1
|
Bergan N, Prachee I, Curran L, McGurk KA, Lu C, de Marvao A, Bai W, Halliday BP, Gregson J, O’Regan DP, Ware JS, Tayal U. Systematic Review, Meta-Analysis, and Population Study to Determine the Biologic Sex Ratio in Dilated Cardiomyopathy. Circulation 2025; 151:442-459. [PMID: 39895490 PMCID: PMC11827689 DOI: 10.1161/circulationaha.124.070872] [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: 06/07/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) appears to be diagnosed twice as often in male than in female patients. This could be attributed to underdiagnosis in female patients or sex differences in susceptibility. Up to 30% of cases have an autosomal dominant monogenic cause, where equal sex prevalence would be expected. The aim of this systematic review, meta-analysis, and population study was to assess the sex ratio in patients with DCM, stratified by genetic status, and evaluate whether this is influenced by diagnostic bias. METHODS A literature search identified DCM patient cohorts with discernible sex ratios. Exclusion criteria were studies with a small (n<100), pediatric, or peripartum population. Meta-analysis and metaregression compared the proportion of female participants for an overall DCM cohort and the following subtypes: all genetic DCM, individual selected DCM genes (TTN and LMNA), and gene-elusive DCM. Population DCM sex ratios generated from diagnostic codes were also compared with those from sex-specific means using the UK Biobank imaging cohort; this established ICD coded, novel imaging-first, and genotype first determined sex ratios. RESULTS A total of 99 studies, with 37 525 participants, were included. The overall DCM cohort had a 0.30 female proportion (95% CI, 0.28-0.32), corresponding to a male:female ratio (M:F) of 2.38:1. This was similar to patients with an identified DCM variant (0.31 [95% CI, 0.26-0.36]; M:F 2.22:1; P=0.56). There was also no significant difference when compared with patients with gene-elusive DCM (0.30 [95% CI, 0.24-0.37]; M:F 2.29:1; P=0.81). Furthermore, the ratio within autosomal dominant gene variants was not significantly different for TTN (0.28 [95% CI, 0.22-0.36]; M:F 2.51:1; P=0.82) or LMNA (0.35 [95% CI, 0.27-0.44]; M:F 1.84:1; P=0.41). Overall, the sex ratio for DCM in people with disease attributed to autosomal dominant gene variants was similar to the all-cause group (0.34 [95% CI, 0.28-0.40]; M:F 1.98:1; P=0.19). In the UK Biobank (n=47 549), DCM defined by International Classification of Diseases, 10th revision, coding had 4.5:1 M:F. However, implementing sex-specific imaging-first and genotype-first diagnostic approaches changed this to 1.7:1 and 2.3:1, respectively. CONCLUSIONS This study demonstrates that DCM is twice as prevalent in male patients. This was partially mitigated by implementing sex-specific DCM diagnostic criteria. The persistent male excess in genotype-positive patients with an equally prevalent genetic risk suggests additional genetic or environmental drivers for sex-biased penetrance. REGISTRATION URL: https://www.crd.york.ac.uk/prospero; Unique identifier: CRD42023451944.
Collapse
Affiliation(s)
- Natalie Bergan
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
| | - Ishika Prachee
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - Lara Curran
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
| | - Kathryn A. McGurk
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - Chang Lu
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - Antonio de Marvao
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
- Department of Women and Children’s Health and British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, UK (A.d.M.)
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing, London, UK (W.B.)
- Department of Brain Sciences, London, UK (W.B.)
| | - Brian P. Halliday
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - John Gregson
- London School of Hygiene and Tropical Medicine, UK (J.G.)
| | - Declan P. O’Regan
- Institute of Clinical Sciences, London, UK (D.P.O.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - James S. Ware
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Imperial College Healthcare NHS Trust, London, UK (J.S.W.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - Upasana Tayal
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
- The George Institute for Global Health, UK (U.T.)
| |
Collapse
|
2
|
Qin Q, Zhou ZY, Liu Y, Zhou F, Cao C, Teng L. Unraveling the nexus of nesprin in dilated cardiomyopathy: From molecular insights to therapeutic prospects. Life Sci 2024; 358:123126. [PMID: 39396640 DOI: 10.1016/j.lfs.2024.123126] [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: 07/19/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Dilated cardiomyopathy is a complex and debilitating heart disorder characterized by the enlargement and weakening of the cardiac chambers, leading to impaired contractility and heart failure. Nesprins, a family of nuclear envelope spectrin repeat proteins that include isoforms Nesprin-1/-2, are integral components of the LInker of Nucleoskeleton and Cytoskeleton complex. They facilitate the connection between the nuclear envelope and the cytoskeleton, crucial for maintaining nuclear architecture, migration and positioning, and mechanical transduction and signaling. Nesprin-1/-2 are abundantly expressed in cardiac and skeletal muscles.They have emerged as key players in the pathogenesis of dilated cardiomyopathy. Mutations in synaptic nuclear envelope-1/-2 genes encoding Nesprin-1/-2 are associated with dilated cardiomyopathy, underscoring their significance in cardiac health. This review highlights the all known cases of Nesprin-1/-2 related dilated cardiomyopathy, focusing on their interactions with the nuclear envelope, their role in mechanical transduction, and their influence on gene expression. Moreover, it delves into the underlying mechanisms through which Nesprin dysfunction disrupts nuclear-cytoskeletal coupling, leading to abnormal nuclear morphology, impaired mechanotransduction, and altered gene regulation. The exploration of Nesprin's impact on dilated cardiomyopathy offers a promising avenue for therapeutic interventions aimed at ameliorating the disease. This review provides a comprehensive overview of recent advancements in understanding the pivotal role of Nesprins in dilated cardiomyopathy research.
Collapse
Affiliation(s)
- Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Zi-Yi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Yangyuanzhi Liu
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China
| | - Chunyu Cao
- School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China; College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, UK.
| |
Collapse
|
3
|
Anastasiou V, Papazoglou AS, Gossios T, Zegkos T, Daios S, Moysidis DV, Koutsiouroumpa O, Parcharidou D, Tziomalos G, Katranas S, Rouskas P, Didagelos M, Karamitsos T, Ziakas A, McKenna WJ, Kamperidis V, Efthimiadis GK. Prognostic implications of genotype findings in non-ischaemic dilated cardiomyopathy: A network meta-analysis. Eur J Heart Fail 2024; 26:2155-2168. [PMID: 39078390 DOI: 10.1002/ejhf.3403] [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: 02/06/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
AIMS Evidence on the relative impact of diverse genetic backgrounds associated with non-ischaemic dilated cardiomyopathy (DCM) remains contradictory. This study sought to synthesize the available data regarding long-term outcomes of different gene groups in DCM. METHODS AND RESULTS Electronic databases were systematically screened to identify studies reporting prognostic data on pre-specified gene groups. Those included pathogenic/likely pathogenic (P/LP) variants, truncating titin variants (TTNtv), lamin A/C variants (LMNA), and desmosomal proteins. Outcomes were divided into composite adverse events (CAEs), malignant ventricular arrhythmic events (MVAEs) and heart failure events (HFEs). A total of 26 studies (n = 7255) were included in the meta-analysis and 6791 patients with genotyped DCM were analysed. Patients with P/LP variants had a higher risk for CAEs (odds ratio [OR] 2.10, 95% confidence interval [CI] 1.67-2.65), MVAEs (OR 1.86, 95% CI 1.52-2.26), and HFEs (OR 2.01, 95% CI 1.08-3.73) than genotype-negative patients. The presence of TTNtv was linked to a higher risk for CAEs (OR 1.78, 95% CI 1.20-2.63), but not MVAEs or HFEs. LMNA and desmosomal groups suffered a higher risk for CAEs, MVAEs, and HFEs compared to non-LMNA and non-desmosomal groups, respectively. When genes were indirectly compared, the presence of LMNA resulted in a more detrimental effect that TTNtv, with respect to all composite outcomes but no significant difference was found between LMNA and desmosomal genes. Desmosomal genes harboured a higher risk for MVAEs compared to TTNtv. CONCLUSIONS Different genetic substrates associated with DCM result in divergent natural histories. Routine utilization of genetic testing should be employed to refine risk stratification and inform therapeutic strategies in DCM.
Collapse
Affiliation(s)
- Vasileios Anastasiou
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Thomas Gossios
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Thomas Zegkos
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stylianos Daios
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Ourania Koutsiouroumpa
- Evidence Synthesis Methods Team, Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece
| | - Despoina Parcharidou
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Tziomalos
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sotiris Katranas
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pavlos Rouskas
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Matthaios Didagelos
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodoros Karamitsos
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonios Ziakas
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - William J McKenna
- Institute of Cardiovascular Medicine, University College London, London, UK
| | - Vasileios Kamperidis
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios K Efthimiadis
- 1st Department of Cardiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
4
|
Myers MC, Breznen B, Zhong Y, Maruyama S, Bueno C, Bastien A, Fazeli MS, Golchin N. Diverse Concepts in Definitions of Dilated Cardiomyopathy: Theory and Practice. Cardiol Res 2024; 15:319-329. [PMID: 39420975 PMCID: PMC11483116 DOI: 10.14740/cr1679] [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: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 10/19/2024] Open
Abstract
Our understanding of dilated cardiomyopathy (DCM) is evolving as new insights into the underlying pathophysiology become available. Professional organizations and clinical experts are improving definitions of DCM, allowing for more accurate treatment recommendations. This review summarized key published literature describing definitions and/or diagnostic criteria for DCM. Embase was searched from database inception to September 19, 2022 for 1) publications reporting definitions of DCM by major professional organizations and related opinion papers, and 2) clinical studies in DCM and heart failure with reduced ejection fraction. Sixty-eight records were included in this review. Definitions of DCM provided by two major professional organizations (American Heart Association (AHA) and European Society of Cardiology (ESC)) agreed on the clinical presentation of DCM; however, they differed in the classification of DCM within the larger context of cardiomyopathy taxonomies. Both organizations agreed that DCM could be clinically defined by the presence of left ventricular dilation and contractile dysfunction in the absence of abnormal loading conditions and severe coronary artery disease. AHA guidelines divided cardiomyopathies into two major groups (primary and secondary) based on predominant organ involvement. DCM was classified as primary cardiomyopathy with mixed (genetic and/or acquired) etiology. Conversely, ESC published a clinically oriented taxonomy in which cardiomyopathies were grouped into specific morphological and functional phenotypes; each was subclassified into familial or non-familial forms. Opinion papers further elaborated on the complex interplay between genetics and environment in the etiology of DCM. Several articles summarized the importance of the new and updated diagnostic tools, such as cardiac magnetic resonance imaging, electrocardiogram, and other biomarkers, in correctly identifying the etiology of DCM. Within clinical studies, most inclusion criteria used standard definitions proposed by leading professional associations (AHA and ESC). Clinical study investigators sometimes used a narrower definition of DCM using additional criteria for the left ventricular ejection fraction threshold value and left ventricular dilatation. Current efforts in cardiology research are focused on a more granular understanding of DCM etiology and the natural history of the disease. Definitions of DCM found in clinical studies mainly rely on published guidelines, with some studies adding idiosyncratic inclusion criteria refining the broad definitions of DCM.
Collapse
Affiliation(s)
| | | | - Yue Zhong
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | |
Collapse
|
5
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [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: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
6
|
Myers MC, Wang S, Zhong Y, Maruyama S, Bueno C, Bastien A, Fazeli MS, Golchin N. Prevalence of Genetically Associated Dilated Cardiomyopathy: A Systematic Literature Review and Meta-Analysis. Cardiol Res 2024; 15:233-245. [PMID: 39205965 PMCID: PMC11349141 DOI: 10.14740/cr1680] [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: 06/14/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is a leading cause of heart failure and cardiac transplantation globally. Disease-associated genetic variants play a significant role in the development of DCM. Accurately determining the prevalence of genetically associated DCM (genetic DCM) is important for developing targeted prevention strategies. This review synthesized published literature on the global prevalence of genetic DCM across various populations, focusing on two of the most common variants: titin (TTN) and myosin heavy chain 7 (MYH7). Methods MEDLINE® and Embase were searched from database inception to September 19, 2022 for English-language studies reporting the prevalence of genetic DCM within any population. Studies using family history as a proxy for genetic DCM were excluded. Results Of 2,736 abstracts, 57 studies were included. Among the global adult or mixed (mostly adults with few pediatric patients) DCM population, median prevalence was 20.2% (interquartile range (IQR): 16.3-36.0%) for overall genetic DCM, 11.4% (IQR: 8.2-17.8%) for TTN-associated DCM, and 3.2% (IQR: 1.8-5.2%) for MYH7-associated DCM. Global prevalence of overall pediatric genetic DCM within the DCM population was similar (weighted mean: 21.3%). Few studies reported data on the prevalence of genetic DCM within the general population. Conclusions Our study identified variable prevalence estimates of genetic DCM across different populations and geographic locations. The current evidence may underestimate the genetic contributions due to limited screening and detection of potential DCM patients. Epidemiological studies using long-read whole genome sequencing to identify structural variants or non-coding variants are needed, as well as large cohort datasets with genotype-phenotype correlation analyses.
Collapse
Affiliation(s)
| | - Su Wang
- Evidinno Outcomes Research Inc., Vancouver, BC, Canada
| | - Yue Zhong
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | |
Collapse
|
7
|
Martini M, Bueno Marinas M, Rigato I, Pilichou K, Bauce B. Clinical Insights in RNA-Binding Protein Motif 20 Cardiomyopathy: A Systematic Review. Biomolecules 2024; 14:702. [PMID: 38927106 PMCID: PMC11202118 DOI: 10.3390/biom14060702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of heart failure (HF) and heart transplantation (HTx), with genetic factors playing a significant role. In recent years, the RNA-binding protein motif 20 (RBM20), which affects the gene splicing of various proteins with different cellular functions, was identified as the first DCM gene with regulatory properties. Variants of RBM20 have been associated with severe forms of DCM. The aim of this critical systematic review was to analyse RBM20 cardiomyopathy clinical features and outcomes. According to PRISMA guidelines, a search was run in the PubMed, Scopus and Web of Science electronic databases using the following keywords: "RBM20"; "cardiomyopathy"; "arrhythmias"; "heart failure". A total of 181 records were screened, of which 27 studies were potentially relevant to the topic. Through the application of inclusion and exclusion criteria, eight papers reporting 398 patients with RBM20 pathogenic variants were analysed. The mean age at presentation was 41 years. Familiarity with cardiomyopathy was available in 59% of cases, with 55% of probands reporting a positive family history. Imaging data indicated a mild reduction of left ventricular ejection fraction (mean LVEF 40%), while tissue characterization was reported in 24.3% of cases, showing late gadolinium enhancement in 33% of patients. Composite outcomes of sustained monomorphic ventricular tachycardia or ventricular fibrillation occurred in 19.4% of patients, with 12% undergoing HTx. There were no gender differences in arrhythmic outcomes, while 96.4% of patients who underwent HTx were male. In conclusion, RBM20 cardiomyopathy exhibits a severe phenotypic expression, both in terms of arrhythmic burden and HF progression.
Collapse
Affiliation(s)
- Marika Martini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.M.); (M.B.M.); (B.B.)
| | - Maria Bueno Marinas
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.M.); (M.B.M.); (B.B.)
| | | | - Kalliopi Pilichou
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.M.); (M.B.M.); (B.B.)
| | - Barbara Bauce
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.M.); (M.B.M.); (B.B.)
| |
Collapse
|
8
|
Voinescu OR, Ionac A, Sosdean R, Ionac I, Ana LS, Kundnani NR, Morariu S, Puiu M, Chirita-Emandi A. Genotype-Phenotype Insights of Inherited Cardiomyopathies-A Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:543. [PMID: 38674189 PMCID: PMC11052121 DOI: 10.3390/medicina60040543] [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: 02/23/2024] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
Background: Cardiomyopathies (CMs) represent a heterogeneous group of primary myocardial diseases characterized by structural and functional abnormalities. They represent one of the leading causes of cardiac transplantations and cardiac death in young individuals. Clinically they vary from asymptomatic to symptomatic heart failure, with a high risk of sudden cardiac death due to malignant arrhythmias. With the increasing availability of genetic testing, a significant number of affected people are found to have an underlying genetic etiology. However, the awareness of the benefits of incorporating genetic test results into the care of these patients is relatively low. Aim: The focus of this review is to summarize the current basis of genetic CMs, including the most encountered genes associated with the main types of cardiomyopathies: hypertrophic, dilated, restrictive arrhythmogenic, and non-compaction. Materials and Methods: For this narrative review, we performed a search of multiple electronic databases, to select and evaluate relevant manuscripts. Results: Advances in genetic diagnosis led to better diagnosis precision and prognosis prediction, especially with regard to the risk of developing arrhythmias in certain subtypes of cardiomyopathies. Conclusions: Implementing the genomic information to benefit future patient care, better risk stratification and management, promises a better future for genotype-based treatment.
Collapse
Affiliation(s)
- Oana Raluca Voinescu
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adina Ionac
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Centre of Timisoara Institute of Cardiovascular Diseases, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Institute for Cardiovascular Diseases, Gheorghe Adam Street 13A, 300310 Timisoara, Romania
| | - Raluca Sosdean
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Centre of Timisoara Institute of Cardiovascular Diseases, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Institute for Cardiovascular Diseases, Gheorghe Adam Street 13A, 300310 Timisoara, Romania
| | - Ioana Ionac
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Luca Silvia Ana
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Institute for Cardiovascular Diseases, Gheorghe Adam Street 13A, 300310 Timisoara, Romania
| | - Nilima Rajpal Kundnani
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Centre of Timisoara Institute of Cardiovascular Diseases, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Stelian Morariu
- General Medicine Faculty, “Vasile Goldis” West University, 473223 Arad, Romania
| | - Maria Puiu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy, “Victor Babeș” Eftimie Murgu Sq., 300041 Timisoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Adela Chirita-Emandi
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy, “Victor Babeș” Eftimie Murgu Sq., 300041 Timisoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| |
Collapse
|
9
|
Chia SPS, Pang JKS, Soh BS. Current RNA strategies in treating cardiovascular diseases. Mol Ther 2024; 32:580-608. [PMID: 38291757 PMCID: PMC10928165 DOI: 10.1016/j.ymthe.2024.01.028] [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: 09/14/2023] [Revised: 12/22/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Cardiovascular disease (CVD) continues to impose a significant global health burden, necessitating the exploration of innovative treatment strategies. Ribonucleic acid (RNA)-based therapeutics have emerged as a promising avenue to address the complex molecular mechanisms underlying CVD pathogenesis. We present a comprehensive review of the current state of RNA therapeutics in the context of CVD, focusing on the diverse modalities that bring about transient or permanent modifications by targeting the different stages of the molecular biology central dogma. Considering the immense potential of RNA therapeutics, we have identified common gene targets that could serve as potential interventions for prevalent Mendelian CVD caused by single gene mutations, as well as acquired CVDs developed over time due to various factors. These gene targets offer opportunities to develop RNA-based treatments tailored to specific genetic and molecular pathways, presenting a novel and precise approach to address the complex pathogenesis of both types of cardiovascular conditions. Additionally, we discuss the challenges and opportunities associated with delivery strategies to achieve targeted delivery of RNA therapeutics to the cardiovascular system. This review highlights the immense potential of RNA-based interventions as a novel and precise approach to combat CVD, paving the way for future advancements in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Shirley Pei Shan Chia
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Jeremy Kah Sheng Pang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Boon-Seng Soh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore.
| |
Collapse
|
10
|
Jolfayi AG, Kohansal E, Ghasemi S, Naderi N, Hesami M, MozafaryBazargany M, Moghadam MH, Fazelifar AF, Maleki M, Kalayinia S. Exploring TTN variants as genetic insights into cardiomyopathy pathogenesis and potential emerging clues to molecular mechanisms in cardiomyopathies. Sci Rep 2024; 14:5313. [PMID: 38438525 PMCID: PMC10912352 DOI: 10.1038/s41598-024-56154-7] [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: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
The giant protein titin (TTN) is a sarcomeric protein that forms the myofibrillar backbone for the components of the contractile machinery which plays a crucial role in muscle disorders and cardiomyopathies. Diagnosing TTN pathogenic variants has important implications for patient management and genetic counseling. Genetic testing for TTN variants can help identify individuals at risk for developing cardiomyopathies, allowing for early intervention and personalized treatment strategies. Furthermore, identifying TTN variants can inform prognosis and guide therapeutic decisions. Deciphering the intricate genotype-phenotype correlations between TTN variants and their pathologic traits in cardiomyopathies is imperative for gene-based diagnosis, risk assessment, and personalized clinical management. With the increasing use of next-generation sequencing (NGS), a high number of variants in the TTN gene have been detected in patients with cardiomyopathies. However, not all TTN variants detected in cardiomyopathy cohorts can be assumed to be disease-causing. The interpretation of TTN variants remains challenging due to high background population variation. This narrative review aimed to comprehensively summarize current evidence on TTN variants identified in published cardiomyopathy studies and determine which specific variants are likely pathogenic contributors to cardiomyopathy development.
Collapse
Affiliation(s)
- Amir Ghaffari Jolfayi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Kohansal
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hesami
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hosseini Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Voinescu OR, Ionescu BI, Militaru S, Afana AS, Sascau R, Vasiliu L, Onciul S, Dobrescu MA, Cozlac RA, Cozma D, Rancea R, Dragulescu B, Andreescu NI, Puiu M, Jurcut RO, Chirita-Emandi A. Genetic Characterization of Dilated Cardiomyopathy in Romanian Adult Patients. Int J Mol Sci 2024; 25:2562. [PMID: 38473809 DOI: 10.3390/ijms25052562] [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: 01/20/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Dilated cardiomyopathy (DCM) represents a group of disorders affecting the structure and function of the heart muscle, leading to a high risk of heart failure and sudden cardiac death (SCD). DCM frequently involves an underlying genetic etiology. Genetic testing is valuable for risk stratification, treatment decisions, and family screening. Romanian population data on the genetic etiology of DCM are lacking. We aimed to investigate the genetic causes for DCM among Romanian adult patients at tertiary referral centers across the country. Clinical and genetic investigations were performed on adult patients presenting to tertiary hospitals in Romania. The genetic investigations used next-generation sequencing panels of disease-associated DCM genes. A total of 122 patients with DCM underwent genetic testing. The mean age at DCM diagnosis was 41.6 ± 12.4 years. The genetic investigations identified pathogenic or likely pathogenic variants in 50.8% of participants, while 25.4% had variants of unknown significance. Disease-causing variants in 15 genes were identified in people with DCM, with 31 previously unreported variants. Variants in TTN, LMNA, and DSP explained 75% of genetic causes for DCM. In total, 52.4% of patients had a family history of DCM/SCD. Left ventricular ejection fraction of <35% was observed in 41.9% of patients with disease-causing variants and 55% with negative or uncertain findings. Further genotype-phenotype correlations were explored in this study population. The substantial percentage (50.8%) of disease-causing variants identified in patients with DCM acknowledges the importance of genetic investigations. This study highlights the genetic landscape in genes associated with DCM in the Romanian population.
Collapse
Affiliation(s)
- Oana Raluca Voinescu
- Department of Cardiology, Cardiology Discipline II, University of Medicine and Pharmacy "Victor Babeș", Eftimie Murgu Sq., 300041 Timișoara, Romania
| | - Bogdana Ioana Ionescu
- Department of Cardiology, University of Medicine and Pharmacy 'Carol Davila', Dionisie Lupu Street, no. 37, Sector 2, 4192910 Bucharest, Romania
- Expert Center for Rare Cardiac Genetic Diseases, Emergency Institute for Cardiovascular Diseases 'Prof.dr.C.C.Iliescu', Fundeni 258, 022328 Bucharest, Romania
| | - Sebastian Militaru
- Department of Cardiology, Craiova University of Medicine and Pharmacy, Petru Rareș Street no 2, 200349 Craiova, Romania
- Cardiomed Hospital, Craiova, Str. Spania, Nr. 35A, 200513 Craiova, Romania
| | - Andreea Sorina Afana
- Department of Cardiology, Craiova University of Medicine and Pharmacy, Petru Rareș Street no 2, 200349 Craiova, Romania
- Cardiomed Hospital, Craiova, Str. Spania, Nr. 35A, 200513 Craiova, Romania
| | - Radu Sascau
- Internal Medicine Department, "Grigore T. Popa" University of Medicine and Pharmacy, 16 Universitatii Street, 700503 Iași, Romania
- Cardiology Department, Cardiovascular Diseases Institute "Prof. Dr. George I. M. Georgescu", 50 Boulevard Carol I, 700503 Iași, Romania
| | - Laura Vasiliu
- Internal Medicine Department, "Grigore T. Popa" University of Medicine and Pharmacy, 16 Universitatii Street, 700503 Iași, Romania
- Cardiology Department, Cardiovascular Diseases Institute "Prof. Dr. George I. M. Georgescu", 50 Boulevard Carol I, 700503 Iași, Romania
| | - Sebastian Onciul
- Department of Cardiology, University of Medicine and Pharmacy 'Carol Davila', Dionisie Lupu Street, no. 37, Sector 2, 4192910 Bucharest, Romania
| | - Mihaela Amelia Dobrescu
- Genetics Department, Craiova University of Medicine and Pharmacy, Petru Rareș 2 Street, 200349 Craiova, Romania
- Regional Centre of Medical Genetics Dolj, Emergency County Hospital Craiova, 200642 Craiova, Romania
| | - Ramona Alina Cozlac
- Department of Cardiology, Cardiology Discipline II, University of Medicine and Pharmacy "Victor Babeș", Eftimie Murgu Sq., 300041 Timișoara, Romania
- Cardiology Department, Institute of Cardiovascular Diseases, Gheorghe Adam Street, 13A, 300310 Timișoara, Romania
| | - Dragos Cozma
- Department of Cardiology, Cardiology Discipline II, University of Medicine and Pharmacy "Victor Babeș", Eftimie Murgu Sq., 300041 Timișoara, Romania
- Cardiology Department, Institute of Cardiovascular Diseases, Gheorghe Adam Street, 13A, 300310 Timișoara, Romania
| | - Raluca Rancea
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001 Cluj-Napoca, Romania
| | - Bogdan Dragulescu
- Communications Department, Politehnica University Timisoara, sq Victoriei 2, 300006 Timișoara, Romania
| | - Nicoleta Ioana Andreescu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy "Victor Babeș" Timișoara, 2 Piaţa Eftimie Murgu Street, 300041 Timişoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children "Louis Țurcanu" Iosif Nemoianu Street N°2, 300011 Timișoara, Romania
| | - Maria Puiu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy "Victor Babeș" Timișoara, 2 Piaţa Eftimie Murgu Street, 300041 Timişoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children "Louis Țurcanu" Iosif Nemoianu Street N°2, 300011 Timișoara, Romania
| | - Ruxandra Oana Jurcut
- Department of Cardiology, University of Medicine and Pharmacy 'Carol Davila', Dionisie Lupu Street, no. 37, Sector 2, 4192910 Bucharest, Romania
- Expert Center for Rare Cardiac Genetic Diseases, Emergency Institute for Cardiovascular Diseases 'Prof.dr.C.C.Iliescu', Fundeni 258, 022328 Bucharest, Romania
| | - Adela Chirita-Emandi
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, University of Medicine and Pharmacy "Victor Babeș" Timișoara, 2 Piaţa Eftimie Murgu Street, 300041 Timişoara, Romania
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children "Louis Țurcanu" Iosif Nemoianu Street N°2, 300011 Timișoara, Romania
| |
Collapse
|
12
|
Gregorich ZR, Yanghai Z, Kamp TJ, Granzier H, Guo W. Mechanisms of RBM20 Cardiomyopathy: Insights From Model Systems. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004355. [PMID: 38288598 PMCID: PMC10923161 DOI: 10.1161/circgen.123.004355] [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] [Indexed: 02/05/2024]
Abstract
RBM20 (RNA-binding motif protein 20) is a vertebrate- and muscle-specific RNA-binding protein that belongs to the serine-arginine-rich family of splicing factors. The RBM20 gene was first identified as a dilated cardiomyopathy-linked gene over a decade ago. Early studies in Rbm20 knockout rodents implicated disrupted splicing of RBM20 target genes as a causative mechanism. Clinical studies show that pathogenic variants in RBM20 are linked to aggressive dilated cardiomyopathy with early onset heart failure and high mortality. Subsequent studies employing pathogenic variant knock-in animal models revealed that variants in a specific portion of the arginine-serine-rich domain in RBM20 not only disrupt splicing but also hinder nucleocytoplasmic transport and lead to the formation of RBM20 biomolecular condensates in the sarcoplasm. Conversely, mice harboring a disease-associated variant in the RRM (RNA recognition motif) do not show evidence of adverse remodeling or exhibit sudden death despite disrupted splicing of RBM20 target genes. Thus, whether disrupted splicing, biomolecular condensates, or both contribute to dilated cardiomyopathy is under debate. Beyond this, additional questions remain, such as whether there is sexual dimorphism in the presentation of RBM20 cardiomyopathy. What are the clinical features of RBM20 cardiomyopathy and why do some individuals develop more severe disease than others? In this review, we summarize the reported observations and discuss potential mechanisms of RBM20 cardiomyopathy derived from studies employing in vivo animal models and in vitro human-induced pluripotent stem cell-derived cardiomyocytes. Potential therapeutic strategies to treat RBM20 cardiomyopathy are also discussed.
Collapse
Affiliation(s)
- Zachery R. Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
| | - Zhang Yanghai
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
| | - Timothy J. Kamp
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
13
|
DePaolo J, Bornstein M, Judy R, Abramowitz S, Verma SS, Levin MG, Arany Z, Damrauer SM. Titin-Truncating variants Predispose to Dilated Cardiomyopathy in Diverse Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.17.24301405. [PMID: 38293092 PMCID: PMC10827233 DOI: 10.1101/2024.01.17.24301405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Importance The effect of high percentage spliced in (hiPSI) TTN truncating variants (TTNtvs) on risk of dilated cardiomyopathy (DCM) has historically been studied among population subgroups defined by genetic similarity to European reference populations. This has raised questions about the effect of TTNtvs in diverse populations, especially among individuals genetically similar to African reference populations. Objective To determine the effect of TTNtvs on risk of DCM in diverse population as measured by genetic distance (GD) in principal component (PC) space. Design Cohort study. Setting Penn Medicine Biobank (PMBB) is a large, diverse biobank. Participants Participants were recruited from across the Penn Medicine healthcare system and volunteered to have their electronic health records linked to biospecimen data including DNA which has undergone whole exome sequencing. Main Outcomes and Measures Risk of DCM among individuals carrying a hiPSI TTNtv. Results Carrying a hiPSI TTNtv was associated with DCM among PMBB participants across a range of GD deciles from the 1000G European centroid; the effect estimates ranged from odds ratio (OR) = 3.29 (95% confidence interval [CI] 1.26 to 8.56) to OR = 9.39 (95% CI 3.82 to 23.13). When individuals were assigned to population subgroups based on genetic similarity to the 1000G reference populations, hiPSI TTNtvs conferred significant risk of DCM among those genetically similar to the 1000G European reference population (OR = 7.55, 95% CI 4.99 to 11.42, P<0.001) and individuals genetically similar to the 1000G African reference population (OR 3.50, 95% CI 1.48 to 8.24, P=0.004). Conclusions and Relevance TTNtvs are associated with increased risk of DCM among a diverse cohort. There is no significant difference in effect of TTNtvs on DCM risk across deciles of GD from the 1000G European centroid, suggesting genetic background should not be considered when screening individuals for titin-related DCM.
Collapse
Affiliation(s)
- John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc Bornstein
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Renae Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Abramowitz
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Michael G Levin
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Zoltan Arany
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Tan K, Foo R, Loh M. Cardiomyopathy in Asian Cohorts: Genetic and Epigenetic Insights. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:496-506. [PMID: 37589150 DOI: 10.1161/circgen.123.004079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Previous studies on cardiomyopathies have been particularly valuable for clarifying pathological mechanisms in heart failure, an etiologically heterogeneous disease. In this review, we specifically focus on cardiomyopathies in Asia, where heart failure is particularly pertinent. There has been an increase in prevalence of cardiomyopathies in Asia, in sharp contrast with the decline observed in Western countries. Indeed, important disparities in cardiomyopathy incidence, clinical characteristics, and prognosis have been reported in Asian versus White cohorts. These have been accompanied by emerging descriptions of a distinct rare and common genetic basis for disease among Asian cardiomyopathy patients marked by an increased burden of variants with uncertain significance, reclassification of variants deemed pathogenic based on evidence from predominantly White cohorts, and the discovery of Asian-specific cardiomyopathy-associated loci with underappreciated pathogenicity under conventional classification criteria. Findings from epigenetic studies of heart failure, particularly DNA methylation studies, have complemented genetic findings in accounting for the phenotypic variability in cardiomyopathy. Though extremely limited, findings from Asian ancestry-focused DNA methylation studies of cardiomyopathy have shown potential to contribute to general understanding of cardiomyopathy pathophysiology by proposing disease and cause-relevant pathophysiological mechanisms. We discuss the value of multiomics study designs incorporating genetic, methylation, and transcriptomic information for future DNA methylation studies in Asian cardiomyopathy cohorts to yield Asian ancestry-specific insights that will improve risk stratification in the Asian population.
Collapse
Affiliation(s)
- Konstanze Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore (R.F.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore (R.F.)
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
- Genome Institute of Singapore, Singapore (GIS), Agency for Science, Technology and Research (A*STAR) (M.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (M.L.)
- National Skin Centre, Singapore (M.L.)
| |
Collapse
|
15
|
Alaei Z, Zamani N, Rabbani B, Mahdieh N. TCAP gene is not a common cause of cardiomyopathy in Iranian patients. Eur J Med Res 2023; 28:376. [PMID: 37752589 PMCID: PMC10523715 DOI: 10.1186/s40001-023-01019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most frequent cardiomyopathies that cause acute heart failure and sudden cardiac death. Previous genetic reports have shown that pathogenic variants of genes encoding Z-disc components such as telethonin protein (TCAP) are the primary cause of DCM and HCM. METHODS This study was the first investigation on the TCAP gene among the Iranian cardiomyopathies population wherein the TCAP gene was analyzed in 40 unrelated patients (17 females and 23 males) who were clinically diagnosed with HCM and DCM. In addition, we conducted a thorough review of all published articles and the databases that were the first to report novel pathogenic or likely pathogenic variants the in TCAP gene. RESULTS In the cohort of this study, we identified only one intronic variant c.111-42G > A in one of the HCM patients that were predicted as polymorphism by in-silico analysis. Moreover, a total of 44 variants were reported for the TCAP gene in the literature where a majority of mutations were found to be missense. Pathogenic mutations in TCAP may cause diseases including limb-girdle muscular dystrophy 2G (LGMD-2G), DCM, HCM, intestinal pseudo-obstruction, and telethonin deficiency. However, a large number of affected patients were clinically diagnosed with limb-girdle 2G compared to other presenting phenotypes. DISCUSSION These findings suggest that the TCAP gene pathogenic mutations might not be a common cause of cardiomyopathies among Iranian patients. These gene disease-causing mutations may cause various manifestations, but it has a high prevalence among LGMD-2G, HCM, and DCM patients.
Collapse
Affiliation(s)
- Zahra Alaei
- Faculty of Basic Sciences, Islamic Azad University, East Tehran Branch, Tehran, Iran
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Zamani
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Rabbani
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nejat Mahdieh
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Genetics Laboratory, Rajaie Cardiovascular Medical and Research Center, Vali-E-Asr Avenue, Tehran, 1996911151, Iran.
| |
Collapse
|
16
|
Heliö K, Cicerchia M, Hathaway J, Tommiska J, Huusko J, Saarinen I, Koskinen L, Muona M, Kytölä V, Djupsjöbacka J, Gentile M, Salmenperä P, Alastalo TP, Steinberg C, Heliö T, Paananen J, Myllykangas S, Koskenvuo J. Diagnostic yield of genetic testing in a multinational heterogeneous cohort of 2088 DCM patients. Front Cardiovasc Med 2023; 10:1254272. [PMID: 37795486 PMCID: PMC10546047 DOI: 10.3389/fcvm.2023.1254272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Background Familial dilated cardiomyopathy (DCM) causes heart failure and may lead to heart transplantation. DCM is typically a monogenic disorder with autosomal dominant inheritance. Currently disease-causing variants have been reported in over 60 genes that encode proteins in sarcomeres, nuclear lamina, desmosomes, cytoskeleton, and mitochondria. Over half of the patients undergoing comprehensive genetic testing are left without a molecular diagnosis even when patient selection follows strict DCM criteria. Methods and results This study was a retrospective review of patients referred for genetic testing at Blueprint Genetics due to suspected inherited DCM. Next generation sequencing panels included 23-316 genes associated with cardiomyopathies and other monogenic cardiac diseases. Variants were considered diagnostic if classified as pathogenic (P) or likely pathogenic (LP). Of the 2,088 patients 514 (24.6%) obtained a molecular diagnosis; 534 LP/P variants were observed across 45 genes, 2.7% (14/514) had two diagnostic variants in dominant genes. Nine copy number variants were identified: two multigene and seven intragenic. Diagnostic variants were observed most often in TTN (45.3%), DSP (6.7%), LMNA (6.7%), and MYH7 (5.2%). Clinical characteristics independently associated with molecular diagnosis were: a lower age at diagnosis, family history of DCM, paroxysmal atrial fibrillation, absence of left bundle branch block, and the presence of an implantable cardioverter-defibrillator. Conclusions Panel testing provides good diagnostic yield in patients with clinically suspected DCM. Causative variants were identified in 45 genes. In minority, two diagnostic variants were observed in dominant genes. Our results support the use of genetic panels in clinical settings in DCM patients with suspected genetic etiology.
Collapse
Affiliation(s)
- Krista Heliö
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Julie Hathaway
- Blueprint Genetics, A Quest Diagnostics Company, Seattle, USA
| | | | - Johanna Huusko
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Inka Saarinen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Lotta Koskinen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Mikko Muona
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Ville Kytölä
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | | | | | | | | | | | - Tiina Heliö
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Jussi Paananen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | | | - Juha Koskenvuo
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| |
Collapse
|
17
|
Lian H, Song S, Chen W, Shi A, Jiang H, Hu S. Genetic characterization of dilated cardiomyopathy patients undergoing heart transplantation in the Chinese population by whole-exome sequencing. J Transl Med 2023; 21:476. [PMID: 37461109 PMCID: PMC10351148 DOI: 10.1186/s12967-023-04282-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/17/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is one of the most frequent causes of heart failure and heart transplantation (HTx). The genetic basis of DCM among patients undergoing HTx remains to be further studied. This study aimed to characterize the genetic basis of DCM HTx in the Chinese population. METHODS In total, 208 unrelated DCM patients who underwent HTx at Fuwai Hospital between June 2004 and June 2017 were included in this study. Whole-exome sequencing (WES) was performed for all patients. Gene burden analysis, variant classification, and genotype-phenotype correlation analysis were subsequently performed. RESULTS After completing the bioinformatics analysis, gene burden analysis suggested that titin (TTN), filamin C (FLNC) and lamin A/C (LMNA) were significantly enriched with rare protein-altering variants. The frequencies of TTN and FLNC truncating variants in our cohort were 18.8% and 8.7%, respectively. Among the 165 rare variants in high evidence DCM-related genes, 27 (16.4%) and 59 (35.8%) were interpreted as pathogenic (P) and likely pathogenic (LP), respectively. In addition, 41 (47.7%) and 16 (18.6%) of these 86 P/LP variants are located in TTN and FLNC, respectively. The FLNC group contained more patients with NYHA class IV than the P/LP-negative group (FLNC, 16/18 vs. P/LP-negative, 81/123, P = 0.049). CONCLUSIONS Based on WES, we provided a primary genetic spectrum of DCM patients undergoing HTx in the Chinese population. TTN and FLNC harbour the most P/LP variants. FLNC truncation may lead to severe clinical symptoms in DCM patients.
Collapse
Affiliation(s)
- Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wenzheng Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Anteng Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Haobin Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
18
|
Hu X, Kong J, Niu T, Chen L, Yang J. Single coronary artery presenting dilated cardiomyopathy and hyperlipidemia with the SCN5A and APOA5 gene mutation: A case report and review of the literature. Front Cardiovasc Med 2023; 10:1113886. [PMID: 37288251 PMCID: PMC10242075 DOI: 10.3389/fcvm.2023.1113886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/23/2023] [Indexed: 06/09/2023] Open
Abstract
We present a 55-year-old man with chest tightness and dyspnoea after activity lasting for 2 months who was diagnosed with single coronary artery (SCA) and presented with dilated cardiomyopathy (DCM) with the c.1858C > T mutation in the SCN5A gene. The computed tomography coronary angiogram (CTCA) showed congenital absence of the right coronary artery (RCA), and the right heart was nourished by the left coronary artery branch with no apparent stenosis. Transthoracic echocardiography (TTE) revealed enlargement of the left heart and cardiomyopathy. Cardiac magnetic resonance imaging (CMR) revealed DCM. Genetic testing showed that the c.1858C > T variant of the SCN5A gene could lead to Brugada syndrome and DCM. SCA is a rare congenital anomaly of the coronary anatomy, and this case reported as SCA accompanied by DCM is even rarer. We present a rare case of a 55-year-old man with DCM with the c.1858C > T (p. Arg620Cys)/c.1008G > A (p.(Pro336=) variant of the SCN5A gene, congenital absence of RCA, and c.990_993delAACA (p. Asp332Valfs*5) variant of the APOA5 gene. To our knowledge, this is the first report of DCM combined with the SCN5A gene mutation in SCA after searching the PubMed, CNKI and Wanfang databases.
Collapse
Affiliation(s)
- Xiaoxia Hu
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Kong
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tingting Niu
- Department of Medical Technology, Jinan Vocational College of Nursing, Jinan, Shandong, China
| | - Liang Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jingjing Yang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
19
|
Heliö K, Brandt E, Vaara S, Weckström S, Harjama L, Kandolin R, Järviö J, Hannula-Jouppi K, Heliö T, Holmström M, Koskenvuo JW. DSP c.6310delA p.(Thr2104Glnfs*12) associates with arrhythmogenic cardiomyopathy, increased trabeculation, curly hair, and palmoplantar keratoderma. Front Cardiovasc Med 2023; 10:1130903. [PMID: 37008330 PMCID: PMC10050721 DOI: 10.3389/fcvm.2023.1130903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundPathogenic variants in DSP associate with cardiac and cutaneous manifestations including arrhythmogenic right ventricular cardiomyopathy, dilated cardiomyopathy, curly or wavy hair, and palmoplantar keratoderma (PPK). Episodes of myocardial inflammation associated with DSP cardiomyopathy might be confused in clinical work with myocarditis of other etiologies such as viral. Cardiac magnetic resonance imaging (CMR) may help in differential diagnosis.Methods and resultsThis study comprised 49 Finnish patients: 34 participants from families with suspected DSP cardiomyopathy (9 index patients and 25 family members) and 15 patients with myocarditis. All 34 participants underwent genetic testing and cardiac evaluation, and 29 of them also underwent CMR. Participants with the DSP variant, numbering 22, were dermatologically examined. The 15 patients with myocarditis underwent CMR and were evaluated during their hospitalization.A heterozygous truncating DSP c.6310delA p.(Thr2104Glnfs*12) variant was confirmed in 29 participants. Only participants with the DSP variant had pacemakers and life-threatening ventricular arrhythmias. Of the participants with the DSP variant, 24% fulfilled cardiomyopathy criteria, and the median age at diagnosis was 53. Upon CMR, myocardial edema was found to be more common in patients with myocarditis. Both groups had a substantial percentage of late gadolinium enhancement (LGE). A ring-like LGE and increased trabeculation were observed only in participants with the DSP variant. All the studied participants with the DSP variant had PPK and curly or wavy hair. Hyperkeratosis developed before the age of 20 in most patients.ConclusionsThe DSP c.6310delA p.(Thr2104Glnfs*12) variant associates with curly hair, PPK, and arrhythmogenic cardiomyopathy with increased trabeculation. Cutaneous symptoms developing in childhood and adolescence might help recognize these patients at an earlier stage. CMR, together with dermatologic characteristics, may help in diagnosis.
Collapse
Affiliation(s)
- Krista Heliö
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Eveliina Brandt
- Department of Dermatology and Allergology, ERN-Skin Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Satu Vaara
- Radiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Sini Weckström
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Liisa Harjama
- Department of Dermatology and Allergology, ERN-Skin Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Riina Kandolin
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Johanna Järviö
- Radiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Katariina Hannula-Jouppi
- Department of Dermatology and Allergology, ERN-Skin Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland and Research Programs Unit, Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Tiina Heliö
- Heart and Lung Center, ERN GUARD-Heart Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Miia Holmström
- Radiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Juha W. Koskenvuo
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
20
|
Pan-Lizcano R, Mariñas-Pardo L, Núñez L, Rebollal-Leal F, López-Vázquez D, Pereira A, Molina-Nieto A, Calviño R, Vázquez-Rodríguez JM, Hermida-Prieto M. Rare Variants in Genes of the Cholesterol Pathway Are Present in 60% of Patients with Acute Myocardial Infarction. Int J Mol Sci 2022; 23:ijms232416127. [PMID: 36555767 PMCID: PMC9786046 DOI: 10.3390/ijms232416127] [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: 11/04/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Acute myocardial infarction (AMI) is a pandemic in which conventional risk factors are inadequate to detect who is at risk early in the asymptomatic stage. Although gene variants in genes related to cholesterol, which may increase the risk of AMI, have been identified, no studies have systematically screened the genes involved in this pathway. In this study, we included 105 patients diagnosed with AMI with an elevation of the ST segment (STEMI) and treated with primary percutaneous coronary intervention (PPCI). Using next-generation sequencing, we examined the presence of rare variants in 40 genes proposed to be involved in lipid metabolism and we found that 60% of AMI patients had a rare variant in the genes involved in the cholesterol pathway. Our data show the importance of considering the wide scope of the cholesterol pathway in order to assess the genetic risk related to AMI.
Collapse
Affiliation(s)
- Ricardo Pan-Lizcano
- Grupo de Investigación en Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), GRINCAR-Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Luis Mariñas-Pardo
- Facultad de Ciencias de la Salud, Universidad Internacional de Valencia (VIU), 46002 Valencia, Spain
| | - Lucía Núñez
- Grupo de Investigación en Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), GRINCAR-Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Departamento de Ciencias de la Salud, GRINCAR Research Group, Universidade da Coruña, 15403 A Coruña, Spain
- Correspondence: ; Tel.: +34-981-178-150
| | - Fernando Rebollal-Leal
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Domingo López-Vázquez
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Ana Pereira
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Aranzazu Molina-Nieto
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Ramón Calviño
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- CIBERCV (Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose Manuel Vázquez-Rodríguez
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- CIBERCV (Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Hermida-Prieto
- Grupo de Investigación en Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC-SERGAS), GRINCAR-Universidade da Coruña (UDC), 15006 A Coruña, Spain
| |
Collapse
|
21
|
Ranta-Aho J, Olive M, Vandroux M, Roticiani G, Dominguez C, Johari M, Torella A, Böhm J, Turon J, Nigro V, Hackman P, Laporte J, Udd B, Savarese M. Mutation update for the ACTN2 gene. Hum Mutat 2022; 43:1745-1756. [PMID: 36116040 PMCID: PMC10087778 DOI: 10.1002/humu.24470] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/29/2022] [Accepted: 09/12/2022] [Indexed: 01/24/2023]
Abstract
ACTN2 encodes alpha-actinin-2, a protein expressed in human cardiac and skeletal muscle. The protein, located in the sarcomere Z-disk, functions as a link between the anti-parallel actin filaments. This important structural protein also binds N-terminal titins, and thus contributes to sarcomere stability. Previously, ACTN2 mutations have been solely associated with cardiomyopathy, without skeletal muscle disease. Recently, however, ACTN2 mutations have been associated with novel congenital and distal myopathy. Previously reported variants are in varying locations across the gene, but the potential clustering effect of pathogenic locations is not clearly understood. Further, the genotype-phenotype correlations of these variants remain unclear. Here we review the previously reported ACTN2-related molecular and clinical findings and present an additional variant, c.1840-2A>T, that further expands the mutation and phenotypic spectrum. Our results show a growing body of clinical, genetic, and functional evidence, which underlines the central role of ACTN2 in the muscle tissue and myopathy. However, limited segregation and functional data are available to support the pathogenicity of most previously reported missense variants and clear-cut genotype-phenotype correlations are currently only demonstrated for some ACTN2-related myopathies.
Collapse
Affiliation(s)
- Johanna Ranta-Aho
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Montse Olive
- Department of Neurology, Neuromuscular Diseases Unit, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marie Vandroux
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Université de Strasbourg, Illkirch, France
| | | | - Cristina Dominguez
- Department of Neurology, Neuromuscular Unit, Hospital Universitario 12 de Octubre, Research Institute imas12, Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Mridul Johari
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Johann Böhm
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Université de Strasbourg, Illkirch, France
| | - Janina Turon
- Department of Neurology, Neuromuscular Diseases Unit, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Peter Hackman
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Jocelyn Laporte
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Université de Strasbourg, Illkirch, France
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| | - Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Vissing CR, Espersen K, Mills HL, Bartels ED, Jurlander R, Skriver SV, Ghouse J, Thune JJ, Axelsson Raja A, Christensen AH, Bundgaard H. Family Screening in Dilated Cardiomyopathy: Prevalence, Incidence, and Potential for Limiting Follow-Up. JACC. HEART FAILURE 2022; 10:792-803. [PMID: 36328645 DOI: 10.1016/j.jchf.2022.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND According to patterns of inheritance and incomplete penetrance, fewer than half of relatives to dilated cardiomyopathy probands will develop disease. OBJECTIVES The purpose of this study was to investigate the prevalence and incidence, and to identify predictors of developing familial dilated cardiomyopathy (FDC) in relatives participating in family screening. METHODS The study was a retrospective, longitudinal cohort study of families screened and followed from 2006 to 2020 at a regional assembly of clinics for inherited cardiomyopathies. RESULTS In total, 211 families (563 relatives, 50% women) were included. At baseline, 124 relatives (22%) were diagnosed with FDC. Genetic sequencing identified the etiology in 37% of screened families and classified 101 (18%) relatives as unaffected carriers (n = 43) or noncarriers (ie, not at risk of FDC [n = 58]). The combined clinical and genetic baseline yield was 30%. During follow-up (2,313 person-years, median 5.0 years), 45 developed FDC (incidence rate of 2.0% per person-year; 95% CI: 1.4%-2.8%), increasing the overall yield to 34%. The incidence rate of FDC was high in relatives with baseline abnormalities on electrocardiogram or echocardiography compared with relatives with normal findings (4.7% vs 0.4% per person-year; HR: 12.9; P < 0.001). In total, baseline screening identified 326 (58%) relatives to be at low risk of FDC. CONCLUSIONS Family screening identified a genetic predisposition to or overt FDC in 1 of 3 relatives at baseline. Genetic and clinical screening was normal in more than half of relatives, and these relatives had a low risk of developing FDC during follow-up. Thus, baseline screening identified a large proportion, in whom follow-up may safely be reduced, allowing focused follow-up of relatives at risk.
Collapse
Affiliation(s)
- Christoffer R Vissing
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Kiri Espersen
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helen L Mills
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil D Bartels
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Jurlander
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sofie V Skriver
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Copenhagen Neuromuscular Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jonas Ghouse
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Thune
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Cardiology, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anna Axelsson Raja
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alex H Christensen
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henning Bundgaard
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Yang Q, Berkman AM, Ezekian JE, Rosamilia M, Rosenfeld JA, Liu P, Landstrom AP. Determining the Likelihood of Disease Pathogenicity Among Incidentally Identified Genetic Variants in Rare Dilated Cardiomyopathy-Associated Genes. J Am Heart Assoc 2022; 11:e025257. [PMID: 36129056 DOI: 10.1161/jaha.122.025257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background As utilization of clinical exome sequencing (ES) has expanded, criteria for evaluating the diagnostic weight of incidentally identified variants are critical to guide clinicians and researchers. This is particularly important in genes associated with dilated cardiomyopathy (DCM), which can cause heart failure and sudden death. We sought to compare the frequency and distribution of incidentally identified variants in DCM-associated genes between a clinical referral cohort with those in control and known case cohorts to determine the likelihood of pathogenicity among those undergoing genetic testing for non-DCM indications. Methods and Results A total of 39 rare, non-TTN DCM-associated genes were identified and evaluated from a clinical ES testing referral cohort (n=14 005, Baylor Genetic Laboratories) and compared with a DCM case cohort (n=9442) as well as a control cohort of population variants (n=141 456) derived from the gnomAD database. Variant frequencies in each cohort were compared. Signal-to-noise ratios were calculated comparing the DCM and ES cohort with the gnomAD cohort. The likely pathogenic/pathogenic variant yield in the DCM cohort (8.2%) was significantly higher than in the ES cohort (1.9%). Based on signal-to-noise and correlation analysis, incidental variants found in FLNC, RBM20, MYH6, DSP, ABCC9, JPH2, and NEXN had the greatest chance of being DCM-associated. Conclusions The distribution of pathogenic variants between the ES cohort and the DCM case cohort was gene specific, and variants found in the ES cohort were similar to variants found in the control cohort. Incidentally identified variants in specific genes are more associated with DCM than others.
Collapse
Affiliation(s)
- Qixin Yang
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC.,Department of Cardiology The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou China
| | - Amy M Berkman
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Jordan E Ezekian
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Michael Rosamilia
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics Baylor College of Medicine and Baylor Genetics Laboratories Houston TX
| | - Pengfei Liu
- Department of Molecular and Human Genetics Baylor College of Medicine and Baylor Genetics Laboratories Houston TX
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
24
|
Yoneda ZT, Anderson KC, Ye F, Quintana JA, O’Neill MJ, Sims RA, Sun L, Glazer AM, Davogustto G, El-Harasis M, Laws JL, Saldivar BN, Crawford DM, Stricker T, Wells Q, Darbar D, Michaud GF, Stevenson LW, Lubitz SA, Ellinor PT, Roden DM, Shoemaker MB. Mortality Among Patients With Early-Onset Atrial Fibrillation and Rare Variants in Cardiomyopathy and Arrhythmia Genes. JAMA Cardiol 2022; 7:733-741. [PMID: 35544069 PMCID: PMC9096694 DOI: 10.1001/jamacardio.2022.0810] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/24/2022] [Indexed: 12/03/2022]
Abstract
Importance Patients with early-onset atrial fibrillation (AF) are enriched for rare variants in cardiomyopathy and arrhythmia genes. The clinical significance of these rare variants in patients with early-onset AF is unknown. Objective To assess the association between rare variants in cardiomyopathy and arrhythmia genes detected in patients with early-onset AF and time to death. Design, Setting, and Participants This prospective cohort study included participants with AF diagnosed before 66 years of age who underwent whole-genome sequencing through the National Heart, Lung and Blood Institute's Trans-Omics for Precision Medicine program. Participants were enrolled from November 23, 1999, to June 2, 2015. Data were analyzed from February 26 to September 19, 2021. Exposures Rare variants identified in a panel of 145 genes that are included in cardiomyopathy and arrhythmia panels used by commercial clinical genetic testing laboratories. Main Outcomes and Measures The primary study outcome was time to death and was adjudicated from medical records and the National Death Index. Multivariable Cox proportional hazards regression was used to evaluate the association of disease-associated variants with risk of death after adjustment for age at AF diagnosis, sex, race, body mass index, left ventricular ejection fraction, and an interaction term of age at AF diagnosis and disease-associated variant status. Results Among 1293 participants (934 [72%] male; median age at enrollment, 56.0 years; IQR, 48.0-61.0 years), disease-associated (pathogenic or likely pathogenic) rare variants were found in 131 (10%). During a median follow-up of 9.9 years (IQR, 6.9-13.2 years), 219 participants (17%) died. In univariable analysis, disease-associated variants were associated with an increased risk of mortality (hazard ratio, [HR], 1.5; 95% CI, 1.0-2.1; P = .05); the association remained significant in multivariable modeling when adjusted for age at AF diagnosis, sex, race, body mass index, left ventricular ejection fraction, and an interaction term between disease-associated variant status and age at AF diagnosis. The interaction demonstrated that disease-associated variants were associated with a significantly higher risk of mortality compared with no disease-associated variant when AF was diagnosed at a younger age (P = .008 for interaction). Higher body mass index (per IQR: HR, 1.4; 95% CI, 1.2-1.6; P < .001) and lower left ventricular ejection fraction (per IQR: HR, 0.8; 95% CI, 0.7-0.8; P < .001) were associated with higher mortality risk. There were 73 cardiomyopathy-related deaths, 40 sudden deaths, and 10 stroke-related deaths. Mortality among patients with the most prevalent genes with disease-associated variants was 26% (10 of 38 patients) for TTN, 33% (6 of 18) for MYH7, 22% (2 of 9) for LMNA, 0% (0 of 10) for MYH6, and 0% (0 of 8) for KCNQ1. Conclusions and Relevance The findings suggest that rare variants in cardiomyopathy and arrhythmia genes may be associated with increased risk of mortality among patients with early-onset AF, especially those diagnosed at a younger age. Genetic testing may provide important prognostic information for patients with early-onset AF.
Collapse
Affiliation(s)
- Zachary T. Yoneda
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Katherine C. Anderson
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fei Ye
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph A. Quintana
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Richard A. Sims
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lili Sun
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Andrew M. Glazer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Giovanni Davogustto
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Majd El-Harasis
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James L. Laws
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brittany N. Saldivar
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Diane M. Crawford
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Thomas Stricker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Quinn Wells
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dawood Darbar
- Department of Medicine, Division of Cardiology, University of Illinois at Chicago
| | - Gregory F. Michaud
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne W. Stevenson
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Steven A. Lubitz
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center & Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center & Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston
| | - Dan M. Roden
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M. Benjamin Shoemaker
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
25
|
Castelletti S, Zorzi A, Ballardini E, Basso C, Biffi A, Bracati F, Cavarretta E, Crotti L, Contursi M, D'Aleo A, D'Ascenzi F, Delise P, Dello Russo A, Gazale G, Mos L, Novelli V, Palamà Z, Palermi S, Palmieri V, Patrizi G, Pelliccia A, Pilichou K, Romano S, Sarto P, Schwartz PJ, Tiberi M, Zeppilli P, Corrado D, Sciarra L. Molecular genetic testing in athletes: Why and when a position statement from the Italian society of sports cardiology. Int J Cardiol 2022; 364:169-177. [PMID: 35662561 DOI: 10.1016/j.ijcard.2022.05.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 12/31/2022]
Abstract
Molecular genetic testing is an increasingly available test to support the clinical diagnosis of inherited cardiovascular diseases through identification of pathogenic gene variants and to make a preclinical genetic diagnosis among proband's family members (so-called "cascade family screening"). In athletes, the added value of molecular genetic testing is to assist in discriminating between physiological adaptive changes of the athlete's heart and inherited cardiovascular diseases, in the presence of overlapping phenotypic features such as ECG changes, imaging abnormalities or arrhythmias ("grey zone"). Additional benefits of molecular genetic testing in the athlete include the potential impact on the disease risk stratification and the implications for eligibility to competitive sports. This position statement of the Italian Society of Sports Cardiology aims to guide general sports medical physicians and sports cardiologists on clinical decision as why and when to perform a molecular genetic testing in the athlete, highlighting strengths and weaknesses for each inherited cardiovascular disease at-risk of sudden cardiac death during sport. The importance of early (preclinical) diagnosis to prevent the negative effects of exercise on phenotypic expression, disease progression and worsening of the arrhythmogenic substrate is also addressed.
Collapse
Affiliation(s)
- Silvia Castelletti
- Cardiomyopathy Center and Rehabilitation Unit, Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Alessandro Zorzi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Enrico Ballardini
- Sports Medicine Centre, Gruppo Mantova Salus, Ospedale San Pellegrino, Mantova, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alessandro Biffi
- Med-Ex, Medicine and Exercise srl, Medical Partner Scuderia Ferrari, Rome, Italy
| | - Francesco Bracati
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Aquila, Italy
| | - Elena Cavarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Naples, Italy
| | - Lia Crotti
- Cardiomyopathy Center and Rehabilitation Unit, Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy; Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Maurizio Contursi
- Sports Cardiology Unit, Centro Polidiagnostico Check-up, Salerno, Italy
| | | | - Flavio D'Ascenzi
- Division of Cardiology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Pietro Delise
- Division of Cardiology, Hospital of Peschiera del Garda, Veneto, Italy
| | - Antonio Dello Russo
- Cardiology and Arrhythmology Clinic, University Hospital "Lancisi-Umberto I- Salesi", Ancona, Italy, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Giovanni Gazale
- Center of Sport Medicine and Sports Cardiology, ASL 1, Sassari, Italy
| | - Lucio Mos
- San Antonio Hospital, San Daniele del Friuli, Udine, Italy
| | | | - Zefferino Palamà
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Aquila, Italy; Casa di Cura Villa Verde, Taranto, Italy
| | - Stefano Palermi
- Med-Ex, Medicine and Exercise srl, Medical Partner Scuderia Ferrari, Rome, Italy
| | - Vincenzo Palmieri
- Sports Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | - Kalliopi Pilichou
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Silvio Romano
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Aquila, Italy
| | | | - Peter J Schwartz
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Monica Tiberi
- Department of Public Health, Azienda Sanitaria Unica Regionale Marche AV 1, Pesaro, Italy
| | - Paolo Zeppilli
- Sports Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Domenico Corrado
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| | - Luigi Sciarra
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Aquila, Italy
| |
Collapse
|
26
|
Malakootian M, Bagheri Moghaddam M, Kalayinia S, Farrashi M, Maleki M, Sadeghipour P, Amin A. Dilated cardiomyopathy caused by a pathogenic nucleotide variant in RBM20 in an Iranian family. BMC Med Genomics 2022; 15:106. [PMID: 35527250 PMCID: PMC9079971 DOI: 10.1186/s12920-022-01262-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/05/2022] [Indexed: 12/22/2022] Open
Abstract
Abstract
Introduction
Dilated cardiomyopathy (DCM) is characterized by the dilation and impaired contraction of 1 or both ventricles and can be caused by a variety of disorders. Up to 50% of idiopathic DCM cases have heritable familial diseases, and the clinical screening of family members is recommended. Identifying a genetic cause that can explain the DCM risk in the family can help with better screening planning and clinical decision-making. Whole-exome sequencing (WES) has aided significantly in the detection of causative genes in many genetically heterogeneous diseases. In the present study, we applied WES to identify the causative genetic variant in a family with heritable DCM.
Methods
WES was applied to identify genetic variants on a 26-year-old man as the proband of a family with DCM. Subsequently, Sanger sequencing was performed to confirm the variant in the patient and all the available affected and unaffected family members. The pathogenicity of the variant was evaluated through co-segregation analysis in the family and employment of in silico predictive software.
Results
WES demonstrated the missense pathogenic heterozygous nucleotide variant, c.1907G > A, (p.Arg636His, rs267607004, NM_0011343), in exon 9 of the RBM20 gene in the proband. The variant was co-segregated in all the affected family members in a heterozygous form and the unaffected family members. The in silico analysis confirmed the variant as pathogenic.
Conclusion
Pathogenic RBM20 nucleotide variants are associated with arrhythmogenic DCM. We believe that our report is the first to show an RBM20 variant in Iranian descent associated with DCM.
Collapse
|
27
|
Huang W, Xu R, Gao N, Wu X, Wen C. Case Report: Family Curse: An SCN5A Mutation, c.611C>A, p.A204E Associated With a Family History of Dilated Cardiomyopathy and Arrhythmia. Front Cardiovasc Med 2022; 9:822150. [PMID: 35600473 PMCID: PMC9120596 DOI: 10.3389/fcvm.2022.822150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Objective We report a 3-generation family with SCN5A c.611 C>A rare variant, whose clinical characteristics are dilated cardiomyopathy (DCM) combined with multifocal ectopic Purkinje-related premature contractions (MEPPC). We tried to explain why the same SCN5A variant carriers had different phenotypes. Methods We collected the clinical data from the family, and followed up this family members. Genetic testing was done for whom DNA samples could be collected. Results Information was collected from 15 people in this family, 8 of whom had genetic testing. The SCN5A variant was present in all patients of this family, whose clinical features showed DCM combined with MEPPC. The proband's children developed DCM and MEPPC in their childhood. They both carried a SCN5A p.A204E mutation from their mother and a mutation PRKAG2 p.D372N from their father. The son did heart transplant and his heart was both dilated and thickened. The pathology confirmed the presence of glycogen accumulation in the myocardium, which were consistent with the diagnosis of PAKAG2 syndrome. Conclusion SCN5A c.611 C>A variant was related to DCM combined with MEPPC. This case report is the first to demonstrate that a combination of SCN5A and PRKAG2 mutations can cause DCM plus MEPPC and PRKAG2 Syndrome.
Collapse
Affiliation(s)
- Wen Huang
- Department of Medical Ultrasound, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong University, Jinan, China
| | - Rui Xu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong University, Jinan, China
- *Correspondence: Rui Xu
| | - Ning Gao
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong University, Jinan, China
| | - Xia Wu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong University, Jinan, China
| | - Cong Wen
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Weifang Medical University, Jinan, China
| |
Collapse
|
28
|
Implication of a novel truncating mutation in titin as a cause of autosomal dominant left ventricular noncompaction. J Geriatr Cardiol 2022; 19:301-314. [PMID: 35572216 PMCID: PMC9068586 DOI: 10.11909/j.issn.1671-5411.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Mutation in the titin gene (TTN) in left ventricular noncompaction (LVNC) has been reported with a highly heterogeneous prevalence, and the molecular mechanisms underlying the pathogenesis of TTN gene mutation are uncharacterized. In the present study, we identified a novel TTN mutation in a pedigree with LVNC and investigated the potential pathogenic mechanism by functional studies. METHODS The whole-genome sequencing with linkage analysis was performed in a 3-generation family affected by autosomal dominant LVNC cardiomyopathy. The clustered regularly interspaced short palindromic repeats associated protein 9 (CRISPR/Cas9) technology was used to establish novel truncating mutation in TTN in a rat cardiomyoblast H9C2 cell line in vitro, in which functional studies were carried out and characterized in comparison to its wild-type counterpart. RESULTS A novel truncating mutation TTN p. R2021X was identified as the only plausible disease-causing variant that segregated with disease among the five surviving affected individuals, with an interrogation of the entire genome excluding other potential causes. Quantitative reverse transcription-polymerase chain reaction and cellular immunofluorescence supported a haploinsufficient disease mechanism in titin truncation mutation cardiomyocytes. Further functional studies suggested mitochondrial abnormities in the presence of mutation, including decreased oxygen consumption rate, reduced adenosine triphosphate production, impaired activity of electron translation chain, and abnormal mitochondrial structure on electron microscopy. Impaired autophagy under electron microscopy accompanied with activation of the Akt-mTORC1 signaling pathway was observed in TTN p. R2021X truncation mutation cardiomyocytes. CONCLUSIONS The TTN p. R2021X mutation has a function in the cause of a highly penetrant familial LVNC. These findings expand the spectrum of titin's roles in cardiomyopathies and provide novel insight into the molecular basis of titin-truncating variants-associated LVNC.
Collapse
|
29
|
Karvonen V, Harjama L, Heliö K, Kettunen K, Elomaa O, Koskenvuo JW, Kere J, Weckström S, Holmström M, Saarela J, Ranki A, Heliö T, Hannula-Jouppi K. A novel desmoplakin mutation causes dilated cardiomyopathy with palmoplantar keratoderma as an early clinical sign. J Eur Acad Dermatol Venereol 2022; 36:1349-1358. [PMID: 35445468 PMCID: PMC9545885 DOI: 10.1111/jdv.18164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/16/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
Background PPKs represent a heterogeneous group of disorders with hyperkeratosis of palmar and/or plantar skin. PPK, hair shaft abnormalities, cardiomyopathy and arrhythmias can be caused by mutations in desmosomal genes, e.g. desmoplakin (DSP). PPK should trigger genetic testing to reveal mutations with possible related cardiac disease. Objectives To report a large multigenerational family with a novel DSP mutation associated with early‐onset PPK and adult‐onset cardiomyopathy and arrhythmias. Methods A custom‐designed in‐house panel of 35 PPK related genes was used to screen mutations in the index patient with focal PPK. The identified DSP mutation was verified by Sanger sequencing. DNA samples from 20 members of the large multigenerational family were sequenced for the DSP mutation. Medical records were reviewed. Clinical dermatological evaluation was performed, including light microscopy of hair samples. Cardiac evaluation included clinical examination, echocardiography, cardiac magnetic resonance imaging (CMR), electrocardiogram (ECG), Holter monitoring and laboratory tests. Results We identified a novel autosomal dominant truncating DSP c.2493delA p.(Glu831Aspfs*33) mutation associated with dilated cardiomyopathy (DCM) with arrhythmia susceptibility and focal PPK as an early cutaneous sign. The mutation was found in nine affected family members, but not in any unaffected members. Onset of dermatological findings preceded cardiac symptoms which were variable and occurred at adult age. Conclusions We report a novel truncating DSP mutation causing focal PPK with varying severity and left ventricular dilatation and ventricular extrasystoles. This finding emphasizes the importance of genetic diagnosis in patients with PPK for clinical counselling and management of cardiomyopathies and arrhythmias.
Collapse
Affiliation(s)
- V Karvonen
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - L Harjama
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - K Heliö
- Department of Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - K Kettunen
- HUS Diagnostic Center, Division of Genetics and Clinical Pharmacology, Laboratory of Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - O Elomaa
- Folkhälsan Research Center, Helsinki, Finland and Research Programs Unit, Stem Cells and Metabolism Research Program, University of Helsinki, Finland
| | | | - J Kere
- Folkhälsan Research Center, Helsinki, Finland and Research Programs Unit, Stem Cells and Metabolism Research Program, University of Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - S Weckström
- Department of Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - M Holmström
- Radiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - J Saarela
- HUS Diagnostic Center, Division of Genetics and Clinical Pharmacology, Laboratory of Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
| | - A Ranki
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - T Heliö
- Department of Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - K Hannula-Jouppi
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland and Research Programs Unit, Stem Cells and Metabolism Research Program, University of Helsinki, Finland
| |
Collapse
|
30
|
Cannatà A, Merlo M, Dal Ferro M, Barbati G, Manca P, Paldino A, Graw S, Gigli M, Stolfo D, Johnson R, Roy D, Tharratt K, Bromage DI, Jirikowic J, Abbate A, Goodwin A, Rao K, Marawan A, Carr-White G, Robert L, Parikh V, Ashley E, McDonagh T, Lakdawala NK, Fatkin D, Taylor MRG, Mestroni L, Sinagra G. Association of Titin Variations With Late-Onset Dilated Cardiomyopathy. JAMA Cardiol 2022; 7:371-377. [PMID: 35138330 PMCID: PMC8829739 DOI: 10.1001/jamacardio.2021.5890] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/09/2021] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Dilated cardiomyopathy (DCM) is frequently caused by genetic factors. Studies identifying deleterious rare variants have predominantly focused on early-onset cases, and little is known about the genetic underpinnings of the growing numbers of patients with DCM who are diagnosed when they are older than 60 years (ie, late-onset DCM). OBJECTIVE To investigate the prevalence, type, and prognostic impact of disease-associated rare variants in patients with late-onset DCM. DESIGN, SETTING, AND PARTICIPANTS A population of patients with late-onset DCM who had undergone genetic testing in 7 international tertiary referral centers worldwide were enrolled from March 1990 to August 2020. A positive genotype was defined as the presence of pathogenic or likely pathogenic (P/LP) variants. MAIN OUTCOMES AND MEASURES The study outcome was all-cause mortality. RESULTS A total of 184 patients older than 60 years (103 female [56%]; mean [SD] age, 67 [6] years; mean [SD] left ventricular ejection fraction, 32% [10%]) were studied. Sixty-six patients (36%) were carriers of a P/LP variant. Titin-truncating variants were the most prevalent (present in 46 [25%] of the total population and accounting for 46 [69%] of all genotype-positive patients). During a median (interquartile range) follow-up of 42 (10-115) months, 23 patients (13%) died; 17 (25%) of these were carriers of P/LP variants, while 6 patients (5.1%) were genotype-negative. CONCLUSIONS AND RELEVANCE Late-onset DCM might represent a distinct subgroup characterized by and a high genetic variation burden, largely due to titin-truncating variants. Patients with a positive genetic test had higher mortality than genotype-negative patients. These findings support the extended use of genetic testing also in older patients.
Collapse
Affiliation(s)
- Antonio Cannatà
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
- Department of Cardiology, King’s College Hospital, London, United Kingdom
| | - Marco Merlo
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Matteo Dal Ferro
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Giulia Barbati
- Biostatistics Unit, University of Trieste, Trieste, Italy
| | - Paolo Manca
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Alessia Paldino
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Sharon Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora
| | - Marta Gigli
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Davide Stolfo
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Renee Johnson
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- Faculty of Medicine, UNSW Sydney, Kensington, New South Wales, Australia
- Cardiology Department, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia
| | - Darius Roy
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kevin Tharratt
- Center for Inherited Heart Disease, Stanford University, Stanford, California
| | - Daniel I. Bromage
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
- Department of Cardiology, King’s College Hospital, London, United Kingdom
| | - Jean Jirikowic
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Allison Goodwin
- VCU Medical Center, Clinical Genetics Services, Richmond, Virginia
| | - Krishnasree Rao
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Amr Marawan
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Gerry Carr-White
- Department of Cardiology, Guys and St Thomas’ NHS Trust, London, United Kingdom
| | - Leema Robert
- Department of Clinical Genetics, Guys and St Thomas' NHS Trust, London, United Kingdom
| | - Victoria Parikh
- Center for Inherited Heart Disease, Stanford University, Stanford, California
| | - Euan Ashley
- Center for Inherited Heart Disease, Stanford University, Stanford, California
| | - Theresa McDonagh
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
- Department of Cardiology, King’s College Hospital, London, United Kingdom
| | - Neal K. Lakdawala
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Diane Fatkin
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- Faculty of Medicine, UNSW Sydney, Kensington, New South Wales, Australia
- Cardiology Department, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia
| | - Matthew R. G. Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora
| | - Luisa Mestroni
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| |
Collapse
|
31
|
Vogiatzi G, Lazaros G, Oikonomou E, Lazarou E, Vavuranakis E, Tousoulis D. Role of genetic testing in cardiomyopathies: Α primer for cardiologists. World J Cardiol 2022; 14:29-39. [PMID: 35126870 PMCID: PMC8788175 DOI: 10.4330/wjc.v14.i1.29] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/18/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent advances in cardiovascular genetics have transformed genetic testing into a valuable part of management of families with inherited cardiomyopathies. As novel mutations have been identified, understanding when to consider genetic testing has emerged as an important consideration in the management of these cases. Specific genetic testing has a paramount importance in the risk stratification of family members, in the prognosis of probands at higher risk of a serious phenotype expression, and finally in the identification of new mutations, all of which are discussed in this review. The indications for each type of cardiomyopathy are described, along with the limitations of genetic testing. Finally, the importance of public sharing of variants in large data sets is emphasized. The ultimate aim of this review is to present key messages about the genetic testing process in order to minimize potential harms and provide suggestions to specialized clinicians who act as a part of a multidisciplinary team in order to offer the best care to families with inherited cardiomyopathies.
Collapse
Affiliation(s)
- Georgia Vogiatzi
- The Third Department of Cardiology, Sotiria Hospital, Athens 11527, Greece
| | - George Lazaros
- The First Department of Cardiology, Hippokration Hospital, Athens 11526, Greece
| | - Evangelos Oikonomou
- The First Department of Cardiology, Hippokration Hospital, Medical School of National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Emilia Lazarou
- The First Department of Cardiology, Hippokration Hospital, Athens 11526, Greece
| | | | - Dimitris Tousoulis
- The First Department of Cardiology, Hippokration Hospital, Athens 11526, Greece
| |
Collapse
|
32
|
It Is Not Carved in Stone—The Need for a Genetic Reevaluation of Variants in Pediatric Cardiomyopathies. J Cardiovasc Dev Dis 2022; 9:jcdd9020041. [PMID: 35200695 PMCID: PMC8875742 DOI: 10.3390/jcdd9020041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/05/2022] Open
Abstract
(1) Background: In cardiomyopathies, identification of genetic variants is important for the correct diagnosis and impacts family cascade screening. A classification system was published by the American College of Medical Genetics and Genomics (ACMG) in 2015 to standardize variants’ classification. The aim of the study was to determine the rate of reclassification of previously identified variants in patients with childhood-onset cardiomyopathies. (2) Methods: Medical records of patients and their relatives were screened for clinical and genetic information at the Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich. Patients without an identified genetic variant were excluded from further analyses. Previously reported variants were reevaluated by the ACMG criteria in November 2021. (3) Results: Data from 167 patients or relatives of patients with childhood-onset cardiomyopathy from 137 families were analyzed. In total, 45 different genetic variants were identified in 71 individuals. Classification changed in 29% (13/45) with the greatest shift in “variants of unknown significance” to “(likely) benign” (9/13). (4) Conclusions: In patients with childhood-onset cardiomyopathies, nearly a third of reported genetic variants change mostly to more benign classes upon reclassification. Given the impact on patient management and cascade screening, this finding underlines the importance of continuous genetic counseling and variant.
Collapse
|
33
|
Wang Y, Han B, Fan Y, Yi Y, Lv J, Wang J, Yang X, Jiang D, Zhao L, Zhang J, Yuan H. Next-Generation Sequencing Reveals Novel Genetic Variants for Dilated Cardiomyopathy in Pediatric Chinese Patients. Pediatr Cardiol 2022; 43:110-120. [PMID: 34350506 DOI: 10.1007/s00246-021-02698-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022]
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disease characterized by bilateral or left ventricular cardiac dilation and systolic dysfunction that can lead to heart failure and sudden cardiac death in children. Many studies have focused on genetic variation in DCM-related genes in adult populations; however, the mutational landscape in pediatric DCM patients remains undetermined, especially in the Chinese population. We applied next-generation sequencing (NGS) technology to genetically analyze 46 pediatric DCM patients to reveal genotype-phenotype correlations. Our results indicated DCM-associated pathogenic mutations in 10 genes related to the structure or function of the sarcomere, desmosome, and cytoskeleton. We also identified 6 pathogenic mutations (5 novel) in the Titin (TTN) gene that resulted in truncated TTN variants in 6 (13%) out of 46 patients. Correlations between TTN mutations and clinical outcomes were assessed. Our data indicate that one-third of pediatric DCM cases are caused by genetic mutations. The role of TTN variants should not be underestimated in pediatric DCM and age-dependent pathogenic penetrance of these mutations should be considered for familial DCM cases. We argue that genetic testing of DCM cases is valuable for predicting disease severity, prognosis, and recurrence risk, and for screening first-degree relatives.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pediatrics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, People's Republic of China
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Bo Han
- Department of Pediatrics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, People's Republic of China.
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China.
| | - Youfei Fan
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Yingchun Yi
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Jianli Lv
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Jing Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Xiaofei Yang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Diandong Jiang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Lijian Zhao
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Jianjun Zhang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Hui Yuan
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| |
Collapse
|
34
|
Heliö K, Mäyränpää MI, Saarinen I, Ahonen S, Junnila H, Tommiska J, Weckström S, Holmström M, Toivonen M, Nikus K, Hathaway J, Siivonen P, Muona M, Sistonen J, Salmenperä P, Gentile M, Paananen J, Myllykangas S, Alastalo TP, Heliö T, Koskenvuo J. GRINL1A Complex Transcription Unit Containing GCOM1, MYZAP, and POLR2M Genes Associates with Fully Penetrant Recessive Dilated Cardiomyopathy. Front Genet 2021; 12:786705. [PMID: 34899865 PMCID: PMC8656111 DOI: 10.3389/fgene.2021.786705] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Familial dilated cardiomyopathy (DCM) is a monogenic disorder typically inherited in an autosomal dominant pattern. We have identified two Finnish families with familial cardiomyopathy that is not explained by a variant in any previously known cardiomyopathy gene. We describe the cardiac phenotype related to homozygous truncating GCOM1 variants. Methods and Results: This study included two probands and their relatives. All the participants are of Finnish ethnicity. Whole-exome sequencing was used to test the probands; bi-directional Sanger sequencing was used to identify the GCOM1 variants in probands' family members. Clinical evaluation was performed, medical records and death certificates were obtained. Immunohistochemical analysis of myocardial samples was conducted. A homozygous GCOM1 variant was identified altogether in six individuals, all considered to be affected. None of the nine heterozygous family members fulfilled any cardiomyopathy criteria. Heart failure was the leading clinical feature, and the patients may have had a tendency for atrial arrhythmias. Conclusions: This study demonstrates the significance of GCOM1 variants as a cause of human cardiomyopathy and highlights the importance of searching for new candidate genes when targeted gene panels do not yield a positive outcome.
Collapse
Affiliation(s)
- Krista Heliö
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko I Mäyränpää
- Department of Pathology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Inka Saarinen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Saija Ahonen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Heidi Junnila
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | | | - Sini Weckström
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Miia Holmström
- Department of Radiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mia Toivonen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Kjell Nikus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Heart Center, Tampere University Hospital, Tampere, Finland
| | - Julie Hathaway
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Pauli Siivonen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | - Mikko Muona
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | | | | | | | - Jussi Paananen
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| | | | | | - Tiina Heliö
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Juha Koskenvuo
- Blueprint Genetics, A Quest Diagnostics Company, Espoo, Finland
| |
Collapse
|
35
|
Saito T, Sato NS, Mozawa K, Adachi A, Sasaki Y, Nakamura K, Oka E, Otsuka T, Kodani E, Asai K, Mizuno K, Shimizu W, Gottlieb RA. Myocardial ultrastructure can augment genetic testing for sporadic dilated cardiomyopathy with initial heart failure. ESC Heart Fail 2021; 8:5178-5191. [PMID: 34486814 PMCID: PMC8712817 DOI: 10.1002/ehf2.13596] [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: 08/10/2020] [Revised: 07/06/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
Aims The aim of the present study was to consider whether the ultrastructural features of cardiomyocytes in dilated cardiomyopathy can be used to guide genetic testing. Methods and results Endomyocardial biopsy and whole‐exome sequencing were performed in 32 consecutive sporadic dilated cardiomyopathy patients [51.0 (40.0–64.0) years, 75% men] in initial phases of decompensated heart failure. The predicted pathogenicity of ultrarare (minor allele frequency ≤0.0005), non‐synonymous variants was determined using the American College of Medical Genetics guidelines. Focusing on 75 cardiomyopathy‐susceptibility and 41 arrhythmia‐susceptibility genes, we identified 404 gene variants, of which 15 were considered pathogenic or likely pathogenic in 14 patients (44% of 32). There were five sarcomeric gene variants (29% of 17 variants) found in five patients (16% of 32), involving a variant of MYBPC3 and four variants of TTN. A patient with an MYBPC3 variant showed disorganized sarcomeres, three patients with TTN variants located in the region encoding the A‐band domain showed sparse sarcomeres, and a patient with a TTN variant in encoding the I‐band domain showed disrupted sarcomeres. The distribution of diffuse myofilament lysis depended on the causal genes; three patients with the same TMEM43 variant had diffuse myofilament lysis near nuclei (P = 0.011), while two patients with different DSP variants had lysis in the peripheral areas of cardiomyocytes (P = 0.033). Conclusions Derangement patterns of myofilament and subcellular distribution of myofilament lysis might implicate causal genes. Large‐scale studies are required to confirm whether these ultrastructural findings are related to the causative genes.
Collapse
Affiliation(s)
- Tsunenori Saito
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.,Department of Cardiovascular Medicine, Nippon Medical School Graduate School, Tokyo, Japan
| | - Naoko Saito Sato
- Department of Neurology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Kosuke Mozawa
- Department of Cardiovascular Medicine, Nippon Medical School Graduate School, Tokyo, Japan
| | - Akiko Adachi
- Division of Morphological and Biomolecular Research, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshihiro Sasaki
- Division of Morphological and Biomolecular Research, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kotoka Nakamura
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Eiichiro Oka
- Department of Cardiovascular Medicine, Nippon Medical School Graduate School, Tokyo, Japan
| | - Toshiaki Otsuka
- Department of Hygiene and Public Health, Nippon Medical School Graduate School, Tokyo, Japan
| | - Eitaro Kodani
- Department of Internal Medicine and Cardiology, Nippon Medical School Tama Nagayama Hospital, Tokyo, Japan
| | - Kuniya Asai
- Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital, Chiba, Japan
| | - Kyoichi Mizuno
- Department of Cardiovascular Medicine, Nippon Medical School Graduate School, Tokyo, Japan
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School Graduate School, Tokyo, Japan
| | - Roberta A Gottlieb
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| |
Collapse
|
36
|
Nguyen TV, Tran Vu MT, Do TNP, Tran THN, Do TH, Nguyen TMH, Tran Huynh BN, Le LA, Nguyen Pham NT, Nguyen TDA, Nguyen TMN, Le NHP, Pham Nguyen V, Ho Huynh TD. Genetic Determinants and Genotype-Phenotype Correlations in Vietnamese Patients With Dilated Cardiomyopathy. Circ J 2021; 85:1469-1478. [PMID: 34011823 DOI: 10.1253/circj.cj-21-0077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is an important cause of heart failure and cardiac transplantation. This study determined the prevalence of DCM-associated genes and evaluated the genotype-phenotype correlation in Vietnamese patients. METHODS AND RESULTS This study analyzed 58 genes from 230 patients. The study cohort consisted of 64.3% men; age at diagnosis 47.9±13.7 years; familial (10.9%) and sporadic DCM (82.2%). The diagnostic yield was 23.5%, 44.0% in familial and 19.6% in sporadic DCM.TTNtruncating variants (TTNtv) were predominant (46.4%), followed byTPM1,DSP,LMNA,MYBPC3,MYH6,MYH7,DES,TNNT2,ACTC1,ACTN2,BAG3,DMD,FKTN,PLN,TBX5,RBM20,TCAP(2-6%). Familial DCM, genotype-positive andTTNtv-positive patients were younger than those with genotype-negative and sporadic DCM. Genotype-positive patients displayed a decreased systolic blood pressure and left ventricular wall thickness compared to genotype-negative patients. Genotype-positive patients, particularly those withTTNtv, had a family history of DCM, higher left atrial volume index and body mass index, and lower right ventricle-fractional area change than genotype-negative patients. Genotype-positive patients reached the combined outcomes more frequently and at a younger age than genotype-negative patients. Major cardiac events occurred more frequently in patients positive with genes other thanTTNtv. CONCLUSIONS The study findings provided an overview of Vietnamese DCM patients' genetic profile and suggested that management of environmental factors may be beneficial for DCM patients.
Collapse
Affiliation(s)
- Thuy Vy Nguyen
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | | | | | | | | | | | | | | | | | | | - Thi My Nuong Nguyen
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | - Ngoc Hong Phuong Le
- Research Center for Genetics and Reproductive Health, School of Medicine, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | | | - Thuy Duong Ho Huynh
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
- Research Center for Genetics and Reproductive Health, School of Medicine, VNUHCM [Vietnam National University, Ho Chi Minh City]
- KTEST Science Company
| |
Collapse
|
37
|
Giri P, Mukhopadhyay A, Gupta M, Mohapatra B. Dilated cardiomyopathy: a new insight into the rare but common cause of heart failure. Heart Fail Rev 2021; 27:431-454. [PMID: 34245424 DOI: 10.1007/s10741-021-10125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
Heart failure is a global health burden responsible for high morbidity and mortality with a prevalence of greater than 60 million individuals worldwide. One of the major causes of heart failure is dilated cardiomyopathy (DCM), characterized by associated systolic dysfunction. During the last few decades, there have been remarkable advances in our understanding about the genetics of dilated cardiomyopathy. The genetic causes were initially thought to be associated with mutations in genes encoding proteins that are localized to cytoskeleton and sarcomere only; however, with the advancement in mechanistic understanding, the roles of ion channels, Z-disc, mitochondria, nuclear proteins, cardiac transcription factors (e.g., NKX-2.5, TBX20, GATA4), and the factors involved in calcium homeostasis have also been identified and found to be implicated in both familial and sporadic DCM cases. During past few years, next-generation sequencing (NGS) has been established as a diagnostic tool for genetic analysis and it has added significantly to the existing candidate gene list for DCM. The animal models have also provided novel insights to develop a better treatment strategy based on phenotype-genotype correlation, epigenetic and phenomic profiling. Most of the DCM biomarkers that are used in routine genetic and clinical testing are structural proteins, but during the last few years, the role of mi-RNA has also emerged as a biomarker due to their accessibility through noninvasive methods. Our increasing genetic knowledge can improve the clinical management of DCM by bringing clinicians and geneticists on one platform, thereby influencing the individualized clinical decision making and leading to precision medicine.
Collapse
Affiliation(s)
- Prerna Giri
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Amrita Mukhopadhyay
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Mohini Gupta
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India.
| |
Collapse
|
38
|
Al-Shafai KN, Al-Hashemi M, Manickam C, Musa R, Selvaraj S, Syed N, Vempalli F, Ali M, Yacoub M, Estivill X. Genetic evaluation of cardiomyopathies in Qatar identifies enrichment of pathogenic sarcomere gene variants and possible founder disease mutations in the Arabs. Mol Genet Genomic Med 2021; 9:e1709. [PMID: 34137518 PMCID: PMC8372065 DOI: 10.1002/mgg3.1709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/03/2020] [Accepted: 05/04/2021] [Indexed: 01/20/2023] Open
Abstract
Background Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are serious inherited heart diseases with various causative mutations identified. The full spectrum of causative mutations remains to be discovered, especially in understudied populations. Methods Here, we established the DOHA Registry and Biobank for cardiomyopathies in Qatar, followed by sequencing of 174 genes on 51 HCM and 53 DCM patients, and 31 relatives. Results In HCM, the analysis of 25 HCM‐associated genes showed that 20% of HCM cases had putative pathogenic variants for cardiomyopathy, mainly in sarcomere genes. Additional 49% of HCM cases had variants of uncertain significance, while 31% of HCM cases had likely benign variant(s) or had no variants identified within the analyzed HCM genes. In DCM, 56 putative DCM genes were analyzed. Eight percent of DCM cases had putative pathogenic variants for DCM, in the TTN gene while 70% of cases had variants of uncertain significance, in the analyzed DCM genes, that will need further pathogenicity assessment. Moreover, 22% of DCM cases remain unexplained, by having likely benign variant(s) or having no variants detected in any of the analyzed DCM genes. Conclusion We identified or replicated at least four recurrent variants among cardiomyopathy patients, which could be founder disease mutations in the Arabic population, including a frameshift variant (c.1371_1381dupTATCCAGTTAT) of unknown significance in the FKTN gene which seems to cause DCM in homozygosity, and HCM in heterozygosity. In vivo and/or in vitro functional validation need to be pursued in order to assess the pathogenicity of the identified variants.
Collapse
Affiliation(s)
- Kholoud N Al-Shafai
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Sidra Research Department, Sidra Medicine, Doha, Qatar
| | | | | | - Rania Musa
- The Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Najeeb Syed
- Sidra Research Department, Sidra Medicine, Doha, Qatar
| | | | - Muneera Ali
- The Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Magdi Yacoub
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Xavier Estivill
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Sidra Research Department, Sidra Medicine, Doha, Qatar.,Quantitative Genomics Laboratories (qGenomics, Barcelona, Spain
| |
Collapse
|
39
|
The Combined Human Genotype of Truncating TTN and RBM20 Mutations Is Associated with Severe and Early Onset of Dilated Cardiomyopathy. Genes (Basel) 2021; 12:genes12060883. [PMID: 34201072 PMCID: PMC8228627 DOI: 10.3390/genes12060883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 12/30/2022] Open
Abstract
A major cause of heart failure is cardiomyopathies, with dilated cardiomyopathy (DCM) as the most common form. Over 40 genes are linked to DCM, among them TTN and RBM20. Next Generation Sequencing in clinical DCM cohorts revealed truncating variants in TTN (TTNtv), accounting for up to 25% of familial DCM cases. Mutations in the cardiac splicing factor RNA binding motif protein 20 (RBM20) are also known to be associated with severe cardiomyopathies. TTN is one of the major RBM20 splicing targets. Most of the pathogenic RBM20 mutations are localized in the highly conserved arginine serine rich domain (RS), leading to a cytoplasmic mislocalization of mutant RBM20. Here, we present a patient with an early onset DCM carrying a combination of (likely) pathogenic TTN and RBM20 mutations. We show that the splicing of RBM20 target genes is affected in the mutation carrier. Furthermore, we reveal RBM20 haploinsufficiency presumably caused by the frameshift mutation in RBM20.
Collapse
|
40
|
Seguret M, Vermersch E, Jouve C, Hulot JS. Cardiac Organoids to Model and Heal Heart Failure and Cardiomyopathies. Biomedicines 2021; 9:563. [PMID: 34069816 PMCID: PMC8157277 DOI: 10.3390/biomedicines9050563] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiac tissue engineering aims at creating contractile structures that can optimally reproduce the features of human cardiac tissue. These constructs are becoming valuable tools to model some of the cardiac functions, to set preclinical platforms for drug testing, or to alternatively be used as therapies for cardiac repair approaches. Most of the recent developments in cardiac tissue engineering have been made possible by important advances regarding the efficient generation of cardiac cells from pluripotent stem cells and the use of novel biomaterials and microfabrication methods. Different combinations of cells, biomaterials, scaffolds, and geometries are however possible, which results in different types of structures with gradual complexities and abilities to mimic the native cardiac tissue. Here, we intend to cover key aspects of tissue engineering applied to cardiology and the consequent development of cardiac organoids. This review presents various facets of the construction of human cardiac 3D constructs, from the choice of the components to their patterning, the final geometry of generated tissues, and the subsequent readouts and applications to model and treat cardiac diseases.
Collapse
Affiliation(s)
- Magali Seguret
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Eva Vermersch
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Charlène Jouve
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Jean-Sébastien Hulot
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
- CIC1418 and DMU CARTE, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| |
Collapse
|
41
|
Valverde-Gómez M, Ruiz-Curiel A, Melendo-Viu M, Salguero-Bodes R, Martín-Arriscado C, Bueno H, Jiménez-López-Guarch C, Rebolo-Bardanca P, Huertas-Nieto S, Montañés-Delmas E, Delgado-Jiménez J, Domínguez-González C, Arribas-Ynsaurriaga F, Palomino-Doza J. Electrocardiogram Changes in the Spectrum of TTNtv Dilated Cardiomyopathy: Accuracy and Predictive Value of a New Index for LV-Changes Identification. Heart Lung Circ 2021; 30:1487-1495. [PMID: 33994281 DOI: 10.1016/j.hlc.2021.04.011] [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: 02/12/2021] [Revised: 03/12/2021] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Truncating TTN variants (TTNtv) are the main cause of dilated cardiomyopathy (DCM). The dynamic nature of this entity has previously been described. Based on own empirical observations and previous evidences, this study assessed repolarisation patterns and the possible association with morphological and functional status of TTNtv-DCM patients. METHODS Electrocardiograms (ECGs) of index patients with TTNtv-DCM and their relatives were included and matched in time with an echocardiogram. All individuals were classified into five phenotype groups: 1) Reduced left ventricular ejection fraction (LVEF <50%); 2) Recovered LVEF: at least 10% increase and LVEF >30% after optimal medical treatment; 3) Borderline phenotype (mildly enlarged ventricle and/or hyper-trabeculation); 4) Genotype positive, phenotype negative; and 5) Non-carriers. All electrocardiograms were evaluated by two blinded observers in qualitative and quantitative terms [T index (mm)=Σ T-wave amplitude (V5, V6, II, aVF)] and these data were compared with demographic and clinical information. The Δ T-index was calculated in those individuals with more than one electrocardiogram. RESULTS Seventy-eight (78) electrocardiograms were included (46% female, mean age 50 years). T-index and prevalence of an abnormal T-wave had significantly different results among the groups (p<0.0001). Age and haemodynamic factors were shown to be ECG-modifiers, especially in phenotype-negative patients. T-index enabled individuals with reduced LVEF (<2.5) to be identified and to differentiate patients with favourable and unfavourable responses to treatment (Δ T index >3.5 and ≤2, respectively). CONCLUSION Repolarisation changes enabled characterisation of the spectrum of TTNtv-DCM. The T-index identified potential carriers and patients with the worst profiles of the spectrum of the disease.
Collapse
Affiliation(s)
- María Valverde-Gómez
- HealthInCode, La Coruña, Spain; Faculty of Medicine, Complutense University of Madrid, Spain.
| | - Aníbal Ruiz-Curiel
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain
| | - María Melendo-Viu
- Cardiology Department, University Hospital Álvaro Cunqueiro, Vigo, Spain
| | - Rafael Salguero-Bodes
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| | | | - Héctor Bueno
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| | - Carmen Jiménez-López-Guarch
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| | | | - Sergio Huertas-Nieto
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain
| | - Elena Montañés-Delmas
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain
| | - Juan Delgado-Jiménez
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| | - Cristina Domínguez-González
- Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; Neurology Department, 12 de Octubre University Hospital, Madrid, Spain; CIBERER (Biomedical Research Networking Centres, Rare Diseases), Institute of Health Carlos III, Madrid, Spain
| | - Fernando Arribas-Ynsaurriaga
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| | - Julián Palomino-Doza
- Cardiology Department, 12 de Octubre University Hospital, Madrid, Spain; Research Institute i+12, 12 de Octubre University Hospital, Madrid, Spain; CIBER-CV (Biomedical Research Networking Centres, Cardiovascular Diseases), Institute of Health Carlos III, Madrid, Spain; Faculty of Medicine, Complutense University of Madrid, Spain
| |
Collapse
|
42
|
Kany S, Reissmann B, Metzner A, Kirchhof P, Darbar D, Schnabel RB. Genetics of atrial fibrillation-practical applications for clinical management: if not now, when and how? Cardiovasc Res 2021; 117:1718-1731. [PMID: 33982075 PMCID: PMC8208749 DOI: 10.1093/cvr/cvab153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The prevalence and economic burden of atrial fibrillation (AF) are predicted to more than double over the next few decades. In addition to anticoagulation and treatment of concomitant cardiovascular conditions, early and standardized rhythm control therapy reduces cardiovascular outcomes as compared with a rate control approach, favouring the restoration, and maintenance of sinus rhythm safely. Current therapies for rhythm control of AF include antiarrhythmic drugs (AADs) and catheter ablation (CA). However, response in an individual patient is highly variable with some remaining free of AF for long periods on antiarrhythmic therapy, while others require repeat AF ablation within weeks. The limited success of rhythm control therapy for AF is in part related to incomplete understanding of the pathophysiological mechanisms and our inability to predict responses in individual patients. Thus, a major knowledge gap is predicting which patients with AF are likely to respond to rhythm control approach. Over the last decade, tremendous progress has been made in defining the genetic architecture of AF with the identification of rare mutations in cardiac ion channels, signalling molecules, and myocardial structural proteins associated with familial (early-onset) AF. Conversely, genome-wide association studies have identified common variants at over 100 genetic loci and the development of polygenic risk scores has identified high-risk individuals. Although retrospective studies suggest that response to AADs and CA is modulated in part by common genetic variation, the development of a comprehensive clinical and genetic risk score may enable the translation of genetic data to the bedside care of AF patients. Given the economic impact of the AF epidemic, even small changes in therapeutic efficacy may lead to substantial improvements for patients and health care systems.
Collapse
Affiliation(s)
- Shinwan Kany
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Bruno Reissmann
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Andreas Metzner
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,The Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston Birmingham B15 2TT, UK
| | - Dawood Darbar
- Division of Cardiology, Departments of Medicine, University of Illinois at Chicago and Jesse Brown Veterans Administration, 840 South Wood Street, Suite 928 M/C 715, Chicago, IL 60612, USA
| | - Renate B Schnabel
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| |
Collapse
|
43
|
Xiao L, Li C, Sun Y, Chen Y, Wei H, Hu D, Yu T, Li X, Jin L, Shi L, Marian AJ, Wang DW. Clinical Significance of Variants in the TTN Gene in a Large Cohort of Patients With Sporadic Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:657689. [PMID: 33996946 PMCID: PMC8120103 DOI: 10.3389/fcvm.2021.657689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Mutations in the TTN gene are the most common causes of dilated cardiomyopathy (DCM). The clinical significance of TTN gene variants remains inadequately understood. Methods: Whole-exome sequencing and phenotypic characterisation were performed, and patients were followed up for a median of 44 months. Results: We analyzed the association of the TTN variants with the clinical outcomes in a prospective study of 1,041 patients with sporadic DCM. TTN truncating variants (tTTN) were detected in 120 (11.5%) patients as compared with 2.4/10,000 East Asian populations in the Genome Aggregation Database (GnomAD; p < 0.0001). Pathogenic TTN missense variants were also enriched in DCM as compared with the GnomAD populations (27.6 vs. 5.9%, p < 0.0001). DCM patients with tTTN had a lower left ventricular ejection fraction (28.89 ± 8.72 vs. 31.81 ± 9.97, p = 0.002) and a lower frequency of the left bundle branch block (3.3 vs. 11.3%, p = 0.011) than those without or with mutations in other known causal genes (OCG). However, tTTN were not associated with the composite primary endpoint of cardiac death and heart transplantation during the follow-up period [adjusted hazard ratio (HR): 0.912; 95% confidence interval: 0.464–1.793; p = 0.790]. There was also no sex-dependent effect. Concomitant tTTN and pathogenic variants in OCG were present in only eight DCM patients and did not affect the outcome. Conclusion: The phenotype of DCM caused by tTTN, major causes of sporadic DCM, is not distinctly different from those caused by other causal genes for DCM.
Collapse
Affiliation(s)
- Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yanghui Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqing Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Li Jin
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Leming Shi
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Ali J Marian
- Center for Cardiovascular Genetics, Houston, TX, United States
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Evaluating the monogenic contribution and genotype-phenotype correlation in patients with isolated thoracic aortic aneurysm. Eur J Hum Genet 2021; 29:1129-1138. [PMID: 33824467 DOI: 10.1038/s41431-021-00857-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 01/09/2023] Open
Abstract
Thoracic aortic aneurysm with or without dissection (TAAD) can be broadly categorized as syndromic TAAD (sTAAD) and isolated TAAD (iTAAD). sTAAD and is highly correlated with genetics. However, although the incidence of iTAAD is much higher, its monogenic contribution is not yet clear. Here, we sequenced 15 known TAAD genes for 578 iTAAD cases from four cardiac centers in China and found that 10.6% patients with a pathogenic/likely pathogenic (P/LP) variant. Other 7.27% of patients carried variants of uncertain significance in these target genes. We further investigated the correlations among genetics, clinical features, and long-term outcomes. Genetic patients showed younger onset ages (P = 1.31E-13) and larger aortic diameter (P = 1.00E-6), with the youngest age in patients with FBN1 P/LP variants. Monogenic variants were also associated with more aortic segments involved (P = 0.043) and complicated with initial dissection (P = 4.50E-5), especially for genetic patients with non-FBN1 P/LP variants. MACEs occurred in 14.9% patients during follow-up of median 55 months. Genetic status (P = 0.001) and initial dissection (P = 3.00E-6) were two major risk factors for poor prognosis. Early onset age was associated with MACEs in non-genetic cases without initial dissection (P = 0.005). Our study revealed the monogenic contribution in known TAAD genes to iTAAD patients. The genotype-phenotype correlations may complement the risk stratification of iTAAD patients and identification of higher risk subgroups, as well as assist the development of tailored precision medicine in iTAAD.
Collapse
|
45
|
Alimohamed MZ, Johansson LF, Posafalvi A, Boven LG, van Dijk KK, Walters L, Vos YJ, Westers H, Hoedemaekers YM, Sinke RJ, Sijmons RH, Sikkema-Raddatz B, Jongbloed JDH, van der Zwaag PA. Diagnostic yield of targeted next generation sequencing in 2002 Dutch cardiomyopathy patients. Int J Cardiol 2021; 332:99-104. [PMID: 33662488 DOI: 10.1016/j.ijcard.2021.02.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Next-generation sequencing (NGS) is increasingly used for clinical evaluation of cardiomyopathy patients as it allows for simultaneous screening of multiple cardiomyopathy-associated genes. Adding copy number variant (CNV) analysis of NGS data is not routine yet and may contribute to the diagnostic yield. OBJECTIVES Determine the diagnostic yield of our targeted NGS gene panel in routine clinical diagnostics of Dutch cardiomyopathy patients and explore the impact of exon CNVs on diagnostic yield. METHODS Patients (N = 2002) referred for clinical genetic analysis underwent diagnostic testing of 55-61 genes associated with cardiomyopathies. Samples were analyzed and evaluated for single nucleotide variants (SNVs), indels and CNVs. CNVs identified in the NGS data and suspected of being pathogenic based on type, size and location were confirmed by additional molecular tests. RESULTS A (likely) pathogenic (L)P variant was detected in 22.7% of patients, including 3 with CNVs and 25 where a variant was identified in a gene currently not associated with the patient's cardiomyopathy subtype. Only 15 out of 2002 patients (0.8%) were found to carry two (L)P variants. CONCLUSION The yield of routine clinical diagnostics of cardiomyopathies was relatively low when compared to literature. This is likely due to the fact that our study reports the outcome of patients in daily routine diagnostics, therefore also including patients not fully fulfilling (subtype specific) cardiomyopathy criteria. This may also explain why (L)P variants were identified in genes not associated with the reported subtype. The added value of CNV analysis was shown to be limited but not negligible.
Collapse
Affiliation(s)
- Mohamed Z Alimohamed
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Lennart F Johansson
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Genomics Coordination Center, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Anna Posafalvi
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Ludolf G Boven
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Krista K van Dijk
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Lisa Walters
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Yvonne J Vos
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Helga Westers
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Yvonne M Hoedemaekers
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Richard J Sinke
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Rolf H Sijmons
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Birgit Sikkema-Raddatz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Jan D H Jongbloed
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Paul A van der Zwaag
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| |
Collapse
|
46
|
Koskenvuo JW, Saarinen I, Ahonen S, Tommiska J, Weckström S, Seppälä EH, Tuupanen S, Kangas-Kontio T, Schleit J, Heliö K, Hathaway J, Gummesson A, Dahlberg P, Ojala TH, Vepsäläinen V, Kytölä V, Muona M, Sistonen J, Salmenperä P, Gentile M, Paananen J, Myllykangas S, Alastalo TP, Heliö T. Biallelic loss-of-function in NRAP is a cause of recessive dilated cardiomyopathy. PLoS One 2021; 16:e0245681. [PMID: 33534821 PMCID: PMC7857588 DOI: 10.1371/journal.pone.0245681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/05/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Familial dilated cardiomyopathy (DCM) is typically a monogenic disorder with dominant inheritance. Although over 40 genes have been linked to DCM, more than half of the patients undergoing comprehensive genetic testing are left without molecular diagnosis. Recently, biallelic protein-truncating variants (PTVs) in the nebulin-related anchoring protein gene (NRAP) were identified in a few patients with sporadic DCM. METHODS AND RESULTS We determined the frequency of rare NRAP variants in a cohort of DCM patients and control patients to further evaluate role of this gene in cardiomyopathies. A retrospective analysis of our internal variant database consisting of 31,639 individuals who underwent genetic testing (either panel or direct exome sequencing) was performed. The DCM group included 577 patients with either a confirmed or suspected DCM diagnosis. A control cohort of 31,062 individuals, including 25,912 individuals with non-cardiac (control group) and 5,150 with non-DCM cardiac indications (Non-DCM cardiac group). Biallelic (n = 6) or two (n = 5) NRAP variants (two PTVs or PTV+missense) were identified in 11 unrelated probands with DCM (1.9%) but none of the controls. None of the 11 probands had an alternative molecular diagnosis. Family member testing supports co-segregation. Biallelic or potentially biallelic NRAP variants were enriched in DCM vs. controls (OR 1052, p<0.0001). Based on the frequency of NRAP PTVs in the gnomAD reference population, and predicting full penetrance, biallelic NRAP variants could explain 0.25%-2.46% of all DCM cases. CONCLUSION Loss-of-function in NRAP is a cause for autosomal recessive dilated cardiomyopathy, supporting its inclusion in comprehensive genetic testing.
Collapse
Affiliation(s)
- Juha W. Koskenvuo
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
- * E-mail:
| | - Inka Saarinen
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | - Saija Ahonen
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | | | - Sini Weckström
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Eija H. Seppälä
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | - Sari Tuupanen
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | | | | | - Krista Heliö
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | - Julie Hathaway
- Blueprint Genetics Inc, a Quest Diagnostics Company, Seattle, Washington, United States of America
| | - Anders Gummesson
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pia Dahlberg
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tiina H. Ojala
- Department of Pediatric Cardiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Ville Kytölä
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | - Mikko Muona
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | | | | | | | - Jussi Paananen
- Blueprint Genetics, a Quest Diagnostics Company, Espoo, Finland
| | | | - Tero-Pekka Alastalo
- Blueprint Genetics Inc, a Quest Diagnostics Company, Seattle, Washington, United States of America
| | - Tiina Heliö
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Bertero A, Rosa-Garrido M. Three-dimensional chromatin organization in cardiac development and disease. J Mol Cell Cardiol 2021; 151:89-105. [PMID: 33242466 PMCID: PMC11056610 DOI: 10.1016/j.yjmcc.2020.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Recent technological advancements in the field of chromatin biology have rewritten the textbook on nuclear organization. We now appreciate that the folding of chromatin in the three-dimensional space (i.e. its 3D "architecture") is non-random, hierarchical, and highly complex. While 3D chromatin structure is partially encoded in the primary sequence and thereby broadly conserved across cell types and states, a substantial portion of the genome seems to be dynamic during development or in disease. Moreover, there is growing evidence that at least some of the 3D structure of chromatin is functionally linked to gene regulation, both being modulated by and impacting on multiple nuclear processes (including DNA replication, transcription, and RNA splicing). In recent years, these new concepts have nourished several investigations about the functional role of 3D chromatin topology dynamics in the heart during development and disease. This review aims to provide a comprehensive overview of our current understanding in this field, and to discuss how this knowledge can inform further research as well as clinical practice.
Collapse
Affiliation(s)
- Alessandro Bertero
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA 98109, USA.
| | - Manuel Rosa-Garrido
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, 650 Charles Young Dr, Los Angeles, CA 90095, USA.
| |
Collapse
|
48
|
Asselbergs FW, Sammani A, Elliott P, Gimeno JR, Tavazzi L, Tendera M, Kaski JP, Maggioni AP, Rubis PP, Jurcut R, Heliö T, Calò L, Sinagra G, Zdravkovic M, Olivotto I, Kavoliūnienė A, Laroche C, Caforio AL, Charron P. Differences between familial and sporadic dilated cardiomyopathy: ESC EORP Cardiomyopathy & Myocarditis registry. ESC Heart Fail 2021; 8:95-105. [PMID: 33179448 PMCID: PMC7835585 DOI: 10.1002/ehf2.13100] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/22/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Dilated cardiomyopathy (DCM) is a complex disease where genetics interplay with extrinsic factors. This study aims to compare the phenotype, management, and outcome of familial DCM (FDCM) and non-familial (sporadic) DCM (SDCM) across Europe. METHODS AND RESULTS Patients with DCM that were enrolled in the prospective ESC EORP Cardiomyopathy & Myocarditis Registry were included. Baseline characteristics, genetic testing, genetic yield, and outcome were analysed comparing FDCM and SDCM; 1260 adult patients were studied (238 FDCM, 707 SDCM, and 315 not disclosed). Patients with FDCM were younger (P < 0.01), had less severe disease phenotype at presentation (P < 0.02), more favourable baseline cardiovascular risk profiles (P ≤ 0.007), and less medication use (P ≤ 0.042). Outcome at 1 year was similar and predicted by NYHA class (HR 0.45; 95% CI [0.25-0.81]) and LVEF per % decrease (HR 1.05; 95% CI [1.02-1.08]. Throughout Europe, patients with FDCM received more genetic testing (47% vs. 8%, P < 0.01) and had higher genetic yield (55% vs. 22%, P < 0.01). CONCLUSIONS We observed that FDCM and SDCM have significant differences at baseline but similar short-term prognosis. Whether modification of associated cardiovascular risk factors provide opportunities for treatment remains to be investigated. Our results also show a prevalent role of genetics in FDCM and a non-marginal yield in SDCM although genetic testing is largely neglected in SDCM. Limited genetic testing and heterogeneity in panels provides a scaffold for improvement of guideline adherence.
Collapse
Affiliation(s)
- Folkert W. Asselbergs
- Department of CardiologyUniversity Medical Centre Utrecht, University of UtrechtHeidelberglaan 100Utrecht3584CXThe Netherlands
- Institute of Cardiovascular Science and Institute of Health Informatics, Faculty of Population Health SciencesUniversity College LondonLondonUK
| | - Arjan Sammani
- Department of CardiologyUniversity Medical Centre Utrecht, University of UtrechtHeidelberglaan 100Utrecht3584CXThe Netherlands
| | - Perry Elliott
- Barts Heart Centre, St Bartholomew's HospitalUniversity College London and Inherited Cardiac Diseases UnitLondonUK
| | - Juan R. Gimeno
- Cardiac DepartmentHospital Universitario Virgen de la ArrixacaMurciaSpain
| | - Luigi Tavazzi
- GVM Care & ResearchMaria Cecilia HospitalCotignolaItaly
| | - Michael Tendera
- Department of Cardiology and Structural Heart Diseases, School of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Juan Pablo Kaski
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street HospitalUK and University College London Institute of Cardiovascular ScienceLondonUK
| | - Aldo P. Maggioni
- GVM Care & ResearchMaria Cecilia HospitalCotignolaItaly
- EUR Observational Research Programme, European Society of CardiologySophia‐AntipolisFrance
| | - Pawel P. Rubis
- Department of Cardiac and Vascular DiseasesJagiellonian University Medical College, John Paul II HospitalKrakowPoland
| | - Ruxandra Jurcut
- Department of CardiologyEmergency Institute of Cardiovascular Diseases C.C. IliescuBucharestRomania
| | - Tiina Heliö
- Department of CardiologyHelsinki University Central Hospital MeilahtiHelsinkiFinland
| | | | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliano IsontinaTriesteItaly
| | - Marija Zdravkovic
- Clinical Hospital Center Bezanijska kosa, Faculty of MedicineUniversity of BelgradeBeogradSerbia
| | | | - Aušra Kavoliūnienė
- Department of CardiologyLithuanian University of Health SciencesKaunasLithuania
| | - Cécile Laroche
- EUR Observational Research Programme, European Society of CardiologySophia‐AntipolisFrance
| | - Alida L.P. Caforio
- Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences and Public HealthUniversity of PadovaPadovaItaly
| | - Philippe Charron
- APHP, Centre de Référence des Maladies Cardiaques Héréditaires, ICAN, Hôpital Pitié‐SalpêtrièreSorbonne UniversitéParisFrance
| | | |
Collapse
|
49
|
Santiago CF, Huttner IG, Fatkin D. Mechanisms of TTNtv-Related Dilated Cardiomyopathy: Insights from Zebrafish Models. J Cardiovasc Dev Dis 2021; 8:jcdd8020010. [PMID: 33504111 PMCID: PMC7912658 DOI: 10.3390/jcdd8020010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder characterized by ventricular dilation and contractile dysfunction that is associated with significant morbidity and mortality. New insights into disease mechanisms and strategies for treatment and prevention are urgently needed. Truncating variants in the TTN gene, which encodes the giant sarcomeric protein titin (TTNtv), are the most common genetic cause of DCM, but exactly how TTNtv promote cardiomyocyte dysfunction is not known. Although rodent models have been widely used to investigate titin biology, they have had limited utility for TTNtv-related DCM. In recent years, zebrafish (Danio rerio) have emerged as a powerful alternative model system for studying titin function in the healthy and diseased heart. Optically transparent embryonic zebrafish models have demonstrated key roles of titin in sarcomere assembly and cardiac development. The increasing availability of sophisticated imaging tools for assessment of heart function in adult zebrafish has revolutionized the field and opened new opportunities for modelling human genetic disorders. Genetically modified zebrafish that carry a human A-band TTNtv have now been generated and shown to spontaneously develop DCM with age. This zebrafish model will be a valuable resource for elucidating the phenotype modifying effects of genetic and environmental factors, and for exploring new drug therapies.
Collapse
Affiliation(s)
- Celine F. Santiago
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Inken G. Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Cardiology Department, St. Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
- Correspondence:
| |
Collapse
|
50
|
Auxerre-Plantié E, Nielsen T, Grunert M, Olejniczak O, Perrot A, Özcelik C, Harries D, Matinmehr F, Dos Remedios C, Mühlfeld C, Kraft T, Bodmer R, Vogler G, Sperling SR. Identification of MYOM2 as a candidate gene in hypertrophic cardiomyopathy and Tetralogy of Fallot, and its functional evaluation in the Drosophila heart. Dis Model Mech 2020; 13:dmm045377. [PMID: 33033063 PMCID: PMC7758640 DOI: 10.1242/dmm.045377] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/01/2020] [Indexed: 01/11/2023] Open
Abstract
The causal genetic underpinnings of congenital heart diseases, which are often complex and multigenic, are still far from understood. Moreover, there are also predominantly monogenic heart defects, such as cardiomyopathies, with known disease genes for the majority of cases. In this study, we identified mutations in myomesin 2 (MYOM2) in patients with Tetralogy of Fallot (TOF), the most common cyanotic heart malformation, as well as in patients with hypertrophic cardiomyopathy (HCM), who do not exhibit any mutations in the known disease genes. MYOM2 is a major component of the myofibrillar M-band of the sarcomere, and a hub gene within interactions of sarcomere genes. We show that patient-derived cardiomyocytes exhibit myofibrillar disarray and reduced passive force with increasing sarcomere lengths. Moreover, our comprehensive functional analyses in the Drosophila animal model reveal that the so far uncharacterized fly gene CG14964 [herein referred to as Drosophila myomesin and myosin binding protein (dMnM)] may be an ortholog of MYOM2, as well as other myosin binding proteins. Its partial loss of function or moderate cardiac knockdown results in cardiac dilation, whereas more severely reduced function causes a constricted phenotype and an increase in sarcomere myosin protein. Moreover, compound heterozygous combinations of CG14964 and the sarcomere gene Mhc (MYH6/7) exhibited synergistic genetic interactions. In summary, our results suggest that MYOM2 not only plays a critical role in maintaining robust heart function but may also be a candidate gene for heart diseases such as HCM and TOF, as it is clearly involved in the development of the heart.This article has an associated First Person interview with Emilie Auxerre-Plantié and Tanja Nielsen, joint first authors of the paper.
Collapse
Affiliation(s)
- Emilie Auxerre-Plantié
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Tanja Nielsen
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marcel Grunert
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Olga Olejniczak
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Andreas Perrot
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Cemil Özcelik
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Dennis Harries
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Faramarz Matinmehr
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Cristobal Dos Remedios
- Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, Sydney, New South Wales 2006, Australia
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Theresia Kraft
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Georg Vogler
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Silke R Sperling
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|