1
|
Liu Y, Jiang Y, Ma T, Dong W, Yang P, Peng L, Wang B, Wu C, Li Z, Zhang H, Sun Y, Niu Y, Ding Y. Cardiomyocyte-specific activation of the sarcomere-localized Dnajb6b chaperone causes cardiomyopathy and heart failure through upregulated sarcoplasmic reticulum stress. Life Sci 2025; 374:123711. [PMID: 40360088 DOI: 10.1016/j.lfs.2025.123711] [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: 02/09/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
AIMS Despite abundant expression of DNAJB6 gene in the heart, its roles in cardiac diseases remain underexplored. We aimed to investigate the function of its zebrafish (Danio rerio) ortholog, the dnajb6b gene, in cardiomyopathy and heart failure. MATERIALS AND METHODS Both loss-of-function mutation and gain-of-function transgenic approaches were employed in zebrafish. High frequency echocardiography was performed to evaluate cardiac function indices in adult zebrafish. 4-phenylbutyric acid (4-PBA) was used to pharmacologically inhibit sarcoplasmic reticulum (SR) stress in zebrafish. Western blot was carried out to determine expression of DNAJB6 isoforms in human patients' heart tissues. KEY FINDINGS Global loss-of-function mutations affecting both the sarcomere-localized short (Dnajb6b[S]) and nucleus-localized long (Dnajb6b[L]) isoforms appeared phenotypically normal. In contrast, cardiomyocyte-specific overexpression of a truncated, sarcomere-localized Dnajb6b(L) isoform (Dnajb6b[∆L]) led to severe cardiomyopathy and heart failure phenotypes. Mechanistically, Dnajb6b responded to sarcoplasmic reticulum (SR) stress and activation of Dnajb6b(∆L) resulted in elevated SR stress, accumulation of ubiquitinated protein aggregation, and aberrant activation of autophagy. 4-PBA treatment partially rescued cardiac dysfunction and extended the lifespan of zebrafish with cardiomyocyte-specific activation of Dnajb6b(∆L). Finally, elevated expression of both DNAJB6(S) and DNAJB6(L) isoforms was detected in failing human hearts, supporting their clinical relevance. SIGNIFICANCE Gain-of-function mutation in Dnajb6b(∆L) isoform causes cardiomyopathy and heart failure, likely mediated by elevated SR stress. This study enhances our understanding of Dnajb6's role in cardiac proteostasis and highlights its potential as a therapeutic target for the treatment of cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Yuting Liu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yajie Jiang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Taiwei Ma
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Wenjing Dong
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Peng Yang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lixia Peng
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Baokun Wang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Hong Zhang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yuanchao Sun
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yujuan Niu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yonghe Ding
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
2
|
Qu KY, Cheng HY, Qiao L, Jiao JC, Chang SQ, Peng XF, Cui C, Zhang F, Huang NP. Construction of engineered cardiac tissue on a heart-on-a-chip device enables modeling of arrhythmogenic right ventricular cardiomyopathy. Biosens Bioelectron 2025; 281:117478. [PMID: 40245609 DOI: 10.1016/j.bios.2025.117478] [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: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive cardiac disorder characterized by the replacement of the right ventricular myocardium with fibrofatty tissue, with an incidence rate of approximately 1 in 5000. To advance our understanding of its pathology and facilitate drug screening, there is an urgent need for myocardial models that closely replicate human physiological conditions. In this study, we developed an engineered cardiac tissue (ECT) model on a chip using cardiomyocytes differentiated from induced pluripotent stem cells (iPSCs) derived from ARVC patients. The disease ECT model successfully recapitulated key phenotypic features of ARVC, including reduced contractility, arrhythmic events, and abnormal calcium transients. We further assessed the drug responses of the model to isoproterenol and amiodarone, confirming increased sensitivity to isoproterenol in the ARVC model, while amiodarone effectively alleviated the arrhythmic events. In conclusion, our ECT model successfully reproduced ARVC phenotypes, providing a novel platform for drug screening and pathological studies.
Collapse
Affiliation(s)
- Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hong-Yi Cheng
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jin-Cheng Jiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Shi-Qi Chang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xia-Feng Peng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Chang Cui
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Feng Zhang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
3
|
Xiong X, Lee HC, Lu T. Impact of Sorbs2 dysfunction on cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167813. [PMID: 40139410 PMCID: PMC12037213 DOI: 10.1016/j.bbadis.2025.167813] [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: 09/05/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Despite significant advancements in prevention and treatment over the past decades, cardiovascular diseases (CVDs) remain the leading cause of death worldwide. CVDs involve multifactorial inheritance, but our understanding of the genetic impact on these diseases is still incomplete. Sorbin and SH3 domain-containing protein 2 (Sorbs2) is ubiquitously expressed in various tissues, including the cardiovascular system. Increasing evidence suggests that Sorbs2 malfunction contributes to CVDs. This manuscript will review our current understanding of the potential mechanisms underlying Sorbs2 dysregulation in the development of CVDs.
Collapse
Affiliation(s)
- Xiaowei Xiong
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Hon-Chi Lee
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Tong Lu
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
4
|
Shetty NS, Pampana A, Gaonkar M, Patel N, Vekariya N, Smith JG, Kalra R, Chahal CAA, Semsarian C, Li P, Arora G, Arora P. Association of Pathogenic/Likely Pathogenic Genetic Variants for Cardiomyopathies With Clinical Outcomes: A Multiancestry Analysis in the All of Us Research Program. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025:e005113. [PMID: 40433684 DOI: 10.1161/circgen.124.005113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND This study aimed to evaluate the prevalence of pathogenic/likely pathogenic cardiomyopathy variant carriers in a multiancestry US population and examine the risk of adverse clinical outcomes. METHODS This retrospective cohort study included multiancestry US adults aged ≥18 years with sequencing data from the All of Us Research Program. Pathogenic/likely pathogenic variants in cardiomyopathy genes were identified using the ClinVar database. The primary outcome was heart failure. Secondary outcomes included cardiomyopathy and arrhythmia. Outcomes were identified from electronic health records. Interval-censored Cox models, taking age on the timescale, were used to assess the risk of outcomes in pathogenic/likely pathogenic variant carriers with noncarriers as the reference group. RESULTS Among 167 435 individuals (median age, 55.2 [39.5-66.3] years; 61.7% female; 40.7% non-European ancestry) included, the proportion of pathogenic/likely pathogenic cardiomyopathy variant carriers was 0.7% of the overall population and 0.8%, 0.8%, 0.5%, and 1.2% of European, African, East Asian, and South Asian ancestry individuals, respectively. Over the lifetime, there were 12 867 heart failure events (205 in carriers and 12 662 in noncarriers), with an incidence rate of 3.05 (95% CI, 2.66-3.49) per 1000 person-years in carriers and 1.37 (95% CI, 1.35-1.40) in noncarriers (HRadj, 2.30 [95% CI, 2.04-2.60]). Cardiomyopathy occurred in 5164 (161 in carriers and 5003 in noncarriers), with an incidence rate of 2.38 (95% CI, 2.04-2.78) per 1000 person-years among carriers and 0.54 (95% CI, 0.53-0.56) in noncarriers (HRadj, 4.31 [95% CI, 3.73-4.97]). There were 19 405 arrhythmia events (263 in carriers and 19 142 in noncarriers), with an incidence rate of 3.93 (95% CI, 3.48-4.44) per 1000 person-years among carriers and 2.09 (95% CI, 2.06-2.12) in noncarriers (HRadj, 2.12 [95% CI, 1.78-2.53]). CONCLUSIONS Pathogenic/likely pathogenic cardiomyopathy variant carriers have an increased risk of heart failure, cardiomyopathy, and arrhythmias. Despite the modest overall prevalence, the associated risks suggest potential benefits of targeted genetic screening for early detection and management.
Collapse
Affiliation(s)
- Naman S Shetty
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston (N.S.S.)
- Harvard Medical School, Boston, MA (N.S.S.)
| | - Akhil Pampana
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
| | - Mokshad Gaonkar
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
| | - Nirav Patel
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
| | - Nehal Vekariya
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
| | - J Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine and Science for Life Laboratory, University of Gothenburg, Sweden (J.G.S.)
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden (J.G.S.)
- Department of Cardiology, Clinical Sciences, Lund University & Skåne University Hospital (J.G.S.)
- Wallenberg Center for Molecular Medicine, Lund University Diabetes Center, Lund University, Sweden (J.G.S.)
| | - Rajat Kalra
- Cardiovascular Division, University of Minnesota, Minneapolis (R.K.)
| | - C Anwar A Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA (C.A.A.C.)
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (C.A.A.C.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (C.A.A.C.)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, New South Wales, Australia. (C.S.)
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia. (C.S.)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (C.S.)
| | - Peng Li
- School of Nursing, University of Alabama at Birmingham. (P.L.)
| | - Garima Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
| | - Pankaj Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham. (A.P., M.G., N.P., N.V., G.A., P.A.)
- Section of Cardiology, Birmingham Veterans Affairs Medical Center, AL (P.A.)
| |
Collapse
|
5
|
Dababneh S, Roston TM. Evolution of the management of ultrarare inherited arrhythmias and cardiomyopathies. Future Cardiol 2025:1-4. [PMID: 40424207 DOI: 10.1080/14796678.2025.2508568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 05/07/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Affiliation(s)
- Saif Dababneh
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Barriales-Villa R, Escobar-López L, Vilanova Larena D, Salazar-Mendiguchía J, Echeto A, Hernández I, Rebollo-Gómez E, Gimeno JR. Clinical management and healthcare resource utilization among patients with obstructive hypertrophic cardiomyopathy in Spain: a real-world study. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025:S1885-5857(25)00138-0. [PMID: 40334777 DOI: 10.1016/j.rec.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION AND OBJECTIVES Obstructive hypertrophic cardiomyopathy (oHCM), whose symptoms range from dyspnea to heart failure or sudden cardiac death, accounts for approximately 70% of all hypertrophic cardiomyopathy cases. This study aimed to analyze the lack of comprehensive data on oHCM management and determine its clinical and economic burden in Spain. METHODS This retrospective observational study, based on electronic medical records (BIG-PAC), enrolled adults from 1 January 2014, to 31 October 2022, from the time of HCM diagnosis. The analysis focused on epidemiology, patient characteristics and management, transitions between New York Heart Association (NYHA) functional classes, healthcare resource utilization (HCRU), and associated costs. RESULTS A total of 752 oHCM patients were included (mean age: 63 years; male: 57.6%). NYHA functional classification at diagnosis was as follows: 12% NYHA-I, 47.9% NYHA-II, 31.5% NYHA-III, and 8.6% NYHA-IV. The prevalence of HCM and oHCM was 28 and 7 per 10 000 individuals, respectively. Patients received a mean of 2.4 (SD 1.5) treatments, mainly beta-blockers. Only patients in NYHA classes III and IV underwent septal reduction therapies (SRT) (13.1% and 47.7%, respectively); 38.7% and 35.5% of NYHA-III and -IV patients who received SRT, respectively, improved to a lower NYHA class. Symptom severity, as measured by NYHA class, was associated with increased rates of hospitalization, cardiovascular events, mortality, and higher HCRU and costs. Mean annualized, direct, adjusted health care costs ranged from €4142 (95%CI: €3110-€5175) in NYHA-I to €16 677 (95%CI: €15 482-€17 872) in NYHA-IV. CONCLUSIONS This is the first Spanish study to evaluate oHCM patient management and to demonstrate its impact in terms of increased hospitalizations, mortality, HCRU, and healthcare costs, trends that parallel the progression of symptoms by NYHA functional class. Patients who underwent SRT showed partial symptom improvement.
Collapse
Affiliation(s)
- Roberto Barriales-Villa
- Unidad de Cardiopatías Familiares, Servicio de Cardiología, Complexo Hospitalario Universitario A Coruña, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | - Ainara Echeto
- Market Access Department, Bristol-Myers Squibb, Madrid, Spain
| | - Ignacio Hernández
- Health Economics and Outcomes Research (HEOR) department, Atrys Health, Madrid, Spain
| | - Elena Rebollo-Gómez
- Health Economics and Outcomes Research (HEOR) department, Atrys Health, Madrid, Spain
| | - Juan Ramón Gimeno
- Unidad de Cardiopatías Familiares, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| |
Collapse
|
7
|
Xie L, Zhang N, Lv Y. Arrhythmogenic Right Ventricular Cardiomyopathy in Monozygotic Twins-A Case Report. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025; 53:920-923. [PMID: 39868601 DOI: 10.1002/jcu.23925] [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: 09/27/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 01/28/2025]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is the classic phenotype of arrhythmogenic cardiomyopathy. ARVC in twins have been reported rarely. Herein, we report an unusual case of young monozygotic twins with early disease onset presenting different course of disease progression and clinical manifestations. Echocardiography with multiparameters was used for the diagnosis, screening, and follow-up of the disease for the family. Cardiac magnetic resonance revealed the fibrosis distribution in the right ventricular with late gadolinium enhancement. DSG2 mutation was found in the family members, which confirmed the final diagnosis of ARVC.
Collapse
Affiliation(s)
- Lijuan Xie
- Department of Gastroenterology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Zhang
- Department of Gastroenterology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yonglai Lv
- Department of Gastroenterology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
8
|
Rasheed S, Jha M, Waheed A, Farooq U, Fatima A, Haq IU, Khan U, Wardak AB, Gul MH. The potential of CRISPR-Cas9 in cardiovascular medicine: a focus on hereditary cardiomyopathies. Ann Med Surg (Lond) 2025; 87:1801-1803. [PMID: 40212198 PMCID: PMC11981303 DOI: 10.1097/ms9.0000000000003170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/02/2025] [Indexed: 04/13/2025] Open
Affiliation(s)
| | - Mayank Jha
- Government Medical College and New Civil Hospital, Surat, India
| | - Aiman Waheed
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Umer Farooq
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | | | | | - Usman Khan
- Hayatabad Medical Complex, Peshawar, Pakistan
| | | | | |
Collapse
|
9
|
Lee DSM, Cardone KM, Zhang DY, Tsao NL, Abramowitz S, Sharma P, DePaolo JS, Conery M, Aragam KG, Biddinger K, Dilitikas O, Hoffman-Andrews L, Judy RL, Khan A, Kullo IJ, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany Z, Cappola TP, Carruth ED, Day SM, Do R, Haggerty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common-variant and rare-variant genetic architecture of heart failure across the allele-frequency spectrum. Nat Genet 2025; 57:829-838. [PMID: 40195560 PMCID: PMC12049093 DOI: 10.1038/s41588-025-02140-2] [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: 04/25/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025]
Abstract
Heart failure is a complex trait, influenced by environmental and genetic factors, affecting over 30 million individuals worldwide. Here we report common-variant and rare-variant association studies of all-cause heart failure and examine how different classes of genetic variation impact its heritability. We identify 176 common-variant risk loci at genome-wide significance in 2,358,556 individuals and cluster these signals into five broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity and arrhythmias. In parallel, we uncover exome-wide significant associations for heart failure and rare predicted loss-of-function variants in TTN, MYBPC3, FLNC and BAG3 using exome sequencing of 376,334 individuals. We find that total burden heritability of rare coding variants is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common-variant heritability is diffusely spread throughout the genome. Finally, we show that common-variant background modifies heart failure risk among carriers of rare pathogenic truncating variants in TTN. Together, these findings discern genetic links between dysregulated metabolism and heart failure and highlight a polygenic component to heart failure not captured by current clinical genetic testing.
Collapse
Affiliation(s)
- David S M Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zoltan Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eric D Carruth
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- BioMe Phenomics Center, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | | | - Jacob Joseph
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
10
|
Cavazza A, Molina-Estévez FJ, Reyes ÁP, Ronco V, Naseem A, Malenšek Š, Pečan P, Santini A, Heredia P, Aguilar-González A, Boulaiz H, Ni Q, Cortijo-Gutierrez M, Pavlovic K, Herrera I, de la Cerda B, Garcia-Tenorio EM, Richard E, Granados-Principal S, López-Márquez A, Köber M, Stojanovic M, Vidaković M, Santos-Garcia I, Blázquez L, Haughton E, Yan D, Sánchez-Martín RM, Mazini L, Aseguinolaza GG, Miccio A, Rio P, Desviat LR, Gonçalves MA, Peng L, Jiménez-Mallebrera C, Molina FM, Gupta D, Lainšček D, Luo Y, Benabdellah K. Advanced delivery systems for gene editing: A comprehensive review from the GenE-HumDi COST Action Working Group. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102457. [PMID: 39991472 PMCID: PMC11847086 DOI: 10.1016/j.omtn.2025.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
In the past decade, precise targeting through genome editing has emerged as a promising alternative to traditional therapeutic approaches. Genome editing can be performed using various platforms, where programmable DNA nucleases create permanent genetic changes at specific genomic locations due to their ability to recognize precise DNA sequences. Clinical application of this technology requires the delivery of the editing reagents to transplantable cells ex vivo or to tissues and organs for in vivo approaches, often representing a barrier to achieving the desired editing efficiency and safety. In this review, authored by members of the GenE-HumDi European Cooperation in Science and Technology (COST) Action, we described the plethora of delivery systems available for genome-editing components, including viral and non-viral systems, highlighting their advantages, limitations, and potential application in a clinical setting.
Collapse
Affiliation(s)
- Alessia Cavazza
- Molecular and Cellular Immunology Section, Department of Infection, Immunity & Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, 20 Guilford Street, London WC1N 1DZ, UK
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via del Pozzo 71, 41125 Modena, Italy
| | - Francisco J. Molina-Estévez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Fundación para la Investigación Biosanitaria de Andalucía Oriental, Alejandro Otero (FIBAO), Avda. de Madrid 15, 18012 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
| | - Álvaro Plaza Reyes
- Department of Regeneration and Cell Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Avda. Americo Vespucio, 24, 41092 Seville, Spain
| | - Victor Ronco
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Asma Naseem
- Molecular and Cellular Immunology Section, Department of Infection, Immunity & Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, 20 Guilford Street, London WC1N 1DZ, UK
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Kongresni trg, 1000 Ljubljana, Slovenia
| | - Peter Pečan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Kongresni trg, 1000 Ljubljana, Slovenia
| | - Annalisa Santini
- Imagine Institute, UMR 163 INSERM, 24 Bd du Montparnasse, 75015 Paris, France
- Paris City University, 45 Rue des Saints-Pères, 75006 Paris, France
| | - Paula Heredia
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Department of Anatomy and Human Embryology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Araceli Aguilar-González
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry applied to Bio-medicine and the Environment, ” Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Houria Boulaiz
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Anatomy and Human Embryology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Marina Cortijo-Gutierrez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Kristina Pavlovic
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Inmaculada Herrera
- Department of Hematology, Reina Sofía University Hospital, Av. Menéndez Pidal, Poniente Sur, 14004 Córdoba, Spain
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cell Therapy, Av. Menéndez Pidal, Poniente Sur, 14004 Córdoba, Spain
| | - Berta de la Cerda
- Department of Regeneration and Cell Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Avda. Americo Vespucio, 24, 41092 Seville, Spain
| | - Emilio M. Garcia-Tenorio
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Sergio Granados-Principal
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Arístides López-Márquez
- Neuromuscular Unit, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, C. de Sta. Rosa, 39, 08950 Barcelona, Spain
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Gran Via de les Corts Catalanes, 585, L'Eixample, 08007 Barcelona, Spain
| | - Mariana Köber
- Biomedical Research Network on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Marijana Stojanovic
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar despota Stefana 142, 10060 Belgrade, Serbia
| | - Melita Vidaković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar despota Stefana 142, 10060 Belgrade, Serbia
| | - Irene Santos-Garcia
- Department of Neurosciences, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain, s/n, 20014 San Sebastián, Gipuzkoa, Spain
| | - Lorea Blázquez
- Department of Neurosciences, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain, s/n, 20014 San Sebastián, Gipuzkoa, Spain
- CIBERNED, ISCIII CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Av. de Monforte de Lemos, 5, Fuencarral-El Pardo, 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Euskadi Pl., 5, Abando, 48009 Bilbao, Biscay, Spain
| | - Emily Haughton
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Dongnan Yan
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Rosario María Sánchez-Martín
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry applied to Bio-medicine and the Environment, ” Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Loubna Mazini
- Technological, Medical and Academic Park (TMAP), N°109, Abdelkrim Elkhatabi, Bd Abdelkrim Al Khattabi, Marrakech 40000, Morocco
| | - Gloria Gonzalez Aseguinolaza
- DNA & RNA Medicine Division, Gene Therapy for Rare Diseases Department, Center for Applied Medical Research (CIMA), University of Navarra, IdisNA, Av. de Pío XII, 55, 31008 Pamplona, Navarra, Spain
- Vivet Therapeutics, Av. de Pío XII 31, 31008 Pamplona, Navarra, Spain
| | - Annarita Miccio
- Imagine Institute, UMR 163 INSERM, 24 Bd du Montparnasse, 75015 Paris, France
- Paris City University, 45 Rue des Saints-Pères, 75006 Paris, France
| | - Paula Rio
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
- Division of Hematopoietic Innovative Therapies, CIEMAT, Av. Complutense, 40, Moncloa - Aravaca, 28040 Madrid, Spain
- Advanced Therapies Unit, IIS-Fundación Jimenez Diaz (IIS-FJD, UAM), Av. de los Reyes Católicos, 2, Moncloa - Aravaca, 28040 Madrid, Spain
| | - Lourdes R. Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Manuel A.F.V. Gonçalves
- Leiden University Medical Center, Department of Cell and Chemical Biology, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Ling Peng
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisee Ligue Ćontre le Cancer”, Campus de Luminy, case 913, 13009 Marseille, France
| | - Cecilia Jiménez-Mallebrera
- Neuromuscular Unit, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, C. de Sta. Rosa, 39, 08950 Barcelona, Spain
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Francisco Martin Molina
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Dhanu Gupta
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- Department of Laboratory Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Huddinge, Sweden
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Centre for Technologies of Gene and Cell Therapy, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, 1000 Ljubljana, Slovenia
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Karim Benabdellah
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| |
Collapse
|
11
|
Sorella A, Galanti K, Iezzi L, Gallina S, Mohammed SF, Sekhri N, Akhtar MM, Prasad SK, Chahal CAA, Ricci F, Khanji MY. Diagnosis and management of dilated cardiomyopathy: a systematic review of clinical practice guidelines and recommendations. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2025; 11:206-222. [PMID: 39674807 PMCID: PMC11879293 DOI: 10.1093/ehjqcco/qcae109] [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: 11/17/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024]
Abstract
Dilated cardiomyopathy (DCM) is extensively discussed in numerous expert consensus documents and international guidelines, with differing recommendations. To support clinicians in daily practice and decision-making, we conducted a systematic review of key guidelines and recommendations concerning the diagnosis and clinical management of DCM. Our research encompassed MEDLINE and EMBASE databases for relevant articles published, as well as the websites of relevant scientific societies. We identified two guidelines and one scientific statement that met stringent criteria, thereby qualifying them for detailed systematic analysis. Our review revealed consensus on several key aspects: the definition of DCM, the use of B-type natriuretic peptides and high-sensitivity troponin in laboratory testing, the essential role of multimodality cardiovascular imaging for initial diagnosis, genetic counselling, and the management of advanced disease. Nonetheless, notable areas of variation included the formation of multidisciplinary management teams, the role of cascade genetic testing, pathways for arrhythmic risk stratification, and the criteria for prophylactic defibrillator implantation. Significant evidence gaps persist, particularly regarding the clinical trajectory of genetic, non-genetic and gene-elusive forms of DCM, the use of cardiovascular magnetic resonance in phenotype-negative family members with genotype-positive probands, and the development of potential aetiology-oriented therapies. Addressing these gaps could enhance clinical outcomes and inform future research directions and guideline development.
Collapse
Affiliation(s)
- Anna Sorella
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Kristian Galanti
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Lorena Iezzi
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, SS. Annunziata Polyclinic, Chieti 66100, Italy
- Institute for Advanced Biomedical Technologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Selma F Mohammed
- Department of Cardiology, Creighton University School of Medicine, Omaha, NE 68124, USA
| | - Neha Sekhri
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- Newham University Hospital, Barts Health NHS Trust, London EC1M 6BQ, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Mohammed Majid Akhtar
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Sanjay K Prasad
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Choudhary Anwar Ahmed Chahal
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Center for Inherited Cardiovascular Diseases, Department of Cardiology, WellSpan Health, 30 Monument Rd, York, PA 17403, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Str, SW Rochester, MN 55905, USA
| | - Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, SS. Annunziata Polyclinic, Chieti 66100, Italy
- Institute for Advanced Biomedical Technologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Mohammed Yunus Khanji
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- Newham University Hospital, Barts Health NHS Trust, London EC1M 6BQ, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
12
|
Zheng SL, Jurgens SJ, McGurk KA, Xu X, Grace C, Theotokis PI, Buchan RJ, Francis C, de Marvao A, Curran L, Bai W, Pua CJ, Tang HC, Jorda P, van Slegtenhorst MA, Verhagen JMA, Harper AR, Ormondroyd E, Chin CWL, Pantazis A, Baksi J, Halliday BP, Matthews P, Pinto YM, Walsh R, Amin AS, Wilde AAM, Cook SA, Prasad SK, Barton PJR, O'Regan DP, Lumbers RT, Goel A, Tadros R, Michels M, Watkins H, Bezzina CR, Ware JS. Evaluation of polygenic scores for hypertrophic cardiomyopathy in the general population and across clinical settings. Nat Genet 2025; 57:563-571. [PMID: 39966645 PMCID: PMC11906360 DOI: 10.1038/s41588-025-02094-5] [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: 03/30/2023] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality, with pathogenic variants found in about a third of cases. Large-scale genome-wide association studies (GWAS) demonstrate that common genetic variation contributes to HCM risk. Here we derive polygenic scores (PGS) from HCM GWAS and genetically correlated traits and test their performance in the UK Biobank, 100,000 Genomes Project, and clinical cohorts. We show that higher PGS significantly increases the risk of HCM in the general population, particularly among pathogenic variant carriers, where HCM penetrance differs 10-fold between those in the highest and lowest PGS quintiles. Among relatives of HCM probands, PGS stratifies risks of developing HCM and adverse outcomes. Finally, among HCM cases, PGS strongly predicts the risk of adverse outcomes and death. These findings support the broad utility of PGS across clinical settings, enabling tailored screening and surveillance and stratification of risk of adverse outcomes.
Collapse
Affiliation(s)
- Sean L Zheng
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kathryn A McGurk
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Xiao Xu
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Chris Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Pantazis I Theotokis
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Rachel J Buchan
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Catherine Francis
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Antonio de Marvao
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Women and Children's Health, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Lara Curran
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Paloma Jorda
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Judith M A Verhagen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elizabeth Ormondroyd
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Calvin W L Chin
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Antonis Pantazis
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - John Baksi
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Brian P Halliday
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul Matthews
- Department of Brain Sciences, Imperial College London, London, UK
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Stuart A Cook
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Cardiology, National Heart Centre, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Sanjay K Prasad
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul J R Barton
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Declan P O'Regan
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - R T Lumbers
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Rafik Tadros
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michelle Michels
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - James S Ware
- National Heart Lung Institute, Imperial College London, London, UK.
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK.
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Imperial College Healthcare NHS Trust, London, UK.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Li XL, Aizezi A, Li YP, Li YH, Liu F, Zhao Q, Ma X, Adi D, Ma YT. Dilated cardiomyopathy signature metabolic marker screening: Machine learning and multi-omics analysis. Heliyon 2025; 11:e41927. [PMID: 39991223 PMCID: PMC11847283 DOI: 10.1016/j.heliyon.2025.e41927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 02/25/2025] Open
Abstract
Objects Our aim was to identify changes in the metabolome in dilated cardiomyopathy (DCM) as well as to construct a metabolic diagnostic model for DCM. Methods We utilized non-targeted metabolomics with a cross-sectional cohort of age- and sex-matched DCM patients and controls. Metabolomics data were analyzed using orthogonal partial least squares-discriminant analysis (OPLS-DA) and pathway analysis. It was validated in combination with transcriptome sequencing data from public databases. Machine learning models were used for the diagnosis of DCM. Results Using multiple analytical techniques, 130 metabolite alterations were identified in DCM compared to healthy controls. Perturbations in glycerophospholipid metabolism (GPL) were identified and validated as a characteristic metabolic pathway in DCM. Through the least absolute shrinkage and selection operator (LASSO), we identified the 7 most important GPL metabolites, including LysoPA (16:0/0:0), LysoPA (18:1(9Z)/0:0), PC (20:3(8Z,11Z,14Z)/20:1(11Z)), PC (20:0/14:0), LysoPC (16:0), PS(15:0/18:0), and PE(16:0/20:4 (5Z,8Z,11Z,14Z)). The machine learning models based on the seven metabolites all had good accuracy in distinguishing DCM [All area under the curve (AUC) > 0.900], and the artificial neural network (ANN) model performed the most consistently (AUC: 0.919 ± 0.075). Conclusions This study demonstrates that GPL metabolism may play a contributing role in the pathophysiological mechanisms of DCM. The 7-GPL metabolite model may help for early diagnosis of DCM.
Collapse
Affiliation(s)
- Xiao-Lei Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
- Hubei University of Medicine, Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Shiyan, Hubei, 442008, China
| | - Aibibanmu Aizezi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yan-Peng Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yan-Hong Li
- Laboratory and Equipment Management, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Qian Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Xiang Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Dilare Adi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yi-Tong Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology,First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| |
Collapse
|
14
|
Fadoni J, Santos A, Amorim A, Cainé L. Sudden Cardiac Death: The Role of Molecular Autopsy with Next-Generation Sequencing. Diagnostics (Basel) 2025; 15:460. [PMID: 40002611 PMCID: PMC11854515 DOI: 10.3390/diagnostics15040460] [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: 01/14/2025] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Molecular autopsy is a term employed to describe the investigation of the cause of death through the analysis of genetic information using biological samples collected post-mortem. Its utility becomes evident in situations where conventional medico-legal autopsy methods are not able to identify the cause of death, i.e., in sudden cardiac death (SCD) cases in young individuals, where deaths are commonly due to genetic cardiac conditions, such as cardiomyopathies and channelopathies. The recent advancement in high-throughput sequencing techniques, such as next-generation sequencing (NGS), has allowed the investigation of a high number of genomic regions in a more cost-effective and faster approach. Unlike traditional sequencing methods, which can only sequence one DNA fragment at a time, NGS can sequence millions of short polynucleotide fragments simultaneously. This parallel approach reduces both the time and cost required to generate large-scale genomic data, making it a useful tool for applications ranging from basic research to molecular autopsy. In the forensic context, by enabling the examination of multiple genes or entire exomes and genomes, NGS enhances the accuracy and depth of genetic investigations, contributing to a better understanding of complex inherited diseases. However, challenges remain, such as the interpretation of variants of unknown significance (VUS), the need for standardized protocols, and the high demand for specialized bioinformatics expertise. Despite these challenges, NGS continues to offer significant promise for enhancing the precision of molecular autopsies. The goal of this review is to assess the effectiveness of contemporary advancements in molecular autopsy methodologies when applied to cases of SCD in young individuals and to present an overview of the steps involved in the analysis of NGS data and the interpretation of genetic variants.
Collapse
Affiliation(s)
- Jennifer Fadoni
- National Institute of Legal Medicine and Forensic Sciences, North Branch, 4050-167 Porto, Portugal; (J.F.)
- LAQV&REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Agostinho Santos
- National Institute of Legal Medicine and Forensic Sciences, North Branch, 4050-167 Porto, Portugal; (J.F.)
- LAQV&REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - António Amorim
- National Institute of Legal Medicine and Forensic Sciences, North Branch, 4050-167 Porto, Portugal; (J.F.)
- LAQV&REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Laura Cainé
- LAQV&REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- National Institute of Legal Medicine and Forensic Sciences, Centre Branch, 3000-548 Coimbra, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
| |
Collapse
|
15
|
Shaikh T, Nguyen D, Dugal JK, DiCaro MV, Yee B, Houshmand N, Lei K, Namazi A. Arrhythmogenic Right Ventricular Cardiomyopathy: A Comprehensive Review. J Cardiovasc Dev Dis 2025; 12:71. [PMID: 39997505 PMCID: PMC11855979 DOI: 10.3390/jcdd12020071] [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: 11/23/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized by structural abnormalities, arrhythmias, and a spectrum of genetic and clinical manifestations. Clinically, ARVC is structurally distinguished by right ventricular dilation due to increased adiposity and fibrosis in the ventricular walls, and it manifests as cardiac arrhythmias ranging from non-sustained ventricular tachycardia to sudden cardiac death. Its prevalence has been estimated to range from 1 in every 1000 to 5000 people, with its large range being attributed to the variability in genetic penetrance from asymptomatic to significant burden. It is even suggested that the prevalence is underestimated, as the presence of genotypic mutations does not always lead to clinical manifestations that would facilitate diagnosis. Additionally, while set criteria have been in place since the 1990s, newer understanding of this condition and advancements in cardiac technology have prompted multiple revisions in the diagnostic criteria for ARVC. Novel discoveries of gene variants predisposing patients to ARVC have led to established screening techniques while providing insight into genetic counseling and management. This review aims to provide an overview of the genetics, pathophysiology, and clinical approach to ARVC. It will also focus on clinical presentation, ARVC diagnostic criteria, electrophysiological findings, including electrocardiogram characteristics, and imaging findings from cardiac MRI, 2D, and 3D echocardiogram. Current management options-including anti-arrhythmic medications, device indications, and ablation techniques-and the effectiveness of treatment will also be reviewed.
Collapse
Affiliation(s)
- Taha Shaikh
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Darren Nguyen
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Jasmine K. Dugal
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Michael V. DiCaro
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Brianna Yee
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Nazanin Houshmand
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| | - KaChon Lei
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| | - Ali Namazi
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| |
Collapse
|
16
|
Tamizuddin F, Stojanovska J, Toussie D, Shmukler A, Axel L, Srinivasan R, Fujikura K, Broncano J, Frank L, Villasana-Gomez G. Advanced Computed Tomography and Magnetic Resonance Imaging in Ischemic and Nonischemic Cardiomyopathies. Echocardiography 2025; 42:e70106. [PMID: 39950567 DOI: 10.1111/echo.70106] [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: 11/29/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 05/09/2025] Open
Abstract
Cardiomyopathies represent a diverse group of heart diseases that can be broadly classified into ischemic and nonischemic etiologies, each requiring distinct diagnostic approaches. Noninvasive imaging modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI), play a pivotal role in the diagnosis, risk stratification, and prognosis of these conditions. This paper reviews the characteristic CT and MRI findings associated with ischemic cardiomyopathy (ICM) and nonischemic cardiomyopathy (NICM), focusing on their ability to provide detailed anatomical, functional, and tissue characterization. In ICM, CT and MRI reveal myocardial scarring, infarct size, and coronary artery disease, while MRI further distinguishes tissue viability through late gadolinium enhancement (LGE). Conversely, nonischemic cardiomyopathies demonstrate a wide array of findings, with MRI's LGE pattern analysis being particularly critical for identifying specific subtypes, such as restrictive, hypertrophic, or dilated cardiomyopathies. By comparing the strengths and limitations of these modalities, this paper highlights their complementary roles in improving diagnostic accuracy, risk stratification, prognosis, and therapeutic decision making in both ischemic and nonischemic cardiomyopathies.
Collapse
Affiliation(s)
- Farah Tamizuddin
- Department of Radiology, New York Langone Health, New York, New York, USA
| | | | - Danielle Toussie
- Department of Radiology, New York Langone Health, New York, New York, USA
| | - Anna Shmukler
- Department of Radiology, New York Langone Health, New York, New York, USA
| | - Leon Axel
- Department of Radiology, New York Langone Health, New York, New York, USA
| | - Ranjini Srinivasan
- Department of Cardiology, New York Langone Health, New York, New York, USA
| | - Kana Fujikura
- Department of Cardiology, New York Langone Health, New York, New York, USA
| | - Jordi Broncano
- Cardiothoracic Imaging Unit, Hospital San Juan de Dios, HT Medica, Córdoba, Spain
| | - Luba Frank
- Baylor College of Medicine, Baylor St. Luke's Medical Center and Texas Heart Institute Department of Radiology Cardiovascular Imaging Section Houston, Texas, USA
| | | |
Collapse
|
17
|
Wenderholm K, Brunet T, Graf E, Arens M, Martens E, Winkelmann J, Hoefele J, Westphal DS. Variants that get straight to your heart - Cardiogenetic secondary findings in exome sequencing. Gene 2025; 935:149063. [PMID: 39486665 DOI: 10.1016/j.gene.2024.149063] [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: 07/26/2024] [Revised: 08/23/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Exome sequencing has been established as a fundamental tool in genetic diagnostics. It may also provide information about variants in genes unrelated to the primary purpose, so-called secondary findings. Especially, diagnoses of unnoticed inborn cardiac diseases are of high clinical relevance due to therapeutic options in context of prevention of sudden cardiac death. METHODS Exome data of 9962 individuals was analysed for relevant cardiogenetic findings. Genes were selected according to ACMG recommendations for secondary findings (v.3.1). First, a filter for (likely) pathogenic variants, published in the ClinVar database, was used. Second, exome data was screened for loss of function (LoF) variants in genes in which LoF is a known disease pathomechanism. All variants were evaluated by geneticists regarding their pathogenicity. RESULTS Pathogenic or likely pathogenic variants were identified in 136 different individuals (136/9962, 1.4%), with the Low-Density Lipoprotein Receptor gene (LDLR, 24/136, 17.6%) and the Titin gene (TTN, 24/136, 17.6%), being the most frequently affected ones. 31.6% (43/136) of the identified variants had been reported beforehand, while 47.1% (64/136) had not been reported. The remaining cases (29/136, 21.3%) were part of research projects with no written reports. In 26.5% (36/136), the finding would have been missed, if only index patients and not their parents had been screened for secondary findings in case of trio ES. CONCLUSION As demonstrated in our study, at least one or two out of one hundred people are likely to carry a pathogenic cardiogenetic variant. Counselling geneticist and clinicians need to be aware of these findings in exome and genome sequencing. Informed consent of the patient regarding the report of secondary findings should absolutely be obtained beforehand.
Collapse
Affiliation(s)
- Kirsten Wenderholm
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Marie Arens
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Eimo Martens
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Juliane Winkelmann
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany
| | - Dominik S Westphal
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany; Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Germany.
| |
Collapse
|
18
|
Yates T, Razieh C, Henson J, Rowlands AV, Goldney J, Gulsin GS, Davies MJ, Khunti K, Zaccardi F, McCann GP. Device-measured physical activity and cardiac structure by magnetic resonance. Eur Heart J 2025; 46:176-186. [PMID: 39140328 PMCID: PMC11704417 DOI: 10.1093/eurheartj/ehae506] [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: 12/15/2023] [Revised: 04/18/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND AND AIMS Although extreme cardiac adaptions mirroring phenotypes of cardiomyopathy have been observed in endurance athletes, adaptions to high levels of physical activity within the wider population are under-explored. Therefore, in this study, associations between device-measured physical activity and clinically relevant cardiac magnetic resonance volumetric indices were investigated. METHODS Individuals without known cardiovascular disease or hypertension were included from the UK Biobank. Cardiac magnetic resonance data were collected between 2015 and 2019, and measures of end-diastolic chamber volume, left ventricular (LV) wall thickness, and LV ejection fraction were extracted. Moderate-to-vigorous-intensity physical activity (MVPA), vigorous-intensity physical activity (VPA), and total physical activity were assessed via wrist-worn accelerometers. RESULTS A total of 5977 women (median age and MVPA: 62 years and 46.8 min/day, respectively) and 4134 men (64 years and 49.8 min/day, respectively) were included. Each additional 10 min/day of MVPA was associated with a 0.70 [95% confidence interval (CI): 0.62, 0.79] mL/m2 higher indexed LV end-diastolic volume (LVEDVi) in women and a 1.08 (95% CI: 0.95, 1.20) mL/m2 higher LVEDVi in men. However, even within the top decile of MVPA, LVEDVi values remained within the normal ranges [79.1 (95% CI: 78.3, 80.0) mL/m2 in women and 91.4 (95% CI: 90.1, 92.7) mL/m2 in men]. Associations with MVPA were also observed for the right ventricle and the left/right atria, with an inverse association observed for LV ejection fraction. Associations of MVPA with maximum or average LV wall thickness were not clinically meaningful. Results for total physical activity and VPA mirrored those for MVPA. CONCLUSIONS High levels of device-measured physical activity were associated with cardiac remodelling within normal ranges.
Collapse
Affiliation(s)
- Thomas Yates
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Cameron Razieh
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Joe Henson
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Alex V Rowlands
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Jonathan Goldney
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Melanie J Davies
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
- Leicester Real World Evidence Unit, University of Leicester, Leicester, UK
| | - Francesco Zaccardi
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
- Leicester Real World Evidence Unit, University of Leicester, Leicester, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
19
|
Olivotto I. Family matters: health policies to tackle cardiomyopathies across Europe. Eur Heart J 2025; 46:6-14. [PMID: 39548843 PMCID: PMC11695914 DOI: 10.1093/eurheartj/ehae419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2024] Open
Affiliation(s)
- Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence
and Meyer University Children Hospital, Viale Pieraccini 24,
50134 Florence, Italy
| |
Collapse
|
20
|
Ewoldt JK, DePalma SJ, Jewett ME, Karakan MÇ, Lin YM, Mir Hashemian P, Gao X, Lou L, McLellan MA, Tabares J, Ma M, Salazar Coariti AC, He J, Toussaint KC, Bifano TG, Ramaswamy S, White AE, Agarwal A, Lejeune E, Baker BM, Chen CS. Induced pluripotent stem cell-derived cardiomyocyte in vitro models: benchmarking progress and ongoing challenges. Nat Methods 2025; 22:24-40. [PMID: 39516564 DOI: 10.1038/s41592-024-02480-7] [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: 12/11/2023] [Accepted: 09/15/2024] [Indexed: 11/16/2024]
Abstract
Recent innovations in differentiating cardiomyocytes from human induced pluripotent stem cells (hiPSCs) have unlocked a viable path to creating in vitro cardiac models. Currently, hiPSC-derived cardiomyocytes (hiPSC-CMs) remain immature, leading many in the field to explore approaches to enhance cell and tissue maturation. Here, we systematically analyzed 300 studies using hiPSC-CM models to determine common fabrication, maturation and assessment techniques used to evaluate cardiomyocyte functionality and maturity and compiled the data into an open-access database. Based on this analysis, we present the diversity of, and current trends in, in vitro models and highlight the most common and promising practices for functional assessments. We further analyzed outputs spanning structural maturity, contractile function, electrophysiology and gene expression and note field-wide improvements over time. Finally, we discuss opportunities to collectively pursue the shared goal of hiPSC-CM model development, maturation and assessment that we believe are critical for engineering mature cardiac tissue.
Collapse
Affiliation(s)
- Jourdan K Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Maggie E Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - M Çağatay Karakan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Yih-Mei Lin
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Paria Mir Hashemian
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Xining Gao
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Lihua Lou
- Department of Mechanical and Material Engineering, Florida International University, Miami, FL, USA
| | - Micheal A McLellan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jonathan Tabares
- Department of Physics, Florida International University, Miami, FL, USA
| | - Marshall Ma
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | | | - Jin He
- Department of Physics, Florida International University, Miami, FL, USA
| | - Kimani C Toussaint
- School of Engineering, Brown University, Providence, RI, USA
- Brown-Lifespan Center for Digital Health, Providence, RI, USA
| | - Thomas G Bifano
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Alice E White
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
- Division of Materials Science and Engineering, Boston University, Boston, MA, USA
- Department of Physics, Boston University, Boston, MA, USA
| | - Arvind Agarwal
- Department of Mechanical and Material Engineering, Florida International University, Miami, FL, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
21
|
Xu M, Liu X, Lu L, Li Z. Metrnl and Cardiomyopathies: From Molecular Mechanisms to Therapeutic Insights. J Cell Mol Med 2025; 29:e70371. [PMID: 39853716 PMCID: PMC11756984 DOI: 10.1111/jcmm.70371] [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: 09/02/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Cardiomyopathies, a diverse group of diseases affecting the heart muscle, continue to pose significant clinical challenges due to their complex aetiologies and limited treatment options targeting underlying genetic and molecular dysregulations. Emerging evidence indicates that Metrnl, a myokine, adipokine and cardiokine, plays a significant role in the pathogenesis of various cardiomyopathies. Therefore, the objective of this review is to examine the role and mechanism of Metrnl in various cardiomyopathies, with the expectation of providing new insights for the treatment of these diseases.
Collapse
Affiliation(s)
- Miaomiao Xu
- School of Physical Education and HealthGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu‐Moxi and RehabilitationGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xiaoguang Liu
- College of Sports and HealthGuangzhou Sport UniversityGuangzhouGuangdongChina
| | - Liming Lu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu‐Moxi and RehabilitationGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhaowei Li
- School of Physical Education and HealthGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| |
Collapse
|
22
|
Roudi HS, Safaei R, Dabbaghi MM, Fadaei MS, Saberifar M, Sakhaee K, Rahimi VB, Askari VR. Mechanistic Insights on Cardioprotective Properties of Ursolic Acid: Regulation of Mitochondrial and Non-mitochondrial Pathways. Curr Pharm Des 2025; 31:1037-1056. [PMID: 39710917 DOI: 10.2174/0113816128344497241120025757] [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: 07/15/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 12/24/2024]
Abstract
Ursolic acid, a natural pentacyclic triterpenoid compound, has been shown to have significant cardioprotective effects in various preclinical studies. This article reviews the various mechanisms by which ursolic acid achieves its cardioprotective effects, highlighting its potent anti-oxidant, anti-inflammatory, and anti- apoptotic properties. Ursolic acid upregulates anti-oxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), effectively reducing oxidative stress, thereby decreasing reactive oxygen species (ROS) and improving lipid peroxidation levels. Furthermore, ursolic acid downregulates pro-inflammatory cytokines and inhibits key inflammatory pathways, such as nuclear factor kappa B (NF-κB), which results in its anti-inflammatory effects. These actions help in protecting cardiac tissues from acute and chronic inflammation. Ursolic acid also promotes mitochondrial function and energy metabolism by enhancing mitochondrial biogenesis and reducing dysfunction, which is critical during ischemia-reperfusion (I/R) injury. Additionally, ursolic acid influences multiple molecular pathways, including B-cell leukemia/lymphoma 2 protein (Bcl- 2)/Bcl-2 associated x-protein (Bax), miR-21/extracellular signal-regulated kinase (ERK), and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), to reduce cardiomyocyte apoptosis. Collectively, these properties make ursolic acid a promising therapeutic agent for cardiovascular diseases (CVDs), warranting further research and clinical trials to harness its potential fully.
Collapse
Affiliation(s)
- Hesan Soleimani Roudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rozhan Safaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mahdi Dabbaghi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Saleh Fadaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Saberifar
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Katayoun Sakhaee
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Hespe S, Gray B, Puranik R, Peters S, Sweeting J, Ingles J. The role of genetic testing in management and prognosis of individuals with inherited cardiomyopathies. Trends Cardiovasc Med 2025; 35:34-44. [PMID: 39004295 DOI: 10.1016/j.tcm.2024.06.002] [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: 03/10/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Inherited cardiomyopathies are a heterogeneous group of heart muscle conditions where disease classification has traditionally been based on clinical characteristics. However, this does not always align with genotype. While there are well described challenges of genetic testing, understanding the role of genotype in patient management is increasingly required. We take a gene-by-gene approach, reviewing current evidence for the role of genetic testing in guiding prognosis and management of individuals with inherited cardiomyopathies. In particular, focusing on causal variants in genes definitively associated with arrhythmogenic cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy. This review identifies genotype-specific disease sub-groups with strong evidence supporting the use of genetics in clinical management and highlights that at present, the spectrum of clinical utility is not reflected in current guidelines. Of 13 guideline or expert consensus statements for management of cardiomyopathies, there are seven gene-specific therapeutic recommendations that have been published from four documents. Understanding how genotype influences phenotype provides evidence for the role of genetic testing for prognostic and therapeutic purposes, moving us closer to precision-medicine based care.
Collapse
Affiliation(s)
- Sophie Hespe
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia
| | - Belinda Gray
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rajesh Puranik
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Stacey Peters
- Department of Cardiology and Genomic Medicine, Royal Melbourne Hospital, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Joanna Sweeting
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
24
|
Patsalis C, Kyriakou S, Georgiadou M, Ioannou L, Constantinou L, Soteriou V, Jossif A, Evangelidou P, Sismani C, Kypri E, Ioannides M, Koumbaris G. Investigating TNNC1 gene inheritance and clinical outcomes through a comprehensive familial study. Am J Med Genet A 2025; 197:e63838. [PMID: 39248034 DOI: 10.1002/ajmg.a.63838] [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: 02/01/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) and restrictive cardiomyopathy (RCM) have significant phenotypic overlap and a similar genetic background, both caused mainly by variants in sarcomeric genes. HCM is the most common cardiomyopathy, while RCM is a rare and often underdiagnosed heart condition, with a poor prognosis. This study focuses on a large family with four infants diagnosed with fatal RCM associated with biventricular hypertrophy. Affected infants were found to be homozygous for NM_003280.3(TNNC1):c.23C>T(p.Ala8Val) variant. Interestingly, this variant resulted in a low penetrance and mild form of hypertrophic cardiomyopathy (HCM) in relatives carrying a single copy of the variant. Overall, this study underscores the complex nature of genetic inheritance in cardiomyopathies and the wide range of clinical presentations they can exhibit. This emphasizes the vital role of genetic testing in providing essential insights crucial for diagnosis, prognosis, early intervention, and the development of potential treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Antonis Jossif
- Paedi Center for Specialized Pediatrics, Nicosia, Cyprus
| | - Paola Evangelidou
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Carolina Sismani
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | | | | |
Collapse
|
25
|
Zou C, Zou H, Jiang Y, Lai S, Liu J. Association between cardiovascular risk factors and dilated and hypertrophic cardiomyopathy: Mendelian randomization analysis. Nutr Metab Cardiovasc Dis 2025; 35:103752. [PMID: 39455334 DOI: 10.1016/j.numecd.2024.09.022] [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: 03/11/2024] [Revised: 07/29/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND AND AIM Dilated cardiomyopathy is a major cause of heart failure, and hypertrophic cardiomyopathy is a common cause of sudden cardiac death in young adults. Epidemiological studies reporting the association between these cardiomyopathies and common cardiovascular risk factors, including smoking, alcohol, and obesity, are limited, and the published studies are mostly observational, making them vulnerable to bias. METHODS AND RESULTS We performed a two-sample Mendelian randomization analysis to assess whether cardiovascular risk factors were causally associated with dilated and hypertrophic cardiomyopathies. Independent genetic variants associated with body mass index, smoking, and alcohol were selected as instrumental variables, with two sets of instrumental variables utilized for alcohol. Dilated cardiomyopathy data on 355,318 samples and hypertrophic cardiomyopathy data on 489,727 samples were obtained from a European population-based genome-wide association study (GWAS) meta-analysis. The large GWAS data sample size improved the statistical power. Our results showed significant associations between a genetic predisposition for smoking and the risk of dilated cardiomyopathy (odds ratio (OR) = 1.33; 95 % confidence level (CI): 1.07-1.67; p = 0.012) and between a genetic predisposition for obesity and the risk of dilated cardiomyopathy (OR = 1.62; 95 % CI, 1.30-2.02; p = 1.51 × 10-5). The results of the other associations were not significant. CONCLUSIONS This study suggests that smoking and obesity are causally associated with an increased risk of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Chenchao Zou
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China; Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Huaxi Zou
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Ying Jiang
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Songqing Lai
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jichun Liu
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
26
|
Tang Q, Meng X, Tu X, Zhang J. Mendelian Randomization Study on the Associations Between Genetically Predicted Cardiovascular Disease Subtypes and the Risk of Developing Cardiomyopathies. Clin Appl Thromb Hemost 2025; 31:10760296251328011. [PMID: 40152048 PMCID: PMC11951890 DOI: 10.1177/10760296251328011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 02/15/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiomyopathies are commonly believed to have genetic origins; however, the connection between cardiomyopathies and cardiovascular diseases remains uncertain. Thus, we employed a Mendelian randomization (MR) approach to investigate the potential causal effects of specific cardiovascular disease subtypes on dilated and hypertrophic cardiomyopathies, focusing primarily on a European population. Summary-level data for cardiomyopathies and other cardiovascular diseases were obtained from public genome-wide association studies. Random-effects inverse-variance weighting was used as the primary analysis, whereas sensitivity analyses, including weighted median, MR-Egger, and multivariable MR methods, were also conducted. A genetic predisposition to atrial fibrillation [odds ratio (OR): 1.33; 95% confidence interval (CI): 1.18-1.50; P < 0.001], heart failure (OR: 3.22; 95% CI: 1.92-5.41; P < 0.001), and hypertension (OR: 1.50; 95% CI: 1.25-1.81; P < 0.001) were causally linked to an increased risk of developing dilated cardiomyopathy. However, there was no direct causal connection between genetically predicted coronary heart disease, pulmonary embolism, or ischemic stroke and the risk of developing dilated cardiomyopathy. In contrast, no significant associations were found between genetically predicted CVD subtypes and the risk of developing hypertrophic cardiomyopathy. Genetically predicted heart failure is significantly associated with the risk of developing dilated cardiomyopathy, underscoring the importance of effective heart failure management for risk prevention. Moreover, individuals with hypertension and atrial fibrillation might have an increased predisposition to dilated cardiomyopathy, highlighting crucial implications for management.
Collapse
Affiliation(s)
- Qiaolin Tang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiangzhu Meng
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiaowen Tu
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Jian Zhang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| |
Collapse
|
27
|
Fan H, Tan X, Xu S, Zeng Y, Zhang H, Shao T, Zhao R, Zhou P, Bo X, Fan J, Fu Y, Ding X, Zhou Y. Identification and validation of differentially expressed disulfidptosis-related genes in hypertrophic cardiomyopathy. Mol Med 2024; 30:249. [PMID: 39701955 DOI: 10.1186/s10020-024-01024-1] [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: 07/22/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common cardiovascular diseases with no effective treatment due to its complex pathogenesis. A novel cell death, disulfidptosis, has been extensively studied in the cancer field but rarely in cardiovascular diseases. This study revealed the potential relationship between disulfidptosis and hypertrophic cardiomyopathy and put forward a predictive model containing disulfidptosis-associated genes (DRGs) of GYS1, MYH10, PDMIL1, SLC3A2, CAPZB, showing excellent performance by SVM machine learning model. The results were further validated by western blot, RNA sequencing and immunohistochemistry in a TAC mice model. In addition, resveratrol was selected as a therapeutic drug targeting core genes using the CTD database. In summary, this study provides new perspectives for exploring disulfidptosis-related biomarkers and potential therapeutic targets for hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Huimin Fan
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Xin Tan
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Shuai Xu
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Yiyao Zeng
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Hailong Zhang
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Tong Shao
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Runze Zhao
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Peng Zhou
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Yangjun Fu
- Department of Neurology, The Third People's Hospital of Hefei, Hefei City, Anhui Province, 230041, China
| | - Xulong Ding
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China.
| | - Yafeng Zhou
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China.
- Institute for Hypertension, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
28
|
Hirono K, Hata Y, Ichimata S, Nishida N, Imamura T, Asano Y, Kuramoto Y, Tsuboi K, Takarada S, Okabe M, Nakaoka H, Ibuki K, Ozawa S, Muneuchi J, Yasuda K, Urayama K, Oka H, Miyamoto T, Baba K, Kato A, Saiki H, Kuwabara N, Harada M, Baba S, Morikawa M, Iwasaki H, Hirata Y, Ito Y, Sakaguchi H, Urata S, Toda K, Kittaka E, Okada S, Hasebe Y, Hoshino S, Fujii T, Mitsushita N, Nii M, Ogino K, Fujino M, Yoshida Y, Fukuda Y, Iwashima S, Takigiku K, Sakata Y, Inuzuka R, Maeda J, Hayabuchi Y, Fujioka T, Namiki H, Fujita S, Nishida K, Kuraoka A, Kan N, Kido S, Watanabe K, Ichida F. Sarcomere gene variants did not improve cardiac function in pediatric patients with dilated cardiomyopathy from Japanese cohorts. Sci Rep 2024; 14:30469. [PMID: 39681577 DOI: 10.1038/s41598-024-77360-3] [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: 03/21/2024] [Accepted: 10/22/2024] [Indexed: 12/18/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a progressive myocardial disorder characterized by impaired cardiac contraction and ventricular dilation. However, some patients with DCM improve when experiencing left ventricular reverse remodeling (LVRR). Currently, the detailed association between genotypes and clinical outcomes, including LVRR, particularly among children, remains uncertain. Pediatric patients with DCM from multiple Japanese institutions recorded between 2014 and 2023 were enrolled. We identified their DCM-related genes and explored the association between gene variants and clinical outcomes, including LVRR. We included 123 pediatric patients (62 males; median age: 8 [1-51] months) and found 50 pathogenic variants in 45 (35.0%) of them. The most identified gene was MYH7 (14.0%), followed by RYR2 (12.0%) and TPM1 (8.0%). LVRR was achieved in 47.5% of these patients. The left ventricular ejection fraction remained unchanged (31.4% to 39.8%, P = 0.1913) in patients with sarcomere gene variants and in those with non-sarcomere gene variants (33.4% to 47.8%, P = 0.0522) but significantly increased in those without gene variants (33.6% to 54.1%, P < 0.0001). LVRR was not uniform across functional gene groups. Hence, an individualized gene-guided prediction approach may be adopted for children with DCM.
Collapse
Affiliation(s)
- Keiichi Hirono
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Yukiko Hata
- Legal Medicine, University of Toyama, Toyama, Japan
| | | | | | - Teruhiko Imamura
- 2Nd Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | | | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kaori Tsuboi
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Shinya Takarada
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Mako Okabe
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hideyuki Nakaoka
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Keijiro Ibuki
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Sayaka Ozawa
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama City, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Jun Muneuchi
- Department of Pediatrics, Kyushu Hospital, Japan Community Healthcare Organization, Fukuoka, Japan
| | - Kazushi Yasuda
- Department of Pediatric Cardiology, Aichi Children's Health and Medical Center, Aichi, Japan
| | - Kotaro Urayama
- Department of Pediatrics, Tsuchiya General Hospital, Hiroshima, Japan
| | - Hideharu Oka
- Department of Pediatrics, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Tomoyuki Miyamoto
- Department of Pediatrics, Yokosuka General Hospital Uwamachi, Kanagawa, Japan
| | - Kenji Baba
- Department of Pediatrics, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Akio Kato
- Department of Pediatric Cardiology, Okinawa Prefectural Nanbu Medical Center and Children's Medical Center, Okinawa, Japan
| | - Hirofumi Saiki
- Department of Pediatrics and Pediatric Cardiology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Naoki Kuwabara
- Department of Pediatric Cardiology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Masako Harada
- Division of Pediatrics, Developmental and Urological-Reproductive Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shiro Baba
- Department of Pediatrics, Kyoto University Hospital, Kyoto, Japan
| | - Mari Morikawa
- Department of Pediatrics, Kanazawa Medical University, Ishikawa, Japan
| | - Hidenori Iwasaki
- Department of Pediatrics, Kanazawa University Hospital, Ishikawa, Japan
| | - Yuichiro Hirata
- Department of Pediatrics, Kyushu University Hospital, Fukuoka, Japan
| | - Yuki Ito
- Department of Pediatric Cardiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Heima Sakaguchi
- Department of Pediatric Cardiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Susumu Urata
- Division of Cardiology, National Center for Child Health and Development, Tokyo, Japan
| | - Koichi Toda
- Department of Pediatric Cardiology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Emi Kittaka
- Department of Cardiology, Saitama Children's Medical Center, Saitama, Japan
| | - Seigo Okada
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yohei Hasebe
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shinsuke Hoshino
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Takanari Fujii
- Pediatric Heart Disease and Adult Congenital Heart Disease Center, Showa University Hospital, Tokyo, Japan
| | - Norie Mitsushita
- Department of Cardiology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Masaki Nii
- Department of Cardiology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Kayo Ogino
- Department of Pediatrics, Kurashiki Central Hospital, Okayama, Japan
| | - Mitsuhiro Fujino
- Department of Pediatric Cardiology, Osaka City General Hospital, Osaka, Japan
| | - Yoko Yoshida
- Decision of Pediatric Electrophysiology, Osaka City General Hospital, Osaka, Japan
| | - Yutaka Fukuda
- Department of Pediatrics, Takeda General Hospital, Fukushinma, Japan
| | - Satoru Iwashima
- Department of Pediatrics, Chutoen General Medical Center, Shizuoka, Japan
| | - Kiyohiro Takigiku
- Department of Pediatric Cardiology, Nagano Children's Hospital, Nagano, Japan
| | - Yasushi Sakata
- Division of Pediatrics and Perinatology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Ryo Inuzuka
- Department of Pediatrics, Tokyo University Hospital, Tokyo, Japan
| | - Jun Maeda
- Division of Cardiology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Yasunobu Hayabuchi
- Department of Pediatrics, Tokushima University Hospital, Tokushima, Japan
| | - Tao Fujioka
- Department of Pediatrics, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Hidemasa Namiki
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Shuhei Fujita
- Department of Pediatrics, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Koichi Nishida
- Department of Pediatrics, Fukui Cardiovascular Center, Fukui, Japan
| | - Ayako Kuraoka
- Department of Cardiology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Nobuhiko Kan
- Department of Fetal and Neonatal Cardiology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Sachiko Kido
- Department of Cardiology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Ken Watanabe
- Department of Pediatrics, Kitano Hospital Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Fukiko Ichida
- Department of Pediatrics, International University of Health and Welfare, Tokyo, Japan
| |
Collapse
|
29
|
Figueiral M, Paldino A, Fazzini L, Pereira NL. Genetic Biomarkers in Heart Failure: From Gene Panels to Polygenic Risk Scores. Curr Heart Fail Rep 2024; 21:554-569. [PMID: 39405019 DOI: 10.1007/s11897-024-00687-5] [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] [Accepted: 09/06/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide a comprehensive overview of the current understanding of genetic markers associated with heart failure (HF) and its underlying causative diseases, such as cardiomyopathies. It highlights the relevance of genetic biomarkers in diagnosing HF, predicting prognosis, potentially identifying its preclinical stages and identifying targets to enable the implementation of individualized medicine approaches. RECENT FINDINGS The prevalence of HF is increasing due to an aging population but with greater access to disease-modifying therapies. Advanced diagnostic tools such as cardiac magnetic resonance, nuclear imaging, and AI-enabled diagnostic testing are now being utilized to further characterize HF patients. Additionally, the importance of genetic testing in HF diagnosis and management is increasingly being recognized. Genetic biomarkers, including single nucleotide polymorphisms (SNPs) and rare genetic variants, are emerging as crucial tools for diagnosing HF substrates, determining prognosis and increasingly directing therapy. These genetic insights are key to optimizing HF management and delivering personalized treatment tailored to individual patients. HF is a complex syndrome affecting millions globally, characterized by high mortality and significant economic burden. Understanding the underlying etiologies of HF is essential for improving management and clinical outcomes. Recent advances highlight the use of multimodal assessments, including AI-enabled diagnostics and genetic testing, to better characterize and manage HF. Genetic biomarkers are particularly promising in identifying preclinical HF stages and providing personalized treatment options. The genetic contribution to HF is heterogeneous, with both monogenic and polygenic bases playing a role. These developments underscore the shift towards personalized medicine in HF management.
Collapse
Affiliation(s)
- Marta Figueiral
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alessia Paldino
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Luca Fazzini
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
30
|
Huang HL, Suchenko A, Grandinetti G, Balasubramanian MK, Chinthalapudi K, Heissler SM. Cryo-EM structures of cardiac muscle α-actin mutants M305L and A331P give insights into the structural mechanisms of hypertrophic cardiomyopathy. Eur J Cell Biol 2024; 103:151460. [PMID: 39393252 PMCID: PMC11611453 DOI: 10.1016/j.ejcb.2024.151460] [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: 09/05/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/13/2024] Open
Abstract
Cardiac muscle α-actin is a key protein of the thin filament in the muscle sarcomere that, together with myosin thick filaments, produce force and contraction important for normal heart function. Missense mutations in cardiac muscle α-actin can cause hypertrophic cardiomyopathy, a complex disorder of the heart characterized by hypercontractility at the molecular scale that leads to diverse clinical phenotypes. While the clinical aspects of hypertrophic cardiomyopathy have been extensively studied, the molecular mechanisms of missense mutations in cardiac muscle α-actin that cause the disease remain largely elusive. Here we used cryo-electron microscopy to reveal the structures of hypertrophic cardiomyopathy-associated actin mutations M305L and A331P in the filamentous state. We show that the mutations have subtle impacts on the overall architecture of the actin filament with mutation-specific changes in the nucleotide binding cleft active site, interprotomer interfaces, and local changes around the mutation site. This suggests that structural changes induced by M305L and A331P have implications for the positioning of the thin filament protein tropomyosin and the interaction with myosin motors. Overall, this study supports a structural model whereby altered interactions between thick and thin filament proteins contribute to disease mechanisms in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andrejus Suchenko
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA; Center for Electron Microscopy and Analysis, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Mohan K Balasubramanian
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
31
|
师 轲, 喻 诗, 夏 冬, 郭 应, 杨 志. [Clincial Research Progress in Using Magnetic Resonance Imaging to Assess Myocardial Fibrosis in Hypertrophic Cardiomyopathy]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1357-1363. [PMID: 39990836 PMCID: PMC11839347 DOI: 10.12182/20241160601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Indexed: 02/25/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common type of primary cardiomyopathy that causes sudden cardiac death in adolescents and athletes. With over 1 million HCM patients, China has the largest population of HCM patients in the world, and the total number of cases is increasing year on year. Myocardial fibrosis is the most important histopathological characterization in HCM and is regarded as the primary cause of malignant ventricular arrhythmia, cardiac remodeling, and heart failure. At present, cardiac magnetic resonance imaging (MRI) serves as the gold-standard imaging modality for noninvasive evaluation of myocardial fibrosis. Several techniques, such as late gadolinium enhancement and T1 mapping, are showing considerable promise for potential applications. These techniques have emerged as viable imaging approaches to the elucidation of HCM tissue characterization. They are also helpful in predicting the long-term prognosis of patients. Herein, we summarized recent advances in using cardiac MRI to assess myocardial fibrosis in HCM from four perspectives, including late gadolinium enhancement, T1 mapping, T1ρ mapping, and MRI-based radiomics and machine learning models.
Collapse
Affiliation(s)
- 轲 师
- 四川大学华西医院 放射科 (成都 610041)Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 诗琴 喻
- 四川大学华西医院 放射科 (成都 610041)Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 冬 夏
- 四川大学华西医院 放射科 (成都 610041)Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
- 中国科学院大学经济与管理学院 (北京 100190)School of Economics and Management, University of Chinese Academy of Sciences, Beijing 100190, China
| | - 应坤 郭
- 四川大学华西医院 放射科 (成都 610041)Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 志刚 杨
- 四川大学华西医院 放射科 (成都 610041)Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
32
|
Draper I, Huang W, Pande S, Zou A, Calamaras TD, Choe RH, Correia-Branco A, Mei AL, Chen HH, Littel HR, Gunasekaran M, Wells NM, Bruels CC, Daugherty AL, Wolf MJ, Kang PB, Yang VK, Slonim DK, Wallingford MC, Blanton RM. The splicing factor hnRNPL demonstrates conserved myocardial regulation across species and is altered in heart failure. FEBS Lett 2024; 598:2670-2682. [PMID: 39300280 PMCID: PMC11560511 DOI: 10.1002/1873-3468.15020] [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: 03/13/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/22/2024]
Abstract
Heart failure (HF) is highly prevalent. Mechanisms underlying HF remain incompletely understood. Splicing factors (SF), which control pre-mRNA alternative splicing, regulate cardiac structure and function. This study investigated regulation of the splicing factor heterogeneous nuclear ribonucleoprotein-L (hnRNPL) in the failing heart. hnRNPL protein increased in left ventricular tissue from mice with transaortic constriction-induced HF and from HF patients. In left ventricular tissue, hnRNPL was detected predominantly in nuclei. Knockdown of the hnRNPL homolog Smooth in Drosophila induced cardiomyopathy. Computational analysis of predicted mouse and human hnRNPL binding sites suggested hnRNPL-mediated alternative splicing of tropomyosin, which was confirmed in C2C12 myoblasts. These findings identify hnRNPL as a sensor of cardiac dysfunction and suggest that disturbances of hnRNPL affect alternative splicing in HF.
Collapse
Affiliation(s)
- Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Wanting Huang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Suchita Pande
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Aaron Zou
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Timothy D Calamaras
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Richard H Choe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | | | - Ariel L Mei
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Howard H Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Hannah R Littel
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Mekala Gunasekaran
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Natalya M Wells
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Christine C Bruels
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Audrey L Daugherty
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Wolf
- Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| | - Peter B Kang
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Vicky K Yang
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | | | - Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
33
|
Shin S, Kowahl N, Hansen T, Ling AY, Barman P, Cauwenberghs N, Rainaldi E, Short S, Dunn J, Shandhi MMH, Shah SH, Mahaffey KW, Kuznetsova T, Daubert MA, Douglas PS, Haddad F, Kapur R. Real-world walking behaviors are associated with early-stage heart failure: a Project Baseline Health Study. J Card Fail 2024; 30:1423-1433. [PMID: 38582256 DOI: 10.1016/j.cardfail.2024.02.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Data collected via wearables may complement in-clinic assessments to monitor subclinical heart failure (HF). OBJECTIVES Evaluate the association of sensor-based digital walking measures with HF stage and characterize their correlation with in-clinic measures of physical performance, cardiac function and participant reported outcomes (PROs) in individuals with early HF. METHODS The analyzable cohort included participants from the Project Baseline Health Study (PBHS) with HF stage 0, A, or B, or adaptive remodeling phenotype (without risk factors but with mild echocardiographic change, termed RF-/ECHO+) (based on available first-visit in-clinic test and echocardiogram results) and with sufficient sensor data. We computed daily values per participant for 18 digital walking measures, comparing HF subgroups vs stage 0 using multinomial logistic regression and characterizing associations with in-clinic measures and PROs with Spearman's correlation coefficients, adjusting all analyses for confounders. RESULTS In the analyzable cohort (N=1265; 50.6% of the PBHS cohort), one standard deviation decreases in 17/18 walking measures were associated with greater likelihood for stage-B HF (multivariable-adjusted odds ratios [ORs] vs stage 0 ranging from 1.18-2.10), or A (ORs vs stage 0, 1.07-1.45), and lower likelihood for RF-/ECHO+ (ORs vs stage 0, 0.80-0.93). Peak 30-minute pace demonstrated the strongest associations with stage B (OR vs stage 0=2.10; 95% CI:1.74-2.53) and A (OR vs stage 0=1.43; 95% CI:1.23-1.66). Decreases in 13/18 measures were associated with greater likelihood for stage-B HF vs stage A. Strength of correlation with physical performance tests, echocardiographic cardiac-remodeling and dysfunction indices and PROs was greatest in stage B, then A, and lowest for 0. CONCLUSIONS Digital measures of walking captured by wearable sensors could complement clinic-based testing to identify and monitor pre-symptomatic HF.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicholas Cauwenberghs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | | | - Sarah Short
- Verily Life Sciences; South San Francisco, CA
| | - Jessilyn Dunn
- Duke University Department of Biomedical Engineering; Durham, NC; Duke University Department of Biostatistics & Bioinformatics; Durham, NC; Duke Clinical Research Institute; Durham, NC
| | - Md Mobashir Hasan Shandhi
- Duke Clinical Research Institute; Durham, NC; Division of Cardiology, Duke University Medical School; Duke University; Durham, NC
| | - Svati H Shah
- Duke Clinical Research Institute; Durham, NC; Division of Cardiology, Duke University Medical School; Duke University; Durham, NC
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford School of Medicine; Stanford, CA
| | - Tatiana Kuznetsova
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Melissa A Daubert
- Duke Clinical Research Institute; Durham, NC; Division of Cardiology, Duke University Medical School; Duke University; Durham, NC
| | - Pamela S Douglas
- Duke Clinical Research Institute; Durham, NC; Division of Cardiology, Duke University Medical School; Duke University; Durham, NC
| | - Francois Haddad
- Stanford Center for Clinical Research, Department of Medicine, Stanford School of Medicine; Stanford, CA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University; Stanford, CA; Stanford Cardiovascular Institute, Stanford University; Stanford, CA
| | - Ritu Kapur
- Verily Life Sciences; South San Francisco, CA; Department of Neurology, Radboud University Medical Center; Nijmegen, The Netherlands
| |
Collapse
|
34
|
Figueiral M, Paldino A, Wilke MVMB, Farris JD, Verheijen J, Giudicessi JR, Ackerman MJ, Olson JE, Arroyo J, Olson RJ, Klee EW, Pereira NL. Prevalence, Penetrance, and Phenotypic Manifestation of Cardiomyopathy-Associated Genetic Variants in the General Population: Insights from a Mayo Clinic Biobank Study. Mayo Clin Proc 2024; 99:1732-1743. [PMID: 39387793 DOI: 10.1016/j.mayocp.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To determine the prevalence, penetrance, and disease expression of cardiomyopathy-related genetic variants in an unselected, richly phenotyped Mayo Clinic population in the setting of preemptive sequencing, with return of incidental findings following the American College of Medical Genetics and Genomics recommendations. PATIENTS AND METHODS We analyzed a quaternary medical center-based biobank cohort (n=983) for reportable variants in 15 cardiomyopathy genes. Prioritization of genetic variants was performed using an internally developed pipeline to identify potentially reportable variants. Prioritized variants were then manually curated. The correlation of likely pathogenic/pathogenic (LP/P) variants with clinical phenotypes and outcomes was established. Artificial intelligence-enabled electrocardiographic predictions of reduced left ventricular ejection fraction and hypertrophic cardiomyopathy were applied to genotype-positive (G+) participants. RESULTS Of the 983 patients, 11 (1%) were G+, with 11 LP/P variants found in the MYBPC3, DSG2, MYH7, DSP, and PKP2 genes. All G+ participants underwent electrocardiography, and 10 (90%) underwent echocardiography. Most patients (10 [90%]) did not have a prior diagnosis of cardiomyopathy. Definitive disease penetrance (heart failure or cardiomyopathy) was present in 4 (36%), while 3 (27%) had possible penetrance (structural heart disease identified by echocardiography). Arrhythmias and/or cardiac conduction disease was present in 4 of 11 G+ individuals (36%). Artificial intelligence-electrocardiography was positive for hypertrophic cardiomyopathy or reduced left ventricular ejection fraction in 5 of the G+ participants (45%), of whom 4 (80%) had definitive or possible disease penetrance. CONCLUSION Cardiomyopathy-associated LP/P variants are present in a small subset of a quaternary medical center population, and disease penetrance in G+ individuals is high in the form of cardiac structural abnormalities and heart failure.
Collapse
Affiliation(s)
- Marta Figueiral
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN; Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Alessia Paldino
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN; Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina(ASUGI), University of Trieste, Trieste, Italy
| | | | - Joseph D Farris
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Jan Verheijen
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - John R Giudicessi
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Janet E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Jennifer Arroyo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Rory J Olson
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Naveen L Pereira
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
35
|
Josephs KS, Seaby EG, May P, Theotokis P, Yu J, Andreou A, Sinclair H, Morris-Rosendahl D, Thomas ERA, Ennis S, Roberts AM, Ware JS. Cardiomyopathies in 100,000 genomes project: interval evaluation improves diagnostic yield and informs strategies for ongoing gene discovery. Genome Med 2024; 16:125. [PMID: 39472908 PMCID: PMC11520845 DOI: 10.1186/s13073-024-01390-9] [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: 07/30/2023] [Accepted: 09/24/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Cardiomyopathies are clinically important conditions, with a strong genetic component. National genomic initiatives such as 100,000 Genome Project (100KGP) provide opportunity to study these rare conditions at scale beyond conventional research studies. METHODS We present the clinical and molecular characteristics of the 100KGP cohort, comparing paediatric and adult probands with diverse cardiomyopathies. We assessed the diagnostic yield and spectrum of genetic aetiologies across clinical presentations. We re-analysed existing genomic data using an updated analytical strategy (revised gene panels; unbiased analyses of de novo variants; and improved variant prioritisation strategies) to identify new causative variants in genetically unsolved children. RESULTS We identified 1918 individuals (1563 probands, 355 relatives) with cardiomyopathy (CM) in 100KGP. Probands, comprising 273 children and 1290 adults, were enrolled under > 55 different recruitment categories. Paediatric probands had higher rates of co-existing congenital heart disease (12%) compared to adults (0.9%). Diagnostic yield following 100KGP's initial analysis was significantly higher for children (19%) than for adults (11%) with 11% of diagnoses overall made in genes not on the existing UK paediatric or syndromic CM panel. Our re-analysis of paediatric probands yields a potential diagnosis in 40%, identifying new probable or possible diagnoses in 49 previously unsolved paediatric cases. Structural and intronic variants accounted for 11% of all potential diagnoses in children while de novo variants were identified in 17%. CONCLUSIONS 100KGP demonstrates the benefit of genome sequencing over a standalone panel in CM. Re-analysis of paediatric CM probands allowed a significant uplift in diagnostic yield, emphasising the importance of iterative re-evaluation in genomic studies. Despite these efforts, many children with CM remain without a genetic diagnosis, highlighting the need for better gene-disease relationship curation and ongoing data sharing. The 100KGP CM cohort is likely to be useful for further gene discovery, but heterogeneous ascertainment and key technical limitations must be understood and addressed.
Collapse
Affiliation(s)
- Katherine S Josephs
- National Heart and Lung Institute, Imperial College London, London, UK.
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Eleanor G Seaby
- Genomic Informatics Group, Faculty of Medicine, University of Southampton, Southampton, UK
- Paediatric Infectious Diseases, Imperial College London, St Mary's Hospital, London, UK
- Translational Genomics Group, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philippa May
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Pantazis Theotokis
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Jing Yu
- The Innovation Building, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Avgi Andreou
- SSt George's University Hospitals NHS Foundation Trust, St George's University of London, London, UK
| | | | - Deborah Morris-Rosendahl
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Sarah Ennis
- Genomic Informatics Group, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Angharad M Roberts
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| |
Collapse
|
36
|
Lee DSM, Cardone KM, Zhang DY, Tsao NL, Abramowitz S, Sharma P, DePaolo JS, Conery M, Aragam KG, Biddinger K, Dilitikas O, Hoffman-Andrews L, Judy RL, Khan A, Kulo I, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany ZP, Cappola TP, Carruth E, Day SM, Do R, Haggarty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common- and rare-variant genetic architecture of heart failure across the allele frequency spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.16.23292724. [PMID: 37503172 PMCID: PMC10371173 DOI: 10.1101/2023.07.16.23292724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Heart failure (HF) is a complex trait, influenced by environmental and genetic factors, which affects over 30 million individuals worldwide. Historically, the genetics of HF have been studied in Mendelian forms of disease, where rare genetic variants have been linked to familial cardiomyopathies. More recently, genome-wide association studies (GWAS) have successfully identified common genetic variants associated with risk of HF. However, the relative importance of genetic variants across the allele-frequency spectrum remains incompletely characterized. Here, we report the results of common- and rare-variant association studies of all-cause heart failure, applying recently developed methods to quantify the heritability of HF attributable to different classes of genetic variation. We combine GWAS data across multiple populations including 207,346 individuals with HF and 2,151,210 without, identifying 176 risk loci at genome-wide significance (P-value < 5×10-8). Signals at newly identified common-variant loci include coding variants in Mendelian cardiomyopathy genes (MYBPC3, BAG3) and in regulators of lipoprotein (LPL) and glucose metabolism (GIPR, GLP1R). These signals are enriched in myocyte and adipocyte cell types and can be clustered into 5 broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity, and arrhythmias. Gene burden studies across three biobanks (PMBB, UKB, AOU), including 27,208 individuals with HF and 349,126 without, uncover exome-wide significant (P-value < 1.57×10-6) associations for HF and rare predicted loss-of-function (pLoF) variants in TTN, MYBPC3, FLNC, and BAG3. Total burden heritability of rare coding variants (2.2%, 95% CI 0.99-3.5%) is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common variant heritability (4.3%, 95% CI 3.9-4.7%) is more diffusely spread throughout the genome. Finally, we show that common-variant background, in the form of a polygenic risk score (PRS), significantly modifies the risk of HF among carriers of pathogenic truncating variants in the Mendelian cardiomyopathy gene TTN. Together, these findings provide a genetic link between dysregulated metabolism and HF, and suggest a significant polygenic component to HF exists that is not captured by current clinical genetic testing.
Collapse
Affiliation(s)
- David S M Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Iftikhar Kulo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Nosheen Reza
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Zoltan P Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Eric Carruth
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York, NY
- Biome Phenomics Center, Mount Sinai Icahn School of Medicine, New York, NY
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York, NY
| | | | - Jacob Joseph
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York, NY
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
- Atlanta VA Health Care System, Decatur, GA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Michael G Levin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
37
|
Binzenhöfer L, Clauss S, Strauß K, Höpler J, Kraft M, Hoffmann S, Brunner S, Tomsits P, Schüttler D, Massberg S, Kääb S, Lüsebrink E. Lifetime cumulative activity burden is associated with symptomatic heart failure and arrhythmic risk in patients with arrhythmogenic right ventricular cardiomyopathy: a retrospective cohort study. Europace 2024; 26:euae236. [PMID: 39305246 PMCID: PMC11481332 DOI: 10.1093/europace/euae236] [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: 07/03/2024] [Accepted: 09/10/2024] [Indexed: 10/17/2024] Open
Abstract
AIMS Sports-related physical activity is associated with an increased risk of ventricular dysfunction and arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC). However, there are currently no standardized strategies for activity assessment. Thresholds for harmful levels of physical activity suggested by previous studies vary substantially and neither lifetime activity burden nor continuous modelling approaches were considered. METHODS AND RESULTS For this single-centre retrospective study, ARVC patients were interviewed to assess sports-related and non-sports-related physical activity between the age of 10 years and the last follow-up. Activity data were aggregated to the median metabolic equivalent of task-hours (METh) per week for each year. The association between cumulative physical activity burden and clinical study endpoints was investigated using Cox regression models. A total of 124 patients (median age: 39.5 years, 48% male) were included in the analysis, of whom 93 had been diagnosed with definite ARVC. Study participants reported a median overall activity of 202.3 METh/week, with 38.7 METh/week attributed to sports-related activity. In the continuous model, cumulative overall activity burden was associated with the occurrence of symptomatic heart failure [hazard ratio (HR) per 100 METh/week: 1.017, 95% CI (1.003, 1.032), P = 0.015], sustained ventricular tachycardia [HR: 1.021, 95% CI (1.006, 1.037), P = 0.007], and implantable cardioverter defibrillator interventions [HR: 1.017, 95%CI (1.000, 1.034), P = 0.048]. This finding was consistent when considering sports-related activity separately as a predictor variable, whereas the resulting hazard ratios did not show a significant association for non-sports-related physical activity. CONCLUSION This study demonstrates for the first time that cumulative physical activity as a continuous predictor variable is associated with symptomatic heart failure and arrhythmic risk in ARVC patients. Collaborative research is required in larger cohorts to investigate the influence of potential confounders on event occurrence and to develop threshold recommendations for clinical practice.
Collapse
Affiliation(s)
- Leonhard Binzenhöfer
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Center of Experimental Medicine, LMU University Hospital, Marchioninistraße 68, 81377 Munich, Germany
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), Feodor-Lynen-Straße 19, 81377 Munich, Germany
| | - Katharina Strauß
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Julia Höpler
- Department of Statistics, Institute of Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Marie Kraft
- Department of Statistics, Institute of Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Sabine Hoffmann
- Department of Statistics, Institute of Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Stefan Brunner
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Center for Sports Medicine, LMU University Hospital, Ziemssenstrasse 5, 80336 Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Center of Experimental Medicine, LMU University Hospital, Marchioninistraße 68, 81377 Munich, Germany
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Center of Experimental Medicine, LMU University Hospital, Marchioninistraße 68, 81377 Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), Feodor-Lynen-Straße 19, 81377 Munich, Germany
| | - Stefan Kääb
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), Feodor-Lynen-Straße 19, 81377 Munich, Germany
| | - Enzo Lüsebrink
- Department of Medicine I, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Pettenkoferstraße 8a/9, 80336 Munich, Germany
| |
Collapse
|
38
|
Gill R, Siddiqui A, Yee B, DiCaro MV, Houshmand N, Tak T. Advancements in the Diagnosis and Treatment of Hypertrophic Cardiomyopathy: A Comprehensive Review. J Cardiovasc Dev Dis 2024; 11:290. [PMID: 39330348 PMCID: PMC11431942 DOI: 10.3390/jcdd11090290] [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: 08/09/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by excessive growth of myocardial tissue, most commonly due to genetic mutations in sarcomere proteins. This can lead to complications such as heart failure, mitral regurgitation, syncope, arrhythmias, sudden cardiac death, and myocardial ischemia. While we have come a long way in our understanding of the pathophysiology, genetics, and epidemiology of HCM, the past 10 years have seen significant advancements in diagnosis and treatment. As the body of evidence on hypertrophic cardiomyopathy continues to grow, a comprehensive review of the current literature is an invaluable resource in organizing this knowledge. By doing so, the vast progress that has been made thus far will be widely available to all experts in the field. This review provides a comprehensive analysis of the scientific literature, exploring both well-established and cutting-edge diagnostic and therapeutic options. It also presents a unique perspective by incorporating topics such as exercise testing, genetic testing, radiofrequency ablation, risk stratification, and symptomatic management in non-obstructive HCM. Lastly, this review highlights areas where current and future research is at the forefront of innovation in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Randeep Gill
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
| | - Arsalan Siddiqui
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
| | - Brianna Yee
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
| | - Michael V DiCaro
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
| | - Nazanin Houshmand
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
| | - Tahir Tak
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV 89102, USA
- VA Southern Nevada Healthcare System, 6900 N. Pecos Road, North Las Vegas, NV 89086, USA
| |
Collapse
|
39
|
Inoue S, Ko T, Shindo A, Nomura S, Yamada T, Jimba T, Dai Z, Nakao H, Suzuki A, Kashimura T, Iwahana T, Goto K, Matsushima S, Ishida J, Amiya E, Zhang B, Kubota M, Sawami K, Heryed T, Yamada S, Katoh M, Katagiri M, Ito M, Nayakama Y, Fujiu K, Hatano M, Takeda N, Takimoto E, Akazawa H, Morita H, Yamaguchi J, Inomata T, Kobayashi Y, Minamino T, Tsutsui H, Kurokawa M, Aiba A, Aburatani H, Komuro I. Association Between Clonal Hematopoiesis and Left Ventricular Reverse Remodeling in Nonischemic Dilated Cardiomyopathy. JACC Basic Transl Sci 2024; 9:956-967. [PMID: 39297129 PMCID: PMC11405799 DOI: 10.1016/j.jacbts.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 09/21/2024]
Abstract
Although clonal hematopoiesis of indeterminate potential (CHIP) is an adverse prognostic factor for atherosclerotic disease, its impact on nonischemic dilated cardiomyopathy (DCM) is elusive. The authors performed whole-exome sequencing and deep target sequencing among 198 patients with DCM and detected germline mutations in cardiomyopathy-related genes and somatic mutations in CHIP driver genes. Twenty-five CHIP driver mutations were detected in 22 patients with DCM. Ninety-two patients had cardiomyopathy-related pathogenic mutations. Multivariable analysis revealed that CHIP was an independent risk factor of left ventricular reverse remodeling, irrespective of known prognostic factors. CHIP exacerbated cardiac systolic dysfunction and fibrosis in a DCM murine model. The identification of germline and somatic mutations in patients with DCM predicts clinical prognosis.
Collapse
Affiliation(s)
- Shunsuke Inoue
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akito Shindo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takanobu Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Jimba
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Zhehao Dai
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Harumi Nakao
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Suzuki
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takeshi Kashimura
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Togo Iwahana
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Keiko Goto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Ishida
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eisuke Amiya
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Bo Zhang
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Kubota
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Sawami
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tuolisi Heryed
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shintaro Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mikako Katagiri
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiteru Nayakama
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaru Hatano
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Yamaguchi
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takayuki Inomata
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- School of Medicine and Graduate School, International University of Health and Welfare, Okawa City, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- International University of Health and Welfare, Tokyo, Japan
- Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Murphy J, Kirk CW, Lambert DM, McGorrian C, Walsh R, McVeigh TP, Prendiville T, Ward D, Galvin J, Lynch SA. Diagnostic yield from cardiac gene testing for inherited cardiac conditions and re-evaluation of pre-ACMG variants of uncertain significance. Ir J Med Sci 2024; 193:1775-1785. [PMID: 38489124 DOI: 10.1007/s11845-024-03650-4] [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] [Accepted: 02/23/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Inherited cardiomyopathies (HCM, DCM, ACM) and cardiac ion channelopathies (long QT/Brugada syndromes, CPVT) are associated with significant morbidity and mortality; however, diagnosis of a familial pathogenic variant in a proband allows for subsequent cascade screening of their at-risk relatives. AIMS We investigated the diagnostic yield from cardiac gene panel testing and reviewed variants of uncertain significance from patients attending three specialist cardiogenetics services in Ireland in the years 2002 to 2020. RESULTS Reviewing molecular genetic diagnostic reports of 834 patients from 820 families, the initial diagnostic yield of pathogenic/likely pathogenic variants was 237/834 patients (28.4%), increasing to 276/834 patients (33.1%) following re-evaluation of cases with variant(s) of uncertain significance. Altogether, 42/85 patients with VUS reviewed (49.4%) had a re-classification that could change their clinical management. Females were more likely to carry pathogenic/likely pathogenic variants than males (139/374, 37.2% vs 137/460, 29.8%, respectively, p = 0.03), and the diagnostic yields were highest in the 0 to < 2 years age group (6/12, 50.0%) and amongst those tested for cardiomyopathy gene panels (13/35, 37.1%). Variants in the MYBPC3/MYH7 (87/109, 79.8%) and KCNQ1/KCNH2 (91/100, 91.0%) genes were the predominant genetic causes for hypertrophic cardiomyopathy and long QT syndrome, respectively. CONCLUSION Our study highlights the importance of collation and review of pre-ACMG genetic variants to increase diagnostic utility of genetic testing for inherited heart disease. Almost half of patients with pre-ACMG VUS reviewed had their variant re-classified to likely pathogenic/likely benign which resulted in a positive clinical impact for patients and their families.
Collapse
Affiliation(s)
- Jane Murphy
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Claire W Kirk
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Deborah M Lambert
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Catherine McGorrian
- Family Heart Screening Clinic, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Terri P McVeigh
- Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, United Kingdom
| | - Terence Prendiville
- Department of Cardiology, Children's Health Ireland at Crumlin, Crumlin, Dublin 12, Ireland
| | - Deirdre Ward
- Centre for Cardiac Risk in the Young Persons, Tallaght University Hospital, Dublin 24, Ireland
| | - Joseph Galvin
- Family Heart Screening Clinic, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Sally Ann Lynch
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Crumlin, Dublin 12, Ireland
| |
Collapse
|
41
|
Liu Y, Luo J, Peng L, Zhang Q, Rong X, Luo Y, Li J. Flavonoids: Potential therapeutic agents for cardiovascular disease. Heliyon 2024; 10:e32563. [PMID: 38975137 PMCID: PMC11225753 DOI: 10.1016/j.heliyon.2024.e32563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Flavonoids are found in the roots, stems, leaves, and fruits of many plant taxa. They are related to plant growth and development, pigment formation, and protection against environmental stress. Flavonoids function as antioxidants and exert anti-inflammatory effects in the cardiovascular system by modulating classical inflammatory response pathways, such as the TLR4-NF-ĸB, PI3K-AKT, and Nrf2/HO-1 signalling pathways. There is increasing evidence for the therapeutic effects of flavonoids on hypertension, atherosclerosis, and other diseases. The potential clinical value of flavonoids for diseases of the cardiovascular system has been widely explored. For example, studies have evaluated the roles of flavonoids in the regulation of blood pressure via endothelium-dependent and non-endothelium-dependent pathways and in the regulation of myocardial systolic and diastolic functions by influencing calcium homeostasis and smooth muscle-related protein expression. Flavonoids also have hypoglycaemic, hypolipidemic, anti-platelet, autophagy, and antibacterial effects. In this paper, the role and mechanism of flavonoids in cardiovascular diseases were reviewed in order to provide reference for the clinical application of flavonoids in the future.
Collapse
Affiliation(s)
- Yingxue Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Peng
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qi Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xi Rong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, China
| |
Collapse
|
42
|
Li Y, Ma K, Dong Z, Gao S, Zhang J, Huang S, Yang J, Fang G, Li Y, Li X, Welch C, Griffin EL, Ramaswamy P, Valivullah Z, Liu X, Dong J, Wang DW, Du, Chung WK, Li Y. Frameshift variants in C10orf71 cause dilated cardiomyopathy in human, mouse, and organoid models. J Clin Invest 2024; 134:e177172. [PMID: 38950288 PMCID: PMC11178530 DOI: 10.1172/jci177172] [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: 11/01/2023] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Ke Ma
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zhujun Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jing Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jie Yang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Guangming Fang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Yujie Li
- Novogene Co. Ltd., Beijing, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Carrie Welch
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Emily L. Griffin
- Department of Pediatrics, Columbia University, New York, New York, USA
| | | | | | | | - Jianzeng Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Wendy K. Chung
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
43
|
Li X, Li J, Samuelsson AM, Thakur H, Kapiloff MS. Protein phosphatase 2A anchoring disruptor gene therapy for familial dilated cardiomyopathy. Mol Ther Methods Clin Dev 2024; 32:101233. [PMID: 38572067 PMCID: PMC10988123 DOI: 10.1016/j.omtm.2024.101233] [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: 11/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
Familial dilated cardiomyopathy is a prevalent cause of heart failure that results from the mutation of genes encoding proteins of diverse function. Despite modern therapy, dilated cardiomyopathy typically has a poor outcome and is the leading cause of cardiac transplantation. The phosphatase PP2A at cardiomyocyte perinuclear mAKAPβ signalosomes promotes pathological eccentric cardiac remodeling, as is characteristic of dilated cardiomyopathy. Displacement of PP2A from mAKAPβ, inhibiting PP2A function in that intracellular compartment, can be achieved by expression of a mAKAPβ-derived PP2A binding domain-derived peptide. To test whether PP2A anchoring disruption would be effective at preventing dilated cardiomyopathy-associated cardiac dysfunction, the adeno-associated virus gene therapy vector AAV9sc.PBD was devised to express the disrupting peptide in cardiomyocytes in vivo. Proof-of-concept is now provided that AAV9sc.PBD improves the cardiac structure and function of a cardiomyopathy mouse model involving transgenic expression of a mutant α-tropomyosin E54K Tpm1 allele, while AAV9sc.PBD has no effect on normal non-transgenic mice. At the cellular level, AAV9sc.PBD restores cardiomyocyte morphology and gene expression in the mutant Tpm1 mouse. As the mechanism of AAV9sc.PBD action suggests potential efficacy in dilated cardiomyopathy regardless of the underlying etiology, these data support the further testing of AAV9sc.PBD as a broad-based treatment for dilated cardiomyopathy.
Collapse
Affiliation(s)
- Xueyi Li
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Jinliang Li
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Anne-Maj Samuelsson
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Hrishikesh Thakur
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Michael S. Kapiloff
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
44
|
Jia H, Chang Y, Song J. The pig as an optimal animal model for cardiovascular research. Lab Anim (NY) 2024; 53:136-147. [PMID: 38773343 DOI: 10.1038/s41684-024-01377-4] [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: 09/02/2022] [Accepted: 04/22/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is a worldwide health problem and a leading cause of morbidity and mortality. Preclinical cardiovascular research using animals is needed to explore potential targets and therapeutic options. Compared with rodents, pigs have many advantages, with their anatomy, physiology, metabolism and immune system being more similar to humans. Here we present an overview of the available pig models for cardiovascular diseases, discuss their advantages over other models and propose the concept of standardized models to improve translation to the clinical setting and control research costs.
Collapse
Affiliation(s)
- Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Sanya Institute of China Agricultural University, Sanya, China.
| |
Collapse
|
45
|
Nosetti L, Zaffanello M, Lombardi C, Gerosa A, Piacentini G, Abramo M, Agosti M. Early Screening for Long QT Syndrome and Cardiac Anomalies in Infants: A Comprehensive Study. Clin Pract 2024; 14:1038-1053. [PMID: 38921260 PMCID: PMC11203353 DOI: 10.3390/clinpract14030082] [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: 02/11/2024] [Revised: 03/26/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
(1) Background: Sudden Infant Death Syndrome (SIDS) represents sudden and unexplained deaths during the sleep of infants under one year of age, despite thorough investigation. Screening for a prolonged QTc interval, a marker for Long QT Syndrome (LQTS), should be conducted on all newborns to reduce the incidence of SIDS. Neonatal electrocardiograms (ECGs) could identify congenital heart defects (CHDs) early, especially those not detected at birth. Infants with prolonged QTc intervals typically undergo genetic analysis for Long QT Syndrome. (2) Methods: The study involved infants aged 20-40 days, born with no apparent clinical signs of heart disease, with initial ECG screening. Infants with prenatal diagnoses or signs/symptoms of CHDs identified immediately after birth, as well as infants who had previously had an ECG or echocardiogram for other medical reasons, were excluded from the study. We used statistical software (SPSS version 22.0) to analyze the data. (3) Results: Of the 42,200 infants involved, 2245 were enrolled, with 39.9% being males. Following this initial screening, 164 children (37.8% males) with prolonged QTc intervals underwent further evaluation. Out of these 164 children, 27 children were confirmed to have LQTS. However, only 18 children were finally investigated for genetic mutations, and mutations were identified in 11 tests. The most common mutations were LQT1 (54.5%), LQT2 (36.4%), and LQT3 (1 patient). Treatment options included propranolol (39.8%), nadolol (22.2%), inderal (11.1%), metoprolol (11.1%), and no treatment (16.7%). The most common abnormalities were focal right bundle branch block (54.5%), left axis deviation (9.2%), and nonspecific ventricular repolarization abnormalities (7.1%). Multiple anomalies were found in 0.47% of children with focal right bundle branch block. Structural abnormalities were associated with specific features in 267 patients (11.9%), primarily isolated patent foramen ovale (PFO) at 61.4%. (4) Conclusions: This screening approach has demonstrated effectiveness in the early identification of LQTS and other cardiac rhythm anomalies, with additional identification of mutations and/or prolonged QTc intervals in family members. Identifying other ECG abnormalities and congenital heart malformations further enhances the benefits of the screening.
Collapse
Affiliation(s)
- Luana Nosetti
- Pediatric Sleep Disorders Center, Division of Pediatrics, “F. Del Ponte” Hospital, University of Insubria, 21100 Varese, Italy; (L.N.)
| | - Marco Zaffanello
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, 37100 Verona, Italy
| | - Carolina Lombardi
- Sleep Disorders Center, Department of Cardiology Istituto Auxologico, IRCCS, 20149 Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Alessandra Gerosa
- Pediatric Sleep Disorders Center, Division of Pediatrics, “F. Del Ponte” Hospital, University of Insubria, 21100 Varese, Italy; (L.N.)
| | - Giorgio Piacentini
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, 37100 Verona, Italy
| | - Michele Abramo
- Pediatric Sleep Disorders Center, Division of Pediatrics, “F. Del Ponte” Hospital, University of Insubria, 21100 Varese, Italy; (L.N.)
| | - Massimo Agosti
- Woman and Child Department, Varese Hospital, Insubria University, Via Ravasi 2, 21100 Varese, Italy;
| |
Collapse
|
46
|
Chakraborti A, Tardiff JC, Schwartz SD. Myosin-Catalyzed ATP Hydrolysis in the Presence of Disease-Causing Mutations: Mavacamten as a Way to Repair Mechanism. J Phys Chem B 2024; 128:4716-4727. [PMID: 38708944 PMCID: PMC11103257 DOI: 10.1021/acs.jpcb.4c01601] [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] [Indexed: 05/07/2024]
Abstract
Hypertrophic cardiomyopathy is one of the most common forms of genetic cardiomyopathy. Mavacamten is a first-in-class myosin modulator that was identified via activity screening on the wild type, and it is FDA-approved for the treatment of obstructive hypertrophic cardiomyopathy (HCM). The drug selectively binds to the cardiac β-myosin, inhibiting myosin function to decrease cardiac contractility. Though the drug is thought to affect multiple steps of the myosin cross-bridge cycle, its detailed mechanism of action is still under investigation. Individual steps in the overall cross-bridge cycle must be queried to elucidate the full mechanism of action. In this study, we utilize the rare-event method of transition path sampling to generate reactive trajectories to gain insights into the action of the drug on the dynamics and rate of the ATP hydrolysis step for human cardiac β-myosin. We study three known HCM causative myosin mutations: R453C, P710R, and R712L to observe the effect of the drug on the alterations caused by these mutations in the chemical step. Since the crystal structure of the drug-bound myosin was not available at the time of this work, we created a model of the drug-bound system utilizing a molecular docking approach. We find a significant effect of the drug in one case, where the actual mechanism of the reaction is altered from the wild type by mutation. The drug restores both the rate of hydrolysis to the wildtype level and the mechanism of the reaction. This is a way to check the effect of the drug on untested mutations.
Collapse
Affiliation(s)
- Ananya Chakraborti
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Jil C Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85724, United States
| | - Steven D Schwartz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
47
|
Gray MP, Fatkin D, Ingles J, Robertson EN, Figtree GA. Genetic testing in cardiovascular disease. Med J Aust 2024; 220:428-434. [PMID: 38571440 DOI: 10.5694/mja2.52278] [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: 04/28/2023] [Accepted: 01/08/2024] [Indexed: 04/05/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality globally and is responsible for an estimated one-third of deaths as well as significant morbidity and health care utilisation. Technological and bioinformatic advances have facilitated the discovery of pathogenic germline variants for some specific CVDs, including familial hypercholesterolaemia, cardiomyopathies and arrhythmic syndromes. Use of these genetic tests for earlier disease identification is increasing due, in part, to decreasing costs, Medicare rebates, and consumer comfort with genetic testing. However, CVDs that occur more commonly, including coronary artery disease and atrial fibrillation, do not display monogenic inheritance patterns. Genetically, these diseases have generally been associated with many genetic variants each with a small effect size. This complexity can be expressed mathematically as a polygenic risk score. Genetic testing kits that provide polygenic risk scoring are becoming increasingly available directly to private-paying consumers outside the traditional clinical setting. An improved understanding of the evidence of genetics in CVD will offer clinicians new opportunities for individualised risk prediction and preventive therapy.
Collapse
Affiliation(s)
- Michael P Gray
- University of Sydney, Sydney, NSW
- Kolling Institute, Sydney, NSW
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Sydney, NSW
| | - Jodie Ingles
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, NSW
| | | | - Gemma A Figtree
- University of Sydney, Sydney, NSW
- Kolling Institute, Sydney, NSW
- Royal North Shore Hospital, Sydney, NSW
| |
Collapse
|
48
|
Dababneh S, Hamledari H, Maaref Y, Jayousi F, Hosseini DB, Khan A, Jannati S, Jabbari K, Arslanova A, Butt M, Roston TM, Sanatani S, Tibbits GF. Advances in Hypertrophic Cardiomyopathy Disease Modelling Using hiPSC-Derived Cardiomyocytes. Can J Cardiol 2024; 40:766-776. [PMID: 37952715 DOI: 10.1016/j.cjca.2023.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/21/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
The advent of human induced pluripotent stem cells (hiPSCs) and their capacity to be differentiated into beating human cardiomyocytes (CMs) in vitro has revolutionized human disease modelling, genotype-phenotype predictions, and therapeutic testing. Hypertrophic cardiomyopathy (HCM) is a common inherited cardiomyopathy and the leading known cause of sudden cardiac arrest in young adults and athletes. On a molecular level, HCM is often driven by single pathogenic genetic variants, usually in sarcomeric proteins, that can alter the mechanical, electrical, signalling, and transcriptional properties of the cell. A deeper knowledge of these alterations is critical to better understanding HCM manifestation, progression, and treatment. Leveraging hiPSC-CMs to investigate the molecular mechanisms driving HCM presents a unique opportunity to dissect the consequences of genetic variants in a sophisticated and controlled manner. In this review, we summarize the molecular underpinnings of HCM and the role of hiPSC-CM studies in advancing our understanding, and we highlight the advances in hiPSC-CM-based modelling of HCM, including maturation, contractility, multiomics, and genome editing, with the notable exception of electrophysiology, which has been previously covered.
Collapse
Affiliation(s)
- Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yasaman Maaref
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dina B Hosseini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aasim Khan
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shayan Jannati
- Faculty of Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kosar Jabbari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shubhayan Sanatani
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
49
|
Bakalakos A, Monda E, Elliott PM. The Diagnostic and Therapeutic Implications of Phenocopies and Mimics of Hypertrophic Cardiomyopathy. Can J Cardiol 2024; 40:754-765. [PMID: 38447917 DOI: 10.1016/j.cjca.2024.02.025] [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: 01/23/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common myocardial disease defined by increased left ventricular wall thickness unexplained by loading conditions. HCM frequently is caused by pathogenic variants in sarcomeric protein genes, but several other syndromic, metabolic, infiltrative, and neuromuscular diseases can result in HCM phenocopies. This review summarizes the current understanding of these HCM mimics, highlighting their importance across the life course. The central role of a comprehensive, multiparametric diagnostic approach and the potential of precision medicine in tailoring treatment strategies are emphasized.
Collapse
Affiliation(s)
- Athanasios Bakalakos
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Emanuele Monda
- Institute of Cardiovascular Science, University College London, London, United Kingdom; Department of Translational Medical Sciences, Inherited and Rare Cardiovascular Diseases, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Perry Mark Elliott
- Institute of Cardiovascular Science, University College London, London, United Kingdom.
| |
Collapse
|
50
|
Zhang Y, Liu M, Li P, Ma L, Yin L, Cui Y, Huang K, Li W, Xin H, Xiang M, Dai C, Chen M, Feng J, Zheng Z, Xu Y, Chen Y, Jiang X, Ma G, Yang P, Wang F, Li X, Jin W, Chen H, Hui R, Wang L, Xiao H, Liu L, Zou Y, Song L. The burden and management competency of cardiomyopathies in China: a nationwide survey study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 46:101062. [PMID: 38623390 PMCID: PMC11017274 DOI: 10.1016/j.lanwpc.2024.101062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/05/2024] [Accepted: 03/26/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND The public health burden of cardiomyopathies and competency in their management by health agencies in China are not well understood. METHODS This study adopted a multi-stage sampling method for hospital selection. In the first stage, nationwide tertiary hospital recruitment was performed. As a result, 88 hospitals with the consent of the director of cardiology and access to an established electronic medical records system, were recruited. In the second stage, we sampled 66 hospitals within each geographic-economic stratification through a random sampling process. Data on (1) the outpatient and inpatient visits for cardiomyopathies between 2017 and 2021 and (2) the competency in the management of patients with cardiomyopathies, were collected. The competency of a hospital to provide cardiomyopathy care was evaluated using a specifically devised scale. FINDINGS The outpatient and inpatient visits for cardiomyopathies increased between 2017 and 2021 by 38.6% and 33.0%, respectively. Most hospitals had basic facilities for cardiomyopathy assessment. However, access to more complex procedures was limited, and the integrated management pathway needs improvement. Only 4 (6.1%) of the 66 participating hospitals met the criteria for being designated as a comprehensive cardiomyopathy center, and only 29 (43.9%) could be classified as a primary cardiomyopathy center. There were significant variations in competency between hospitals with different administrative and economic levels. INTERPRETATION The health burden of cardiomyopathies has increased significantly between 2017 and 2021 in China. Although most tertiary hospitals in China can offer basic cardiomyopathy care, more advanced facilities are not yet universally available. Moreover, inconsistencies in the management of cardiomyopathies across hospitals due to differing administrative and economic levels warrants a review of the nation allocation of medical resources. FUNDING This work was supported by the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (2023-I2M-1-001) and the National High Level Hospital Clinical Research Funding (2022-GSP-GG-17).
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
| | - Minghao Liu
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peijin Li
- 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
| | - Likun Ma
- Division of Life Sciences and Medicine, Department of Cardiology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), University of Science and Technology of China, Hefei, Anhui, China
| | - Lixue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Cardiovascular Ultrasound and Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yinghua Cui
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Kai Huang
- Department of Cardiology, Western Hospital, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Li
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cuilian Dai
- Department of Cardiology, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Mulei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jinping Feng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihan Chen
- Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuejun Jiang
- Department of Cardiology, Wuhan University People's Hospital, Wuhan, Hubei, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Fang Wang
- Department of Cardiology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Wei Jin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Rutai Hui
- 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
| | - Lian Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hongyan Xiao
- Department of Cardiac Surgery, Wuhan Asia Heart Hospital Affiliated with the Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Liwen Liu
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yubao 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
|