R Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Department of Internal Medicine, Columbia University, New York, NY 10032
| | - Lindsey Howland
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - William Lehman
- Department of Physiology/Biophysics, Boston University, Boston, MA 02215
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| |
Collapse
|
88
|
Tadros HJ, Choudhry S, Kearney DL, Hope K, Yesso A, Miyake CY, Price J, Spinner J, Tunuguntla H, Puri K, Dreyer W, Denfield SW. Arrhythmogenic cardiomyopathy is under-recognized in end-stage pediatric heart failure: A 36-year single-center experience. Pediatr Transplant 2023; 27:e14442. [PMID: 36451335 DOI: 10.1111/petr.14442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/27/2022] [Accepted: 11/09/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND Although ventricular failure is a late finding in adults with AC, we hypothesize that this is a presenting symptom in pediatric heart failure patients who undergo HT and that their ventricular arrhythmia burden could differentiate AC from other cardiomyopathies. METHODS We performed a single-center retrospective cohort study reviewing 457 consecutive pediatric (≤18 years) HT recipients at our institution. Explanted hearts were examined to establish the primary diagnosis, based on pathologic findings. Demographic and clinical variables were compared between AC versus non-HCM cardiomyopathy cases. RESULTS Forty-five percent (n = 205/457) had non-HCM cardiomyopathies as the underlying primary diagnosis. Ten cases (10/205 = 4.9%) were diagnosed with AC. All 10 had biventricular disease. In 8/10 patients (80%), AC diagnosis was unrecognized pre-HT. Compared with non-AC cardiomyopathies, the AC group was older at diagnosis (9.3 years vs. 4.3 years, p = .012) and transplant (11.1 years vs. 6.5 years, p = .010), had more ventricular arrhythmias (80.0% vs 32.8%, p = .003), and required more anti-arrhythmic use (80.0% vs 32.3%, p = .001). Genetic testing yielded causative pathogenic variants in all tested individuals (n = 5/5, 100%). CONCLUSION AC is often an unrecognized cardiomyopathy pretransplant in children who undergo HT. Pediatric non-HCM phenotypes with heart failure who have a significant ventricular arrhythmia burden should be investigated for AC.
Collapse
Affiliation(s)
- Hanna J Tadros
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Swati Choudhry
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Debra L Kearney
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Kyle Hope
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Abigail Yesso
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Christina Y Miyake
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Jack Price
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph Spinner
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Hari Tunuguntla
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Kriti Puri
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA.,Department of Pediatrics, Division of Pediatric Critical Care Medicine, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - William Dreyer
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Susan W Denfield
- Department of Pediatrics, Lille Frank Abercrombie Division of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
89
|
Ma W, You S, Regnier M, McCammon JA. Integrating comparative modeling and accelerated simulations reveals conformational and energetic basis of actomyosin force generation. Proc Natl Acad Sci U S A 2023; 120:e2215836120. [PMID: 36802417 PMCID: PMC9992861 DOI: 10.1073/pnas.2215836120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/15/2023] [Indexed: 02/23/2023] Open
Abstract
Muscle contraction is performed by arrays of contractile proteins in the sarcomere. Serious heart diseases, such as cardiomyopathy, can often be results of mutations in myosin and actin. Direct characterization of how small changes in the myosin-actin complex impact its force production remains challenging. Molecular dynamics (MD) simulations, although capable of studying protein structure-function relationships, are limited owing to the slow timescale of the myosin cycle as well as a lack of various intermediate structures for the actomyosin complex. Here, employing comparative modeling and enhanced sampling MD simulations, we show how the human cardiac myosin generates force during the mechanochemical cycle. Initial conformational ensembles for different myosin-actin states are learned from multiple structural templates with Rosetta. This enables us to efficiently sample the energy landscape of the system using Gaussian accelerated MD. Key myosin loop residues, whose substitutions are related to cardiomyopathy, are identified to form stable or metastable interactions with the actin surface. We find that the actin-binding cleft closure is allosterically coupled to the myosin motor core transitions and ATP-hydrolysis product release from the active site. Furthermore, a gate between switch I and switch II is suggested to control phosphate release at the prepowerstroke state. Our approach demonstrates the ability to link sequence and structural information to motor functions.
Collapse
Affiliation(s)
- Wen Ma
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
| | - Shengjun You
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA98109
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
- Department of Pharmacology, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
90
|
Zhang Y, Liu MH, Zhang M, Wu GX, Liu J, Wang JZ, Sun XL, Jiang W, Wang D, Kang LM, Wu XY, Zou YB, Song L. Different clinical characteristics and outcomes of hypertrophic cardiomyopathy with and without hypertension: seeking the truth. J Geriatr Cardiol 2023; 20:109-120. [PMID: 36910243 PMCID: PMC9992951 DOI: 10.26599/1671-5411.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023] Open
Abstract
OBJECTIVE To determine the different clinical characteristics and outcomes of hypertrophic cardiomyopathy (HCM) patients with and without hypertension (HT). METHODS A total of 696 HCM patients were included in this study and all HCM diagnoses were confirmed by the genetic test. Patients were analyzed separately in the septal reduction therapy (SRT) cohort and the non-SRT cohort. The primary endpoint was cardiovascular death and the secondary endpoint was all-cause death. Outcome analyses were conducted to evaluate the associations between HT and outcomes in HCM. Medications before enrollment and at discharge were collected in the post-hoc analyses. RESULTS HCM patients without HT were younger, had a lower body mass index, were more likely to have a family history of HCM, and had a smaller left ventricular (LV) end-diastolic diameter than those with HT in both cohorts. A thicker LV wall, a higher level of N-terminal pro-B-type natriuretic peptide, and a higher extent of LV late gadolinium enhancement were additionally observed in patients without HT in the non-SRT cohort. The presence of HT did not alter the distribution pattern of late gadolinium enhancement, as well as the constituent ratio of eight disease-causing sarcomeric gene variants in both cohorts. Outcome analyses showed that in the non-SRT cohort, patients without HT had higher risks of cardiovascular death (HR = 2.537, P = 0.032) and all-cause death (HR = 3.309, P = 0.032). While such prognostic divergence was not observed in the SRT cohort. Further post-hoc analyses in the non-SRT cohort found that patients without HT received fewer non-dihydropyridine calcium channel blockers and angiotensin-converting enzyme inhibitors/angiotensin receptor blockers before enrollment and at discharge. CONCLUSIONS HCM patients without HT had worse clinical conditions and higher mortality than patients with HT overall, which may result from active medical therapy in HT patients. Active SRT may have a substantial de-risking effect on patients meeting the indications.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Hao Liu
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mo Zhang
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gui-Xin Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji-Zheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Lu Sun
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Jiang
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Wang
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lian-Ming Kang
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue-Yi Wu
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Bao Zou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
91
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 1550] [Impact Index Per Article: 1550.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
92
|
Paul C, Peters S, Perrin M, Fatkin D, Amerena J. Non-ischaemic dilated cardiomyopathy: recognising the genetic links. Intern Med J 2023; 53:178-185. [PMID: 36043846 DOI: 10.1111/imj.15921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/25/2022] [Indexed: 11/27/2022]
Abstract
The landscape of genetically related cardiac disease continues to evolve. Heritable genetic variants can be a primary cause of familial or sporadic dilated cardiomyopathy (DCM). There is also increasing recognition that genetic variation is an important determinant of susceptibility to acquired causes of DCM. Genetic forms of DCM can show a wide variety of phenotypic manifestations. Identifying patients who are most likely to benefit from genetic testing is paramount. The objective of this review is to highlight the importance of recognising genetic DCM, key genotype-phenotype correlations and the value of genetic testing in clinical management for both the individual and their family. This is likely to become more relevant as management strategies continue to be refined with genotype-specific recommendations and disease-modifying therapies.
Collapse
Affiliation(s)
- Caitlin Paul
- Department of Cardiology, University Hospital Geelong, Geelong, Victoria, Australia.,Department of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Stacey Peters
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Mark Perrin
- Department of Cardiology, University Hospital Geelong, Geelong, Victoria, Australia.,Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia.,School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, New South Wales, Australia.,Cardiology Department, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - John Amerena
- Department of Cardiology, University Hospital Geelong, Geelong, Victoria, Australia.,Department of Medicine, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
93
|
Reichart D, Newby GA, Wakimoto H, Lun M, Gorham JM, Curran JJ, Raguram A, DeLaughter DM, Conner DA, Marsiglia JDC, Kohli S, Chmatal L, Page DC, Zabaleta N, Vandenberghe L, Liu DR, Seidman JG, Seidman C. Efficient in vivo genome editing prevents hypertrophic cardiomyopathy in mice. Nat Med 2023; 29:412-421. [PMID: 36797483 PMCID: PMC9941048 DOI: 10.1038/s41591-022-02190-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/16/2022] [Indexed: 02/18/2023]
Abstract
Dominant missense pathogenic variants in cardiac myosin heavy chain cause hypertrophic cardiomyopathy (HCM), a currently incurable disorder that increases risk for stroke, heart failure and sudden cardiac death. In this study, we assessed two different genetic therapies-an adenine base editor (ABE8e) and a potent Cas9 nuclease delivered by AAV9-to prevent disease in mice carrying the heterozygous HCM pathogenic variant myosin R403Q. One dose of dual-AAV9 vectors, each carrying one half of RNA-guided ABE8e, corrected the pathogenic variant in ≥70% of ventricular cardiomyocytes and maintained durable, normal cardiac structure and function. An additional dose provided more editing in the atria but also increased bystander editing. AAV9 delivery of RNA-guided Cas9 nuclease effectively inactivated the pathogenic allele, albeit with dose-dependent toxicities, necessitating a narrow therapeutic window to maintain health. These preclinical studies demonstrate considerable potential for single-dose genetic therapies to correct or silence pathogenic variants and prevent the development of HCM.
Collapse
Affiliation(s)
- Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Mingyue Lun
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Justin J Curran
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Aditya Raguram
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Daniel M DeLaughter
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David A Conner
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Sajeev Kohli
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - David C Page
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Whitehead Institute, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nerea Zabaleta
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA
- Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Luk Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA
- Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - Christine Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
94
|
Kontorovich AR. Approaches to Genetic Screening in Cardiomyopathies: Practical Guidance for Clinicians. JACC. HEART FAILURE 2023; 11:133-142. [PMID: 36754525 DOI: 10.1016/j.jchf.2022.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 02/08/2023]
Abstract
Patients and families benefit when the genetic etiology of cardiomyopathy is elucidated through a multidisciplinary approach including genetic counseling and judicious use of genetic testing. The yield of genetic testing is optimized when performed on a proband with a clear phenotype, and interrogates genes that are validated in association with that specific form of cardiomyopathy. Variants of uncertain significance are frequently uncovered and should not be overinterpreted. Identifying an impactful genetic variant as the cause of a patient's cardiomyopathy can have important prognostic impact, and enable streamlined cascade testing to highlight at risk relatives. Certain genotypes are associated with unique potential cardiac and noncardiac risk factors and may dictate personalized approaches to treatment.
Collapse
Affiliation(s)
- Amy R Kontorovich
- Center for Inherited Cardiovascular Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
95
|
Vadrot N, Ader F, Moulin M, Merlant M, Chapon F, Gandjbakhch E, Labombarda F, Maragnes P, Réant P, Rooryck C, Probst V, Donal E, Richard P, Ferreiro A, Buendia B. Abnormal Cellular Phenotypes Induced by Three TMPO/LAP2 Variants Identified in Men with Cardiomyopathies. Cells 2023; 12:337. [PMID: 36672271 PMCID: PMC9857342 DOI: 10.3390/cells12020337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
A single missense variant of the TMPO/LAP2α gene, encoding LAP2 proteins, has been associated with cardiomyopathy in two brothers. To further evaluate its role in cardiac muscle, we included TMPO in our cardiomyopathy diagnostic gene panel. A screening of ~5000 patients revealed three novel rare TMPO heterozygous variants in six males diagnosed with hypertrophic or dilated cardiomypathy. We identified in different cellular models that (1) the frameshift variant LAP2α p.(Gly395Glufs*11) induced haploinsufficiency, impeding cell proliferation and/or producing a truncated protein mislocalized in the cytoplasm; (2) the C-ter missense variant LAP2α p.(Ala240Thr) led to a reduced proximity events between LAP2α and the nucleosome binding protein HMGN5; and (3) the LEM-domain missense variant p.(Leu124Phe) decreased both associations of LAP2α/β with the chromatin-associated protein BAF and inhibition of the E2F1 transcription factor activity which is known to be dependent on Rb, partner of LAP2α. Additionally, the LAP2α expression was lower in the left ventricles of male mice compared to females. In conclusion, our study reveals distinct altered properties of LAP2 induced by these TMPO/LAP2 variants, leading to altered cell proliferation, chromatin structure or gene expression-regulation pathways, and suggests a potential sex-dependent role of LAP2 in myocardial function and disease.
Collapse
Affiliation(s)
- Nathalie Vadrot
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Flavie Ader
- APHP—Sorbonne Université, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, HU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
- Faculté de Pharmacie Paris Descartes, Département 3, Université Paris Cité, F-75006 Paris, France
| | - Maryline Moulin
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Marie Merlant
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | | | - Estelle Gandjbakhch
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
- Département de cardiologie, APHP—Sorbonne Université, HU Pitié Salpêtrière- Charles Foix, F-75610 Paris, France
| | - Fabien Labombarda
- Service de Cardiologie, CHU de Caen, Université de Caen Normandie, F-14000 Caen, France
| | - Pascale Maragnes
- Cardiologie pédiatrique, Service de pédiatrie, CHU de Caen, F-14000 Caen, France
| | - Patricia Réant
- Service de Cardiologie, Hôpital Haut Lévêque, CHU de Bordeaux, INSERM 1045, Université de Bordeaux, F-33000 Bordeaux, France
| | - Caroline Rooryck
- Service de Génétique Médicale, CHU Bordeaux, F-33000 Bordeaux, France
| | - Vincent Probst
- Centre de référence des maladies rythmiques cardiaques, CHU de Nantes, F-44000 Nantes, France
| | - Erwan Donal
- Centre Cardio-Pneumologique, CHU de Rennes Hôpital de Pontchaillou, F-35000 Rennes, France
| | - Pascale Richard
- APHP—Sorbonne Université, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, HU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
- APHP, Centre de référence des Maladies Neuromusculaires, Institut de Myologie, Neuromyology Department, CHU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
| | - Brigitte Buendia
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| |
Collapse
|
96
|
Marston S, Pinto JR. Suppression of lusitropy as a disease mechanism in cardiomyopathies. Front Cardiovasc Med 2023; 9:1080965. [PMID: 36698941 PMCID: PMC9870330 DOI: 10.3389/fcvm.2022.1080965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
In cardiac muscle the action of adrenaline on β1 receptors of heart muscle cells is essential to adjust cardiac output to the body's needs. Adrenergic activation leads to enhanced contractility (inotropy), faster heart rate (chronotropy) and faster relaxation (lusitropy), mainly through activation of protein kinase A (PKA). Efficient enhancement of heart output under stress requires all of these responses to work together. Lusitropy is essential for shortening the heartbeat when heart rate increases. It therefore follows that, if the lusitropic response is not present, heart function under stress will be compromised. Current literature suggests that lusitropy is primarily achieved due to PKA phosphorylation of troponin I (TnI) and phospholamban (PLB). It has been well documented that PKA-induced phosphorylation of TnI releases Ca2+ from troponin C faster and increases the rate of cardiac muscle relaxation, while phosphorylation of PLB increases SERCA activity, speeding up Ca2+ removal from the cytoplasm. In this review we consider the current scientific evidences for the connection between suppression of lusitropy and cardiac dysfunction in the context of mutations in phospholamban and thin filament proteins that are associated with cardiomyopathies. We will discuss what advances have been made into understanding the physiological mechanism of lusitropy due to TnI and PLB phosphorylation and its suppression by mutations and we will evaluate the evidence whether lack of lusitropy is sufficient to cause cardiomyopathy, and under what circumstances, and consider the range of pathologies associated with loss of lusitropy. Finally, we will discuss whether suppressed lusitropy due to mutations in thin filament proteins can be therapeutically restored.
Collapse
Affiliation(s)
- Steven Marston
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
97
|
Siguero-Álvarez M, Salguero-Jiménez A, Grego-Bessa J, de la Barrera J, MacGrogan D, Prados B, Sánchez-Sáez F, Piñeiro-Sabarís R, Felipe-Medina N, Torroja C, Gómez MJ, Sabater-Molina M, Escribá R, Richaud-Patin I, Iglesias-García O, Sbroggio M, Callejas S, O'Regan DP, McGurk KA, Dopazo A, Giovinazzo G, Ibañez B, Monserrat L, Pérez-Pomares JM, Sánchez-Cabo F, Pendas AM, Raya A, Gimeno-Blanes JR, de la Pompa JL. A Human Hereditary Cardiomyopathy Shares a Genetic Substrate With Bicuspid Aortic Valve. Circulation 2023; 147:47-65. [PMID: 36325906 DOI: 10.1161/circulationaha.121.058767] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.
Collapse
Affiliation(s)
- Marcos Siguero-Álvarez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Center for Chromosome Stability and Institut for Cellulær og Molekylær Medicin, University of Copenhagen, Denmark (M.S.)
| | - Alejandro Salguero-Jiménez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Joaquim Grego-Bessa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Jorge de la Barrera
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Belén Prados
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Fernando Sánchez-Sáez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Natalia Felipe-Medina
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Carlos Torroja
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Manuel José Gómez
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - María Sabater-Molina
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Rubén Escribá
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Ivonne Richaud-Patin
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
- Regenerative Medicine Program, Cima Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain (O.I.-G.)
| | - Mauro Sbroggio
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Sergio Callejas
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Declan P O'Regan
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
| | - Kathryn A McGurk
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
- National Heart and Lung Institute (K.A.M.), Imperial College London, United Kingdom
| | - Ana Dopazo
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Borja Ibañez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Translational Laboratory (B.I.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Cardiology Department, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Hospital, Madrid, Spain (B.I.)
| | - Lorenzo Monserrat
- Instituto de Investigación Biomédica de A Coruña and Departamento Científico, Health in Code S.L., A Coruña, Spain (L.M.)
| | - José María Pérez-Pomares
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Department of Animal Biology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga and Centro Andaluz de Nanomedicina y Biotecnología, Universidad de Málaga, Spain (J.M.P.-P.)
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Alberto M Pendas
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Juan R Gimeno-Blanes
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| |
Collapse
|
98
|
Bhat V, Adzhubei IA, Fife JD, Lebo M, Cassa CA. Informing variant assessment using structured evidence from prior classifications (PS1, PM5, and PVS1 sequence variant interpretation criteria). Genet Med 2023; 25:16-26. [PMID: 36305854 DOI: 10.1016/j.gim.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE This study aimed to explore whether evidence of pathogenicity from prior variant classifications in ClinVar could be used to inform variant interpretation using the American College of Medical Genetics and Genomics/Association for Molecular Pathology clinical guidelines. METHODS We identified distinct single-nucleotide variants (SNVs) that are either similar in location or in functional consequence to pathogenic variants in ClinVar and analyzed evidence in support of pathogenicity using 3 interpretation criteria. RESULTS Thousands of variants, including many in clinically actionable disease genes (American College of Medical Genetics and Genomics secondary findings v3.0), have evidence of pathogenicity from existing variant classifications, accounting for 2.5% of nonsynonymous SNVs within ClinVar. Notably, there are many variants with uncertain or conflicting classifications that cause the same amino acid substitution as other pathogenic variants (PS1, N = 323), variants that are predicted to cause different amino acid substitutions in the same codon as pathogenic variants (PM5, N = 7692), and loss-of-function variants that are present in genes in which many loss-of-function variants are classified as pathogenic (PVS1, N = 3635). Most of these variants have similar computational predictions of pathogenicity and splicing effect as their associated pathogenic variants. CONCLUSION Broadly, for >1.4 million SNVs exome wide, information from previously classified variants could be used to provide evidence of pathogenicity. We have developed a pipeline to identify variants meeting these criteria that may inform interpretation efforts.
Collapse
Affiliation(s)
- Vineel Bhat
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ivan A Adzhubei
- Department of Biomedical Informatics, Blavatnik Institute, Harvard Medical School, Boston, MA
| | - James D Fife
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Matthew Lebo
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Boston, MA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Christopher A Cassa
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
99
|
Lin L, Liu S, Chen Z, Xia Y, Xie J, Fu M, Lu D, Wu Y, Shen H, Yang P, Qian J. Anatomically resolved transcriptome and proteome landscapes reveal disease‐relevant molecular signatures and systematic changes in heart function of end‐stage dilated cardiomyopathy. VIEW 2022. [DOI: 10.1002/viw.20220040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Ling Lin
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Shanshan Liu
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Zhangwei Chen
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Yan Xia
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Juanjuan Xie
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Mingqiang Fu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Danbo Lu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Yuan Wu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Huali Shen
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
- Department of chemistry Fudan University Shanghai China
| | - Juying Qian
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| |
Collapse
|
100
|
Titin-truncating variants in hiPSC cardiomyocytes induce pathogenic proteinopathy and sarcomere defects with preserved core contractile machinery. Stem Cell Reports 2022; 18:220-236. [PMID: 36525964 PMCID: PMC9860080 DOI: 10.1016/j.stemcr.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
Titin-truncating variants (TTNtv) are the single largest genetic cause of dilated cardiomyopathy (DCM). In this study we modeled disease phenotypes of A-band TTNtv-induced DCM in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using genome editing and tissue engineering technologies. Transcriptomic, cellular, and micro-tissue studies revealed that A-band TTNtv hiPSC-CMs exhibit pathogenic proteinopathy, sarcomere defects, aberrant Na+ channel activities, and contractile dysfunction. These phenotypes establish a dual mechanism of poison peptide effect and haploinsufficiency that collectively contribute to DCM pathogenesis. However, TTNtv cellular defects did not interfere with the function of the core contractile machinery, the actin-myosin-troponin-Ca2+ complex, and preserved the therapeutic mechanism of sarcomere modulators. Treatment of TTNtv cardiac micro-tissues with investigational sarcomere modulators augmented contractility and resulted in sustained transcriptomic changes that promote reversal of DCM disease signatures. Together, our findings elucidate the underlying pathogenic mechanisms of A-band TTNtv-induced DCM and demonstrate the validity of sarcomere modulators as potential therapeutics.
Collapse
|