1
|
Imai Y, Kusano K, Aiba T, Ako J, Asano Y, Harada-Shiba M, Kataoka M, Kosho T, Kubo T, Matsumura T, Minamino T, Minatoya K, Morita H, Nishigaki M, Nomura S, Ogino H, Ohno S, Takamura M, Tanaka T, Tsujita K, Uchida T, Yamagishi H, Ebana Y, Fujita K, Ida K, Inoue S, Ito K, Kuramoto Y, Maeda J, Matsunaga K, Neki R, Sugiura K, Tada H, Tsuji A, Yamada T, Yamaguchi T, Yamamoto E, Kimura A, Kuwahara K, Maemura K, Minamino T, Morisaki H, Tokunaga K. JCS/JCC/JSPCCS 2024 Guideline on Genetic Testing and Counseling in Cardiovascular Disease. Circ J 2024; 88:2022-2099. [PMID: 39343605 DOI: 10.1253/circj.cj-23-0926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Affiliation(s)
- Yasushi Imai
- Division of Clinical Pharmacology and Division of Cardiovascular Medicine, Jichi Medical University
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | - Yoshihiro Asano
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center
| | | | - Masaharu Kataoka
- The Second Department of Internal Medicine, University of Occupational and Environmental Health
| | - Tomoki Kosho
- Department of Medical Genetics, Shinshu University School of Medicine
| | - Toru Kubo
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | - Takayoshi Matsumura
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Masakazu Nishigaki
- Department of Genetic Counseling, International University of Health and Welfare
| | - Seitaro Nomura
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo
| | | | - Seiko Ohno
- Medical Genome Center, National Cerebral and Cardiovascular Center
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Tetsuro Uchida
- Department of Surgery II (Division of Cardiovascular, Thoracic and Pediatric Surgery), Yamagata University Faculty of Medicine
| | | | - Yusuke Ebana
- Life Science and Bioethics Research Center, Tokyo Medical and Dental University Hospital
| | - Kanna Fujita
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Department of Computational Diagnostic Radiology and Preventive Medicine, Graduate School of Medicine, The University of Tokyo
| | - Kazufumi Ida
- Division of Counseling for Medical Genetics, National Cerebral and Cardiovascular Center
| | - Shunsuke Inoue
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Jun Maeda
- Department of Cardiology, Tokyo Metropolitan Children's Medical Center
| | - Keiji Matsunaga
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Reiko Neki
- Division of Counseling for Medical Genetics, Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center
| | - Kenta Sugiura
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | - Hayato Tada
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University
| | - Akihiro Tsuji
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | | | | | | | - Akinori Kimura
- Institutional Research Office, Tokyo Medical and Dental University
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University School of Medicine
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | | | - Katsushi Tokunaga
- Genome Medical Science Project, National Center for Global Health and Medicine
| |
Collapse
|
2
|
Busse L, Huth EA, Abraham MR, Abraham T, Padmanabhan A, Wojciak J, Wright G, Aatre R, Campagna R, Jackson E, Kreykes S, Lane K, Sawyer L, Stevens C, Thomas M, VanDyke R, Vedantham V, Higgs EJ. ALPK3 heterozygous truncating variants cause late-onset hypertrophic cardiomyopathy with frequent apical involvement and apical aneurysm. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.14.24317359. [PMID: 39606411 PMCID: PMC11601730 DOI: 10.1101/2024.11.14.24317359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetically heterogeneous disorder with several established genotype-phenotype relationships. While biallelic truncating variants in the ALPK3 gene cause severe congenital HCM, recent studies have associated heterozygous truncating variants (ALPK3tv) with milder adult-onset HCM. Here we describe a multicenter cohort of 21 individuals with heterozygous ALPK3tv from 10 institutions in the United States, highlighting distinctive clinical characteristics compared to a control group of 132 patients with HCM caused by deleterious variants in sarcomeric genes. As compared to other HCM genotypes, ALPK3tv patients present at an older age (mean 57.25 years) with significantly lower left ventricular wall thickness (14.09 vs 19.78 mm with echocardiogram and 16.13 vs 21.13 mm with cardiac MRI), a lower prevalence of obstructive HCM (15% of ALPK3tv vs 45% of controls), and a strikingly higher incidence of apical aneurysm (22.22% vs. 2.40% in the control group). These results suggest a milder degree of hypertrophy in heterozygous ALPK3-related HCM as compared to other Mendelian causes of HCM, although the increased occurrence of apical aneurysms could have implications for ventricular arrhythmia risk. Our study underscores the importance of recognizing heterozygous ALPK3tv as a cause of adult-onset HCM and provides a comprehensive characterization of its clinical phenotype.
Collapse
|
3
|
Farrant JP, Schmitt M, Reid AB, Garratt CJ, Newman WG, Malhotra A, Beynon R, Mahmod M, Raman B, Cooper RM, Dawson D, Green T, Prasad SK, Singh A, Dodd S, Watkins H, Neubauer S, Miller CA. Considerations for drug trials in hypertrophic cardiomyopathy. ESC Heart Fail 2024. [PMID: 39462184 DOI: 10.1002/ehf2.15138] [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: 09/12/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heterogeneous condition with potentially serious manifestations. Management has traditionally comprised therapies to palliate symptoms and implantable cardioverter-defibrillators to prevent sudden cardiac death. The need for disease-modifying therapies has been recognized for decades. More recently, an increasing number of novel and repurposed therapies hypothesized to target HCM disease pathways have been evaluated, culminating in the recent regulatory approval of mavacamten, a novel oral myosin inhibitor. HCM poses several unique challenges for clinical trials, which are important to recognize when designing trials and interpreting findings. This manuscript discusses the key considerations in the context of recent and ongoing randomized trials, including the roles of genotype, phenotype and symptom status in patient selection, the evidence base for clinical and mechanistic outcome measurements, trial duration and sample size.
Collapse
Affiliation(s)
- John P Farrant
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Matthias Schmitt
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Anna B Reid
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Clifford J Garratt
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - William G Newman
- Manchester Centre for Genomic Medicine Manchester University NHS Foundation Trust, Oxford Road, Manchester, M13 9WL, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Aneil Malhotra
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
- Institute of Sport, Manchester Metropolitan University, 99 Oxford Rd, Manchester, M1 7EL, UK
| | - Rhys Beynon
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, OX3 9DU, UK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, OX3 9DU, UK
| | - Robert M Cooper
- Liverpool Heart and Chest Hospital, Thomas Dr, Liverpool, L14 3PE, UK
- Liverpool John Moores University, 70 Mount Pleasant, Merseyside, L3 5UX, UK
| | - Dana Dawson
- School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
- Cardiology Department, Aberdeen Royal Infirmary, Aberdeen, AB25 2ZN, UK
| | - Thomas Green
- Cardiology Department, Northumbria Healthcare NHS Trust, Northumberland, UK
| | - Sanjay K Prasad
- Royal Brompton and Harefield NHS Foundation Trust, Sydney St, London, SW3 6NP, UK
- National Heart and Lung Institute, Imperial College London, London
| | - Anvesha Singh
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Susanna Dodd
- Department of Health Data Sciences, Institute of Population Health, Faculty of Health and Life Sciences, University of Liverpool, Block F, Waterhouse Boulevard, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, OX3 9DU, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, OX3 9DU, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
4
|
Verheyen N, Auer J, Bonaros N, Buchacher T, Dalos D, Grimm M, Mayr A, Rab A, Reinstadler S, Scherr D, Toth GG, Weber T, Zach DK, Zaruba MM, Zimpfer D, Rainer PP, Pölzl G. Austrian consensus statement on the diagnosis and management of hypertrophic cardiomyopathy. Wien Klin Wochenschr 2024; 136:571-597. [PMID: 39352517 PMCID: PMC11445290 DOI: 10.1007/s00508-024-02442-1] [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] [Accepted: 08/27/2024] [Indexed: 10/04/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease that is characterized by left ventricular hypertrophy unexplained by secondary causes. Based on international epidemiological data, around 20,000-40,000 patients are expected to be affected in Austria. Due to the wide variety of clinical and morphological manifestations the diagnosis can be difficult and the disease therefore often goes unrecognized. HCM is associated with a substantial reduction in quality of life and can lead to sudden cardiac death, especially in younger patients. Early and correct diagnosis, including genetic testing, is essential for comprehensive counselling of patients and their families and for effective treatment. The latter is especially true as an effective treatment of outflow tract obstruction has recently become available in the form of a first in class cardiac myosin ATPase inhibitor, as a noninvasive alternative to established septal reduction therapies. The aim of this Austrian consensus statement is to summarize the recommendations of international guidelines with respect to the genetic background, pathophysiology, diagnostics and management in the context of the Austrian healthcare system and resources, and to present them in easy to understand algorithms.
Collapse
Affiliation(s)
- Nicolas Verheyen
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Johannes Auer
- Department of Internal Medicine 1 with Cardiology and Intensive Care, St. Josef Hospital Braunau, Braunau, Austria
- Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Tamara Buchacher
- Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Daniel Dalos
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Grimm
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Agnes Mayr
- University Clinic of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Rab
- Department Internal Medicine I, Kardinal Schwarzenberg Klinikum, Schwarzach, Austria
| | - Sebastian Reinstadler
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Daniel Scherr
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Gabor G Toth
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Thomas Weber
- Department Innere Medizin II, Cardiology and Intensive Care Medicine, Klinikum Wels-Grieskirchen, Wels, Austria
| | - David K Zach
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Marc-Michael Zaruba
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Daniel Zimpfer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
- BioTech Med, Graz, Austria
- Department of Medicine, St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| | - Gerhard Pölzl
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
5
|
Mukhopadhyay S, Dixit P, Khanom N, Sanghera G, McGurk KA. The Genetic Factors Influencing Cardiomyopathies and Heart Failure across the Allele Frequency Spectrum. J Cardiovasc Transl Res 2024; 17:1119-1139. [PMID: 38771459 PMCID: PMC11519107 DOI: 10.1007/s12265-024-10520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
Heart failure (HF) remains a major cause of mortality and morbidity worldwide. Understanding the genetic basis of HF allows for the development of disease-modifying therapies, more appropriate risk stratification, and personalised management of patients. The advent of next-generation sequencing has enabled genome-wide association studies; moving beyond rare variants identified in a Mendelian fashion and detecting common DNA variants associated with disease. We summarise the latest GWAS and rare variant data on mixed and refined HF aetiologies, and cardiomyopathies. We describe the recent understanding of the functional impact of titin variants and highlight FHOD3 as a novel cardiomyopathy-associated gene. We describe future directions of research in this field and how genetic data can be leveraged to improve the care of patients with HF.
Collapse
Affiliation(s)
- Srinjay Mukhopadhyay
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
- School of Medicine, Cardiff University, Wales, UK
| | - Prithvi Dixit
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Najiyah Khanom
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Gianluca Sanghera
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK.
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK.
| |
Collapse
|
6
|
Pisklova M, Osmak G. Unveiling MiRNA-124 as a biomarker in hypertrophic cardiomyopathy: An innovative approach using machine learning and intelligent data analysis. Int J Cardiol 2024; 410:132220. [PMID: 38815672 DOI: 10.1016/j.ijcard.2024.132220] [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: 04/17/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a widespread hereditary cardiac pathology characterized by thickened heart walls and rearrangement of cardiomyocytes. Despite extensive research, the mechanisms underlying HCM development remain poorly understood, impeding the development of effective therapeutic and diagnostic strategies. Recent studies have suggested a polygenic nature of HCM development alongside monogenic forms. Transcriptomic profiling is a valuable tool for investigating such diseases. In this study, we propose a novel approach to study regulatory microRNAs (miRNAs) in the context of HCM, utilizing state-of-the-art data analysis tools. METHODS AND RESULTS Our method involves applying the Monte Carlo simulation and machine learning algorithm to transcriptomic data to generate high-capacity classifiers for HCM. From these classifiers, we extract key genes crucial for their performance, resulting in the identification of 16 key genes. Subsequently, we narrow down the pool of miRNAs by selecting those that may target the greatest number of key genes within the best models. We particularly focused on miR-124-3p, which we validated to have an association with HCM on an independent dataset. Subsequent investigation of its function revealed involvement of miR-124-3p in the RhoA signaling pathway. CONCLUSIONS In this study we propose a new approach to analyze transcriptomic data to search for microRNAs associated with a disease. Using this approach for transcriptomic profiling data of patients with HCM, we identified miR-124-3p as a potential regulator of the RhoA signaling pathway in the pathogenesis of HCM.
Collapse
Affiliation(s)
- Maria Pisklova
- E.I. Chazov National Medical Research Center for Cardiology, Academician Chazov st. 15a, 121552 Moscow, Russia; Pirogov Russian National Research Medical University, Ostrovitianov st. 1, 117997 Moscow, Russia
| | - German Osmak
- E.I. Chazov National Medical Research Center for Cardiology, Academician Chazov st. 15a, 121552 Moscow, Russia; Pirogov Russian National Research Medical University, Ostrovitianov st. 1, 117997 Moscow, Russia.
| |
Collapse
|
7
|
Wu X, Chen N, Hsu P, Sun J, Li W, Wang Q, Samira M, Wei Q, Yu J, Cao G, Yang H, Wang L, Wang J, Jin Y, Liu W, Wu J, He J, Lyu C, Zhang J. Pharmacokinetics and safety of mavacamten in healthy Chinese participants with different CYP2C19 phenotypes. Clin Transl Sci 2024; 17:e13877. [PMID: 39014868 PMCID: PMC11252221 DOI: 10.1111/cts.13877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Obstructive hypertrophic cardiomyopathy (oHCM) is a subtype of HCM characterized by left ventricular outflow tract obstruction resulting from cardiac muscle hypertrophy and anatomic alterations in the mitral valve and apparatus. Mavacamten, a cardiac myosin inhibitor metabolized primarily by CYP2C19 in the liver, is the first and only targeted medication approved for the treatment of symptomatic New York Heart Association (NYHA) class II-III oHCM. Previous pharmacokinetic (PK) results of mavacamten in healthy Caucasian, Japanese, and Asian participants demonstrated that mavacamten exposure was affected by CYP2C19 metabolism status. This open-label, parallel-group, phase I trial aimed to determine the PK and safety of mavacamten in healthy Chinese participants with different CYP2C19 genotypes. The primary outcome was to define the PK of mavacamten in healthy Chinese participants; the secondary outcome was to examine safety and tolerability. After a single oral dose of 15 or 25 mg mavacamten in fasted healthy adult Chinese individuals, Cmax was reached within a median Tmax of 0.6-1.5 h, indicating rapid absorption. Inter-individual variability was moderate, and individuals carrying non-functional CYP2C19 alleles (*2/*2, *3/*3, or *2/*3) exhibited longer half-life and increased total exposure. After stratification of CYP2C19 genotypes, total mavacamten exposures were similar among different ethnic groups when compared with prior PK studies. No significant adverse events were observed in this study. Single oral administration of mavacamten at 15 mg was well tolerated across all CYP2C19 genotypes, and 25 mg dose was well tolerated in healthy participants with CYP2C19 genotypes UM/RM/NM. The PK profile of mavacamten in the healthy Chinese population was consistent with that in other healthy populations.
Collapse
Affiliation(s)
- Xiaojie Wu
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan Hospital, Fudan UniversityShanghaiChina
- Research Ward of Huashan HospitalFudan UniversityShanghaiChina
| | - Nanye Chen
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Peiwen Hsu
- Shanghai LianBio Development Co., LtdShanghaiChina
| | - Jing Sun
- Bristol Myers SquibbShanghaiChina
| | - Wenting Li
- dMed Biopharmaceutical Co., LtdShanghaiChina
| | - Qi Wang
- Bristol Myers SquibbShanghaiChina
| | | | - Qiong Wei
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jicheng Yu
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan Hospital, Fudan UniversityShanghaiChina
- Research Ward of Huashan HospitalFudan UniversityShanghaiChina
| | - Guoying Cao
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan Hospital, Fudan UniversityShanghaiChina
- Research Ward of Huashan HospitalFudan UniversityShanghaiChina
| | - Haijing Yang
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Lili Wang
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jingjing Wang
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Yi Jin
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Wei Liu
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jufang Wu
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jinjie He
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Cheng Lyu
- Shanghai LianBio Development Co., LtdShanghaiChina
| | - Jing Zhang
- Phase 1 Clinical Research CenterHuashan Hospital, Fudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan Hospital, Fudan UniversityShanghaiChina
- Research Ward of Huashan HospitalFudan UniversityShanghaiChina
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
8
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Substrate mechanics unveil early structural and functional pathology in iPSC micro-tissue models of hypertrophic cardiomyopathy. iScience 2024; 27:109954. [PMID: 38827401 PMCID: PMC11141149 DOI: 10.1016/j.isci.2024.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024] Open
Abstract
Hypertension is a major cause of morbidity and mortality in patients with hypertrophic cardiomyopathy (HCM), suggesting a potential role for mechanics in HCM pathogenesis. Here, we developed an in vitro physiological model to investigate how mechanics acts together with HCM-linked myosin binding protein C (MYBPC3) mutations to trigger disease. Micro-heart muscles (μHM) were engineered from induced pluripotent stem cell (iPSC)-derived cardiomyocytes bearing MYBPC3+/- mutations and challenged to contract against substrates of different elasticity. μHMs that worked against substrates with stiffness at or exceeding the stiffness of healthy adult heart muscle exhibited several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in MYBPC3+/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca2+ intake through membrane-embedded channels underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease with iPSC technology.
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Ghiska Ramahdita
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Druv Bhagavan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan Silva
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
9
|
Hou C, Fei S, Jia F. Necroptosis and immune infiltration in hypertrophic cardiomyopathy: novel insights from bioinformatics analyses. Front Cardiovasc Med 2024; 11:1293786. [PMID: 38947229 PMCID: PMC11211569 DOI: 10.3389/fcvm.2024.1293786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Background Hypertrophic Cardiomyopathy (HCM), a widespread genetic heart disorder, is largely associated with sudden cardiac fatality. Necroptosis, an emerging type of programmed cell death, plays a fundamental role in several cardiovascular diseases. Aim This research utilized bioinformatics analysis to investigate necroptosis's implication in HCM. Methods The study retrieved RNA sequencing datasets GSE130036 and GSE141910 from the Gene Expression Omnibus (GEO) database. It detected necroptosis-linked differentially expressed genes (NRDEGs) by reviewing both the gene set for necroptosis and the differently expressed genes (DEGs). The enriched signaling pathway of HCM was assessed using GSEA, while common DEGs were studied through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Concurrently, the Protein-Protein Interaction network (PPI) proved useful for identifying central genes. CIBERSORT facilitated evaluating the correlation between distinct immune cell-type prevalence and NRDEGs by analyzing immune infiltration patterns. Lastly, GSE141910 dataset validated the expression ranks of NRDEGs and immune-cell penetration. Results The investigation disclosed significant enrichment and activation of the necroptosis pathway in HCM specimens. Seventeen diverse genes, including CYBB, BCL2, and JAK2 among others, were identified in the process. PPI network scrutiny classified nine of these genes as central genes. Results from GO and KEGG enrichment analyses showed substantial connections of these genes to pathways pertaining to the HIF-1 signaling track, necroptosis, and NOD-like receptor signaling process. Moreover, an imbalance in M2 macrophage cells in HCM samples was observed. Finally, CYBB, BCL2, and JAK2 emerged as vital genes and were validated using the GSE141910 dataset. Conclusion These results indicate necroptosis as a probable underlying factor in HCM, with immune cell infiltration playing a part. Additionally, CYBB, BCL2, JAK2 could act as potential biomarkers for recognizing HCM. This information forms crucial insights into the basic mechanisms of HCM and could enhance its diagnosis and management.
Collapse
Affiliation(s)
| | | | - Fang Jia
- Department of Cardiovascular Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
10
|
Bonaventura J, Rowin EJ, Chan RH, Chin MT, Puchnerova V, Polakova E, Macek M, Votypka P, Batorsky R, Perera G, Koethe B, Veselka J, Maron BJ, Maron MS. Relationship Between Genotype Status and Clinical Outcome in Hypertrophic Cardiomyopathy. J Am Heart Assoc 2024; 13:e033565. [PMID: 38757491 PMCID: PMC11179794 DOI: 10.1161/jaha.123.033565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND The genetic basis of hypertrophic cardiomyopathy (HCM) is complex, and the relationship between genotype status and clinical outcome is incompletely resolved. METHODS AND RESULTS We assessed a large international HCM cohort to define in contemporary terms natural history and clinical consequences of genotype. Consecutive patients (n=1468) with established HCM diagnosis underwent genetic testing. Patients with pathogenic (or likely pathogenic) variants were considered genotype positive (G+; n=312; 21%); those without definite disease-causing mutations (n=651; 44%) or variants of uncertain significance (n=505; 35%) were considered genotype negative (G-). Patients were followed up for a median of 7.8 years (interquartile range, 3.5-13.4 years); HCM end points were examined by cumulative event incidence. Over follow-up, 135 (9%) patients died, 33 from a variety of HCM-related causes. After adjusting for age, all-cause and HCM-related mortality did not differ between G- versus G+ patients (hazard ratio [HR], 0.78 [95% CI, 0.46-1.31]; P=0.37; HR, 0.93 [95% CI, 0.38-2.30]; P=0.87, respectively). Adverse event rates, including heart failure progression to class III/IV, heart transplant, or heart failure death, did not differ (G- versus G+) when adjusted for age (HR, 1.20 [95% CI, 0.63-2.26]; P=0.58), nor was genotype independently associated with sudden death event risk (HR, 1.39 [95% CI, 0.88-2.21]; P=0.16). In multivariable analysis, age was the only independent predictor of all-cause and HCM-related mortality, heart failure progression, and sudden death events. CONCLUSIONS In this large consecutive cohort of patients with HCM, genotype (G+ or G-) was not a predictor of clinical course, including all-cause and HCM-related mortality and risk for heart failure progression or sudden death. G+ status should not be used to dictate clinical management or predict outcome in HCM.
Collapse
Affiliation(s)
- Jiri Bonaventura
- Department of Cardiology, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
- Hypertrophic Cardiomyopathy CenterLahey Hospital and Medical CenterBurlingtonMAUSA
| | - Ethan J. Rowin
- Hypertrophic Cardiomyopathy CenterLahey Hospital and Medical CenterBurlingtonMAUSA
| | - Raymond H. Chan
- Division of Cardiology, Peter Munk Cardiac CentreToronto General Hospital, University Health NetworkOntarioCanada
| | - Michael T. Chin
- Molecular Cardiology Research InstituteTufts Medical CenterBostonMAUSA
| | - Veronika Puchnerova
- Department of Cardiology, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Eva Polakova
- Department of Cardiology, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Milan Macek
- Department of Biology and Medical Genetics, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Pavel Votypka
- Department of Biology and Medical Genetics, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Rebecca Batorsky
- Molecular Cardiology Research InstituteTufts Medical CenterBostonMAUSA
| | - Gayani Perera
- Molecular Cardiology Research InstituteTufts Medical CenterBostonMAUSA
| | - Benjamin Koethe
- Institute for Clinical Research and Health Policy Studies, Tufts Medical CenterBostonMAUSA
| | - Josef Veselka
- Department of Cardiology, 2nd Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Barry J. Maron
- Hypertrophic Cardiomyopathy CenterLahey Hospital and Medical CenterBurlingtonMAUSA
| | - Martin S. Maron
- Hypertrophic Cardiomyopathy CenterLahey Hospital and Medical CenterBurlingtonMAUSA
| |
Collapse
|
11
|
Pepe G, Appierdo R, Ausiello G, Helmer-Citterich M, Gherardini PF. A Meta-Analysis Approach to Gene Regulatory Network Inference Identifies Key Regulators of Cardiovascular Diseases. Int J Mol Sci 2024; 25:4224. [PMID: 38673810 PMCID: PMC11049946 DOI: 10.3390/ijms25084224] [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: 03/08/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) represent a major concern for global health, whose mechanistic understanding is complicated by a complex interplay between genetic predisposition and environmental factors. Specifically, heart failure (HF), encompassing dilated cardiomyopathy (DC), ischemic cardiomyopathy (ICM), and hypertrophic cardiomyopathy (HCM), is a topic of substantial interest in basic and clinical research. Here, we used a Partial Correlation Coefficient-based algorithm (PCC) within the context of a meta-analysis framework to construct a Gene Regulatory Network (GRN) that identifies key regulators whose activity is perturbed in Heart Failure. By integrating data from multiple independent studies, our approach unveiled crucial regulatory associations between transcription factors (TFs) and structural genes, emphasizing their pivotal roles in regulating metabolic pathways, such as fatty acid metabolism, oxidative stress response, epithelial-to-mesenchymal transition, and coagulation. In addition to known associations, our analysis also identified novel regulators, including the identification of TFs FPM315 and OVOL2, which are implicated in dilated cardiomyopathies, and TEAD1 and TEAD2 in both dilated and ischemic cardiomyopathies. Moreover, we uncovered alterations in adipogenesis and oxidative phosphorylation pathways in hypertrophic cardiomyopathy and discovered a role for IL2 STAT5 signaling in heart failure. Our findings underscore the importance of TF activity in the initiation and progression of cardiac disease, highlighting their potential as pharmacological targets.
Collapse
Affiliation(s)
- Gerardo Pepe
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.P.); (R.A.)
| | - Romina Appierdo
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.P.); (R.A.)
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Gabriele Ausiello
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.P.); (R.A.)
| | | | | |
Collapse
|
12
|
Chen P, Yawar W, Farooqui AR, Ali S, Lathiya N, Ghous Z, Sultan R, Alhomrani M, Alghamdi SA, Almalki AA, Alghamdi AA, ALSuhaymi N, Razi Ul Islam Hashmi M, Hameed Y. Transcriptomics data integration and analysis to uncover hallmark genes in hypertrophic cardiomyopathy. Am J Transl Res 2024; 16:637-653. [PMID: 38463581 PMCID: PMC10918138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is a heterogeneous disease that mainly affects the myocardium. In the current study, we aim to explore HCM-related hub genes through the analysis of differentially expressed genes (DEGs) between HCM and normal sample groups. METHODS The GSE68316 and GSE36961 expression profiles were obtained from the Gene Expression Omnibus (GEO) database for the identification of DEGs, to explore hub genes, and to perform their expression analysis. Clinical HCM and control tissue samples were taken for expression and promoter methylation validation analysis via RNA-sequencing (RNA-seq) and targeted bisulfite sequencing (bisulfite-seq) analyses. Then, other different bioinformatics tools were employed to perform STRING, lncRNA-miRNA-mRNA regulatory networks, gene enrichment, and drug prediction analyses. RESULTS In total, the top 20 DEGs, including 10 up-regulated and 10 down-regulated, were obtained from GSE68316. Out of the 20 DEGs, we subsequently identified the 8 most important hub genes including 5 up-regulated genes (EPB42, UQCRH, CA1, PFDN5, and LSM5) and 3 down-regulated genes (RPS24, TNS1, and RPL26). Expression and promoter methylation dysregulation of these genes were further validated on clinical HCM samples paired with controls. Next, we further investigated hub genes' regulatory 6 miRNAs (has-mir-1-3p, has-mir-129-5p, has-mir-16-5p, has-mir-23b-3p, has-mir-27-3p, and has-mir-182-5p) and miRNAs regulatory 4 lncRNAs (NUTMB2-AS1, NEAT1, XIST, and GABPB1-AS1) in this study via the lncRNA-cricRNA-miRNA-mRNA regulatory network. Later on, gene enrichment analysis revealed that hub genes were enriched in various important pathways including Nitrogen metabolism, Ribosome, RNA degradation, Cardiac muscle contraction, and Coronavirus disease, etc. Finally, the drug prediction analysis highlighted different potential candidate drugs for altering the expression of hub genes in the treatment of HCM. CONCLUSION In summary, the identification of key hub genes and their enrichment analysis in the current study may shed light on the mechanisms behind the occurrence and development of HCM.
Collapse
Affiliation(s)
- Peng Chen
- Department of Cardiovascular Medicine, Taiyuan Central HospitalTaiyuan 030000, Shanxi, China
| | - Warda Yawar
- Department of Emergency, PPHISindh, Karachi 74800, Pakistan
| | | | - Saqib Ali
- Department of Computer Science, University of AgricultureFaisalabad 38040, Pakistan
| | - Nida Lathiya
- Department of Physiology, Jinnah Medical and Dental College, Sohail UniversityKarachi 74800, Pakistan
| | - Zeeshan Ghous
- Department of Cardiology, Punjab Institute of CardiologyLahore 54000, Pakistan
| | - Rizwana Sultan
- Department of Pathology, Faculty of Veterinary and Animal Sciences, Cholistan University of Veterinary and Animal SciencesBahawalpur, Pakistan
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Saleh A Alghamdi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Abdulraheem Ali Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Ahmad A Alghamdi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura UniversityMekkah, Saudi Arabia
| | | | - Yasir Hameed
- Department of Biotechnology, Institute of Biochemistry Biotechnology and Bioinformatics, The Islamia University of BahawalpurBahawalpur 63100, Pakistan
| |
Collapse
|
13
|
Kwak S, Lee HJ, Lee H, Park JB, Kim YJ, Han K, Kim HK. Age-dependent association of metabolic dyslipidemia with clinical expression of hypertrophic cardiomyopathy. Int J Cardiol 2024; 396:131574. [PMID: 37935337 DOI: 10.1016/j.ijcard.2023.131574] [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: 08/23/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Previous studies have shown that the clinical expression of hypertrophic cardiomyopathy (HCM) can be determined by obesity and metabolic syndrome. The present study aimed to investigate the association between triglyceride and high-density lipoprotein cholesterol (HDLC) level, the two dyslipidemia-related components of metabolic syndrome, and the incidence of HCM. We also explored an age-dependent association between them. METHODS Individuals without previous HCM diagnosis who underwent a designated national health examination in 2009 were recruited. Individuals who used lipid-lowering medications within 1-year of the baseline were excluded. The outcome of interest was a newly diagnosed HCM. RESULTS Our cohort consisted of 8,652,709 individuals (mean 46 years, 55.6% men). During the median 9.3 years of follow-up, 5932 (0.07%) individuals were newly diagnosed with HCM. There was a gradual increase in the incidence of HCM towards higher triglyceride and lower HDL-C levels (log-rank p < 0.001). When stratified by age, the incidence of HCM was highest in individuals aged ≥65 years, followed by those aged 40-64 and 20-39 years (0.22% vs. 0.07% vs. 0.03%, log-rank p < 0.001). In individuals aged 20-39 years, a higher triglyceride level was associated with a higher incidence of HCM (i.e., ≥200 vs. <100 mg/dL: adjusted hazard ratio 2.28, 95% confidence interval 1.89-2.75), whereas there was no significant association in older groups (p-for-interaction<0.001). Similarly, a lower HDL-C level was associated with a higher incidence of HCM, particularly in individuals aged 20-39 years (p-for-interaction = 0.001). CONCLUSIONS High triglyceride and low HDL-C levels are associated with a higher incidence of HCM, particularly in young individuals.
Collapse
Affiliation(s)
- Soongu Kwak
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyun-Jung Lee
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Heesun Lee
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jun-Bean Park
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yong-Jin Kim
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea.
| | - Hyung-Kwan Kim
- Department of Internal medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Matthews EL, Guers JJ, Ramick MG, Hosick PA. Inverse Association between Exercising Blood Pressure Response and Left Ventricular Chamber Size and Mass in Women Who Habitually Resistance Train. Healthcare (Basel) 2024; 12:353. [PMID: 38338238 PMCID: PMC10855375 DOI: 10.3390/healthcare12030353] [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: 12/04/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Exercise is a major modifiable lifestyle factor that leads to temporarily increased systolic blood pressure (SBP), which is thought to influence left ventricular mass normalized to body surface area (LVM/BSA). This relationship has never been studied in women who habitually perform resistance exercise. PURPOSE To determine if a direct correlation exists between the SBP response to resistance exercise (change from rest; eSBP) and LVM/BSA in young healthy women who habitually resistance train. METHODS Leg extension resistance exercise was performed while continuously monitoring blood pressure using finger plethysmography. LVM was estimated using echocardiography. Data are shown as mean ± SD. RESULTS Thirty-one women participated (age 23 ± 3 years, height 164 ± 7 cm, body mass 63.7 ± 10.3 kg). Resting SBP (110 ± 8 mmHg, r = 0.355, p = 0.049) was shown to be directly correlated to LVM/BSA (72.0 ± 28.4 g/m2). Conversely, eSBP (30.8 ± 14.6 ∆mmHg, r = -0.437, p = 0.014) was inversely related to LVM/BSA. eSBP was not correlated to interventricular septum width (0.88 ± 0.12 cm, r = -0.137, p = 0.463) or posterior wall thickness (0.91 ± 0.15 cm, r = -0.084, p = 0.654). eSBP was inversely related to left ventricle internal diameter during diastole (LVIDd) (4.25 ± 0.33 cm, r = -0.411, p = 0.021). CONCLUSION Counter to the hypothesis, these data suggest an inverse association between eSBP during resistance exercise and LVM/BSA in healthy young women who resistance train. This relationship is due to a smaller LVIDd with greater eSBP.
Collapse
Affiliation(s)
- Evan L. Matthews
- Department of Exercise Science and Physical Education, Montclair State University, Montclair, NJ 07043, USA;
| | - John J. Guers
- Department of Biology, Behavioral Neuroscience and Health Sciences, Rider University, Lawrenceville, NJ 08648, USA;
| | - Meghan G. Ramick
- Department of Kinesiology, West Chester University, West Chester, PA 19383, USA;
| | - Peter A. Hosick
- Department of Exercise Science and Physical Education, Montclair State University, Montclair, NJ 07043, USA;
| |
Collapse
|
15
|
Liao X, Zhou S, Zeng D, Ying W, Lian D, Zhang M, Ge J, Chen M, Liu Y, Lin Y. Roles of the crucial mitochondrial DNA in hypertrophic cardiomyopathy prognosis and diagnosis: A review. Medicine (Baltimore) 2023; 102:e36368. [PMID: 38050313 PMCID: PMC10695538 DOI: 10.1097/md.0000000000036368] [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: 07/18/2023] [Accepted: 11/08/2023] [Indexed: 12/06/2023] Open
Abstract
Mitochondrial DNA is implicated in hypertrophic cardiomyopathy (HCM) development. We aimed to identify valuable mtDNAs that contribute to the development of HCM. Differentially expressed mitochondrial DNAs (DEMGs) between HCM and controls were screened. GO and KEGG functional enrichment analyses were performed, and the optimum genes were explored using the LASSO regression mode and SVM-RFE model. A diagnostic scoring model was constructed and verified using ROC curves. Mitochondria-based subtypes were identified. Immune performance among the subtypes including immune cells, immune checkpoint genes, and HLA family genes was analyzed. Finally, an mRNA-transcription factor (TF)-miRNA network was constructed using Cytoscape software. Twelve DEMGs in HCM were selected. Among them, 6 DEMGs, including PDK4, MGST1, TOMM40, LYPLAL1, GATM, and CPT1B were demonstrated as DEMGs at the point of intersection of Lasso regression and SVM-RFE. The ROC of the model for the training and validation datasets was 0.999 and 0.958, respectively. Two clusters were divided, and 4 immune cell types were significantly different between the 2 clusters, including resting mast cells, macrophages M2, and plasma cells. Nine upregulated KEGG pathways were enriched in cluster 1 vs. cluster 2 including O-glycan biosynthesis, the ErbB signaling pathway, and the GnRH signaling pathway. Meanwhile, 49 down-regulated pathways were enriched such as the toll-like signaling pathway and natural killer cell-mediated cytotoxicity pathway. The 6 gene-based mRNA-TF-miRNA networks included other 133 TFs and 18 miRNAs. Six DEMGs in HCM, including PDK4, MGST1, TOMM40, LYPLAL1, GATM, and CPT1B, can be indicative of HCM prognosis or disease progression.
Collapse
Affiliation(s)
- Xuewen Liao
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou City, China
| | - Shunkai Zhou
- Department of Thoracic and Cardiac Surgery, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Dehua Zeng
- Department of Pathology, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Wenmin Ying
- Department of Radiotherapy, Fuding Hospital, Fuding City, China
| | - Duohuang Lian
- Department of Thoracic and Cardiac Surgery, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Meiqing Zhang
- Department of Thoracic and Cardiac Surgery, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Jianjun Ge
- Department of Thoracic Surgery, No. 2 Hospital of Nanping City, Nanping City, China
| | - Mengmeng Chen
- Department of Thoracic and Cardiac Surgery, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Yaming Liu
- Department of Thoracic and Cardiac Surgery, 900th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Fuzhou City, China
| | - Yazhou Lin
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou City, China
| |
Collapse
|
16
|
Hilderink S, Schuldt M, Goebel M, Jansen VJ, Manders E, Moorman S, Dorsch LM, van Steenbeek FG, van der Velden J, Kuster DWD. Characterization of heterozygous and homozygous mouse models with the most common hypertrophic cardiomyopathy mutation MYBPC3 c.2373InsG in the Netherlands. J Mol Cell Cardiol 2023; 185:65-76. [PMID: 37844837 DOI: 10.1016/j.yjmcc.2023.10.008] [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: 01/25/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in the cardiac myosin binding protein-C (cMyBP-C) encoding gene MYBPC3. In the Netherlands, approximately 25% of patients carry the MYBPC3c.2373InsG founder mutation. Most patients are heterozygous (MYBPC3+/InsG) and have highly variable phenotypic expression, whereas homozygous (MYBPC3InsG/InsG) patients have severe HCM at a young age. To improve understanding of disease progression and genotype-phenotype relationship based on the hallmarks of human HCM, we characterized mice with CRISPR/Cas9-induced heterozygous and homozygous mutations. At 18-28 weeks of age, we assessed the cardiac phenotype of Mybpc3+/InsG and Mybpc3InsG/InsG mice with echocardiography, and performed histological analyses. Cytoskeletal proteins and cardiomyocyte contractility of 3-4 week old and 18-28 week old Mybpc3c.2373InsG mice were compared to wild-type (WT) mice. Expectedly, knock-in of Mybpc3c.2373InsG resulted in the absence of cMyBP-C and our 18-28 week old homozygous Mybpc3c.2373InsG model developed cardiac hypertrophy and severe left ventricular systolic and diastolic dysfunction, whereas HCM was not evident in Mybpc3+/InsG mice. Mybpc3InsG/InsG cardiomyocytes also presented with slowed contraction-relaxation kinetics, to a greater extent in 18-28 week old mice, partially due to increased levels of detyrosinated tubulin and desmin, and reduced cardiac troponin I (cTnI) phosphorylation. Impaired cardiomyocyte contraction-relaxation kinetics were successfully normalized in 18-28 week old Mybpc3InsG/InsG cardiomyocytes by combining detyrosination inhibitor parthenolide and β-adrenergic receptor agonist isoproterenol. Both the 3-4 week old and 18-28 week old Mybpc3InsG/InsG models recapitulate HCM, with a severe phenotype present in the 18-28 week old model.
Collapse
Affiliation(s)
- Sarah Hilderink
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Maike Schuldt
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Max Goebel
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Valentijn J Jansen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Emmy Manders
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands
| | - Stan Moorman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Larissa M Dorsch
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Frank G van Steenbeek
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands; Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, the Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Mechanical Resistance to Micro-Heart Tissue Contractility unveils early Structural and Functional Pathology in iPSC Models of Hypertrophic Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564856. [PMID: 37961198 PMCID: PMC10634965 DOI: 10.1101/2023.10.30.564856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Hypertrophic cardiomyopathy is the most common cause of sudden death in the young. Because the disease exhibits variable penetrance, there are likely nongenetic factors that contribute to the manifestation of the disease phenotype. Clinically, hypertension is a major cause of morbidity and mortality in patients with HCM, suggesting a potential synergistic role for the sarcomeric mutations associated with HCM and mechanical stress on the heart. We developed an in vitro physiological model to investigate how the afterload that the heart muscle works against during contraction acts together with HCM-linked MYBPC3 mutations to trigger a disease phenotype. Micro-heart muscle arrays (μHM) were engineered from iPSC-derived cardiomyocytes bearing MYBPC3 loss-of-function mutations and challenged to contract against mechanical resistance with substrates stiffnesses ranging from the of embryonic hearts (0.4 kPa) up to the stiffness of fibrotic adult hearts (114 kPa). Whereas MYBPC3 +/- iPSC-cardiomyocytes showed little signs of disease pathology in standard 2D culture, μHMs that included components of afterload revealed several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in the MYBPC3 +/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca 2+ intake through membrane-embedded channels, rather than sarcoplasmic reticulum Ca 2+ ATPase (SERCA) dysfunction or Ca 2+ buffering at myofilaments underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease mechanisms with iPSC technology.
Collapse
|
18
|
Schlittler M, Pramstaller PP, Rossini A, De Bortoli M. Myocardial Fibrosis in Hypertrophic Cardiomyopathy: A Perspective from Fibroblasts. Int J Mol Sci 2023; 24:14845. [PMID: 37834293 PMCID: PMC10573356 DOI: 10.3390/ijms241914845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease and the leading cause of sudden cardiac death in young people. Mutations in genes that encode structural proteins of the cardiac sarcomere are the more frequent genetic cause of HCM. The disease is characterized by cardiomyocyte hypertrophy and myocardial fibrosis, which is defined as the excessive deposition of extracellular matrix proteins, mainly collagen I and III, in the myocardium. The development of fibrotic tissue in the heart adversely affects cardiac function. In this review, we discuss the latest evidence on how cardiac fibrosis is promoted, the role of cardiac fibroblasts, their interaction with cardiomyocytes, and their activation via the TGF-β pathway, the primary intracellular signalling pathway regulating extracellular matrix turnover. Finally, we summarize new findings on profibrotic genes as well as genetic and non-genetic factors involved in the pathophysiology of HCM.
Collapse
Affiliation(s)
| | | | | | - Marzia De Bortoli
- Eurac Research, Institute for Biomedicine (Affiliated to the University of Lübeck), 39100 Bolzano, Italy
| |
Collapse
|
19
|
Anderson EL, Côté É, Burton S, Saleh T. Serum C-terminal telopeptide of Type-I collagen (CTx) concentration and myocardial hyperechogenicity in cats with hypertrophic cardiomyopathy. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2023; 64:864-870. [PMID: 37663017 PMCID: PMC10426254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Objective This study sought to determine the serum concentrations of C-terminal telopeptide of Type-I collagen (CTx), a marker of collagen degradation, in a hospital population of cats with hypertrophic cardiomyopathy (HCM). The study also evaluated the prevalence of myocardial hyperechogenicity of the left ventricle (LV) in the same cats. Animals and procedure Cats brought to a university veterinary cardiology service entered the study when they had an echocardiographic diagnosis of HCM; echocardiographically normal cats served as controls. Serum CTx concentrations were assessed using ELISA. Results There was no difference in serum CTx concentrations between cats with HCM and controls (HCM: median 0.248 ng/mL, controls: median 0.253 ng/mL; P = 0.4). Significantly more cats with HCM (60%) showed echocardiographic LV myocardial hyperechogenicity compared to normal controls (17%; P = 0.0057), but serum CTx concentrations were not different between these 2 groups. Conclusion and clinical relevance These results indicate that, as in human patients with HCM and in contrast to earlier feline studies, there was no evidence of enhanced collagen degradation indicated by serum CTx concentrations in cats with HCM compared to normal controls.
Collapse
Affiliation(s)
- Erin L Anderson
- Departments of Companion Animals (Anderson, Côté), Pathology and Microbiology (Burton), and Biomedical Sciences (Saleh), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Étienne Côté
- Departments of Companion Animals (Anderson, Côté), Pathology and Microbiology (Burton), and Biomedical Sciences (Saleh), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Shelley Burton
- Departments of Companion Animals (Anderson, Côté), Pathology and Microbiology (Burton), and Biomedical Sciences (Saleh), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Tarek Saleh
- Departments of Companion Animals (Anderson, Côté), Pathology and Microbiology (Burton), and Biomedical Sciences (Saleh), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| |
Collapse
|
20
|
Bonaventura J, Rowin EJ, Maron MS, Maron BJ. Is Hypertrophic Cardiomyopathy Always a Familial and Inherited Disease? J Am Heart Assoc 2023:e028974. [PMID: 37301765 DOI: 10.1161/jaha.122.028974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 05/15/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Jiri Bonaventura
- Hypertrophic Cardiomyopathy Center Lahey Hospital and Medical Center Burlington MA USA
- Department of Cardiology, 2nd Faculty of Medicine Charles University and Motol University Hospital Prague Czech Republic
| | - Ethan J Rowin
- Hypertrophic Cardiomyopathy Center Lahey Hospital and Medical Center Burlington MA USA
| | - Martin S Maron
- Hypertrophic Cardiomyopathy Center Lahey Hospital and Medical Center Burlington MA USA
| | - Barry J Maron
- Hypertrophic Cardiomyopathy Center Lahey Hospital and Medical Center Burlington MA USA
| |
Collapse
|
21
|
Wang RS, Maron BA, Loscalzo J. Multiomics Network Medicine Approaches to Precision Medicine and Therapeutics in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2023; 43:493-503. [PMID: 36794589 PMCID: PMC10038904 DOI: 10.1161/atvbaha.122.318731] [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: 11/07/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide and display complex phenotypic heterogeneity caused by many convergent processes, including interactions between genetic variation and environmental factors. Despite the identification of a large number of associated genes and genetic loci, the precise mechanisms by which these genes systematically influence the phenotypic heterogeneity of CVD are not well understood. In addition to DNA sequence, understanding the molecular mechanisms of CVD requires data from other omics levels, including the epigenome, the transcriptome, the proteome, as well as the metabolome. Recent advances in multiomics technologies have opened new precision medicine opportunities beyond genomics that can guide precise diagnosis and personalized treatment. At the same time, network medicine has emerged as an interdisciplinary field that integrates systems biology and network science to focus on the interactions among biological components in health and disease, providing an unbiased framework through which to integrate systematically these multiomics data. In this review, we briefly present such multiomics technologies, including bulk omics and single-cell omics technologies, and discuss how they can contribute to precision medicine. We then highlight network medicine-based integration of multiomics data for precision medicine and therapeutics in CVD. We also include a discussion of current challenges, potential limitations, and future directions in the study of CVD using multiomics network medicine approaches.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Division of Cardiovascular Medicine
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Joseph Loscalzo
- Division of Cardiovascular Medicine
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Maurer C, Boleti O, Najarzadeh Torbati P, Norouzi F, Fowler ANR, Minaee S, Salih KH, Taherpour M, Birjandi H, Alizadeh B, Salih AF, Bijari M, Houlden H, Pittman AM, Maroofian R, Almashham YH, Karimiani EG, Kaski JP, Faqeih EA, Vakilian F, Jamshidi Y. Genetic Insights from Consanguineous Cardiomyopathy Families. Genes (Basel) 2023; 14:182. [PMID: 36672924 PMCID: PMC9858866 DOI: 10.3390/genes14010182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Inherited cardiomyopathies are a prevalent cause of heart failure and sudden cardiac death. Both hypertrophic (HCM) and dilated cardiomyopathy (DCM) are genetically heterogeneous and typically present with an autosomal dominant mode of transmission. Whole exome sequencing and autozygosity mapping was carried out in eight un-related probands from consanguineous Middle Eastern families presenting with HCM/DCM followed by bioinformatic and co-segregation analysis to predict the potential pathogenicity of candidate variants. We identified homozygous missense variants in TNNI3K, DSP, and RBCK1 linked with a dilated phenotype, in NRAP linked with a mixed phenotype of dilated/hypertrophic, and in KLHL24 linked with a mixed phenotype of dilated/hypertrophic and non-compaction features. Co-segregation analysis in family members confirmed autosomal recessive inheritance presenting in early childhood/early adulthood. Our findings add to the mutational spectrum of recessive cardiomyopathies, supporting inclusion of KLHL24, NRAP and RBCK1 as disease-causing genes. We also provide evidence for novel (recessive) modes of inheritance of a well-established gene TNNI3K and expand our knowledge of the clinical heterogeneity of cardiomyopathies. A greater understanding of the genetic causes of recessive cardiomyopathies has major implications for diagnosis and screening, particularly in underrepresented populations, such as those of the Middle East.
Collapse
Affiliation(s)
- Constance Maurer
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Olga Boleti
- Centre for Paediatric Inherited and Rare Cardiovascular Disease, University College London and Great Ormond Street Hospital, London WC1N 1DZ, UK
| | | | - Farzaneh Norouzi
- Department of Cardiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Anna Nicole Rebekah Fowler
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Shima Minaee
- Department of Cardiovascular Diseases, Razavi Hospital, Mashhad 9177948954, Iran
| | - Khalid Hama Salih
- Department of Pediatrics, College of Medicine, Sulaimani University, Sulaymaniyah 46001, Iraq
| | - Mehdi Taherpour
- Department of Cardiovascular Diseases, Razavi Hospital, Mashhad 9177948954, Iran
| | - Hassan Birjandi
- Division of Congenital and Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Behzad Alizadeh
- Division of Congenital and Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Aso Faeq Salih
- Department of Pediatrics, College of Medicine, Sulaimani University, Sulaymaniyah 46001, Iraq
| | - Moniba Bijari
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Alan Michael Pittman
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Reza Maroofian
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Yahya H. Almashham
- Pediatric Cardiology, King Salman Heart Center, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Ehsan Ghayoor Karimiani
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
- Department of Medical Genetics, Next Generation Genetic Polyclinic, Mashhad 009851, Iran
| | - Juan Pablo Kaski
- Centre for Paediatric Inherited and Rare Cardiovascular Disease, University College London and Great Ormond Street Hospital, London WC1N 1DZ, UK
| | - Eissa Ali Faqeih
- Section of Medical Genetics, Children’s Specialist Hospital, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Farveh Vakilian
- Department of Cardiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Yalda Jamshidi
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| |
Collapse
|
23
|
Subtypes and Mechanisms of Hypertrophic Cardiomyopathy Proposed by Machine Learning Algorithms. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101566. [PMID: 36294999 PMCID: PMC9605444 DOI: 10.3390/life12101566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a relatively common inherited cardiac disease that results in left ventricular hypertrophy. Machine learning uses algorithms to study patterns in data and develop models able to make predictions. The aim of this study is to identify HCM subtypes and examine the mechanisms of HCM using machine learning algorithms. Clinical and laboratory findings of 143 adult patients with a confirmed diagnosis of nonobstructive HCM are analyzed; HCM subtypes are determined by clustering, while the presence of different HCM features is predicted in classification machine learning tasks. Four clusters are determined as the optimal number of clusters for this dataset. Models that can predict the presence of particular HCM features from other genotypic and phenotypic information are generated, and subsets of features sufficient to predict the presence of other features of HCM are determined. This research proposes four subtypes of HCM assessed by machine learning algorithms and based on the overall phenotypic expression of the participants of the study. The identified subsets of features sufficient to determine the presence of particular HCM aspects could provide deeper insights into the mechanisms of HCM.
Collapse
|