1
|
Cardoso I, Melo M, Brás P, Viegas JM, Almeida I, Nunes S, Custódio I, Trigo C, Laranjo S, Graça R, Cruz Ferreira R, Oliveira M, Aguiar Rosa S, Antunes D. The contribution of genetics to the understanding and management of cardiomyopathies: Part 2. Rev Port Cardiol 2025; 44:321-329. [PMID: 40154602 DOI: 10.1016/j.repc.2024.11.016] [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: 11/28/2023] [Revised: 05/19/2024] [Accepted: 11/06/2024] [Indexed: 04/01/2025] Open
Abstract
Cardiomyopathies may present as a manifestation of various inherited syndromes. Recognizing the rarity and diagnostic challenges of syndromic and metabolic cardiomyopathies is crucial, as their identification holds significant implications for targeted treatment and enables the use of specific risk stratification tools. Genetics has assumed a pivotal role in clarifying the pathophysiology of cardiomyopathies, facilitating molecular diagnosis, and enabling effective family screening. The advent of next-generation sequencing has revolutionized genetic testing, enabling cost-effective, high-throughput analyses, facilitating the diagnosis of these rare conditions, and allowing the provision of specific management and therapeutics.
Collapse
Affiliation(s)
- Isabel Cardoso
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal.
| | - Mafalda Melo
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Pedro Brás
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - José Miguel Viegas
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Inês Almeida
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Sofia Nunes
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Inês Custódio
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Conceição Trigo
- Serviço de Cardiologia Pediátrica, Centro de Referência de Cardiopatias Congénitas do CHULC, Hospital de Santa Marta, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network Guard-Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Sérgio Laranjo
- Serviço de Cardiologia Pediátrica, Centro de Referência de Cardiopatias Congénitas do CHULC, Hospital de Santa Marta, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network Guard-Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Rafael Graça
- GenoMed - Diagnósticos de Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Rui Cruz Ferreira
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Mário Oliveira
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Centro Clínico Académico de Lisboa, Lisboa, Portugal; Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, CHLC, Lisbon, Portugal; Institute of Physiology and CCUL, Faculty of Medicine of Lisbon, Portugal; Cardiology Center, Hospital CUF Tejo, Lisbon, Portugal
| | - Sílvia Aguiar Rosa
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal; CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Diana Antunes
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal; GenoMed - Diagnósticos de Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| |
Collapse
|
2
|
Ding Q, Fine J, Hoffman FT, Kluge ML, Kuennen RK, Thieke SM, Hoppman NL, Marcou CA, Rowsey RA, Thorland EC, Baudhuin LM, Moyer AM, Buglioni A. Follow the LINE: A novel case of dilated cardiomyopathy caused by a LINE-1 insertion in the TTN gene. Am J Clin Pathol 2025; 163:511-515. [PMID: 39928391 DOI: 10.1093/ajcp/aqae170] [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: 09/10/2024] [Accepted: 12/01/2024] [Indexed: 02/11/2025] Open
Abstract
OBJECTIVES Protein-truncating variants in the TTN gene are a well-established cause of dilated cardiomyopathy (DCM). We report a novel case of DCM caused by a mobile element insertion (MEI) in TTN, through which we highlight the key features of MEIs in next-generation sequencing data. Because of the rarity of MEIs, the next-generation sequencing data features associated with these events may be mistaken as noise, potentially leading to missed diagnoses. METHODS Next-generation sequencing gene panel testing for DCM was performed on a 17-year-old male patient presenting with severe left ventricular dilatation and systolic dysfunction. Manta was used for structural variant detection, followed by manual review of NGS data for potential structural variants. RESULTS Manta detected a potential insertion in TTN. Manual review identified hallmark features consistent with a LINE-1 MEI. This finding was orthogonally confirmed by long-range polymerase chain reaction and gel electrophoresis, which indicated an insertion of approximately 4 to 5 kilobase pairs. The insertion disrupted the reading frame of TTN within an A-band exon, resulting in protein truncation that was classified as likely pathogenic. CONCLUSIONS This case expands the mutational spectrum of TTN protein-truncating variants. It also underscores the importance of recognizing rarer types of pathogenic variants (eg, MEIs) to produce accurate genetic diagnostics.
Collapse
Affiliation(s)
- Qiliang Ding
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Jenna Fine
- Pediatric Critical Care Medicine, Goryeb Children's Hospital, Morriston, NJ, US
| | - Frank T Hoffman
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Michelle L Kluge
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Rhonda K Kuennen
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Sarah M Thieke
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Nicole L Hoppman
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Cherisse A Marcou
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Ross A Rowsey
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Erik C Thorland
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Linnea M Baudhuin
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Ann M Moyer
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| | - Alessia Buglioni
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, US
| |
Collapse
|
3
|
Melo M, Ribeiro M, Silva PF, Valente S, Alves F, Venâncio M, Sequeiros J, Freixo JP, Antunes D, Oliveira J. Medically Actionable Secondary Findings from Whole-Exome Sequencing (WES) Data in a Sample of 3972 Individuals. Int J Mol Sci 2025; 26:3509. [PMID: 40332002 PMCID: PMC12027037 DOI: 10.3390/ijms26083509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
The application of whole-exome sequencing (WES) for diagnostic purposes has the potential to unravel secondary findings unrelated with the primary reason of testing. Some of those might be of high clinical utility and comprise disease-causing variants in genes, related to life-threatening and clinically actionable diseases. Clarifying the allelic frequencies of such variants in specific populations is a crucial step for the large-scale deployment of genomic medicine. We analysed medically relevant variants in the 81 genes from the American College of Medical Genetics and Genomics (ACMG) v3.2 list of actionable loci, using WES data from a diagnostic laboratory cohort of 3972 persons, tentatively resampled to represent the Portuguese population geographic distribution. We identified medically actionable variants in 6.2% of our cohort, distributed across several disease domains: cardiovascular disorders (3.0%), cancer predisposition (2.0%), miscellaneous disorders (1.1%), and metabolic disorders (0.1%). Additionally, we estimated a frequency of heterozygotes for recessive disease alleles of 11.1%. Overall, our results suggest that medically actionable findings can be identified in approximately 6.2% of persons from our population. This is the first study estimating medically actionable findings in Portugal. These results provide valuable insight for patients, healthcare providers, and policymakers involved in advancing genomic medicine at the national and international level.
Collapse
Affiliation(s)
- Mafalda Melo
- Medical Genetics Unit, Hospital Dona Estefânia, Unidade Local de Saúde de Sao José, 1169-045 Lisbon, Portugal (D.A.)
| | - Mariana Ribeiro
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Paulo Filipe Silva
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Susana Valente
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Filipe Alves
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Margarida Venâncio
- Medical Genetics Unit, Hospital Dona Estefânia, Unidade Local de Saúde de Sao José, 1169-045 Lisbon, Portugal (D.A.)
| | - Jorge Sequeiros
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
- ICBAS School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine (UMIB), ICBAS/ITR-Laboratory for Integrative and Translational Research in Population Health, University of Porto, 4050-313 Porto, Portugal
| | - João Parente Freixo
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Diana Antunes
- Medical Genetics Unit, Hospital Dona Estefânia, Unidade Local de Saúde de Sao José, 1169-045 Lisbon, Portugal (D.A.)
- NOVA National School of Public Health (ENSP), NOVA University Lisbon, 1600-560 Lisbon, Portugal
| | - Jorge Oliveira
- Centre for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), 4200-135 Porto, Portugal; (M.R.); (P.F.S.); (S.V.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
- ICBAS School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine (UMIB), ICBAS/ITR-Laboratory for Integrative and Translational Research in Population Health, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
4
|
Ma W, Wright DL, Parra O, Shah ND, Black CC, Baker ML, Khan WA. Identification of novel TTN gene variant in a patient exhibiting severe dilated cardiomyopathy co-occurring with acute fibrinoid organizing pneumonia. Am J Clin Pathol 2025; 163:102-108. [PMID: 39180762 DOI: 10.1093/ajcp/aqae100] [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: 01/05/2024] [Accepted: 07/08/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVES Dilated cardiomyopathy (DCM) is often hereditary, with 20% to 40% of nonischemic cases showing familial linkage, yet genetic testing is underused. This report describes an unreported pathogenic nonsense variant in the Titin (TTN) gene (NM_001267550.2:c.92603G>A) in a 24-year-old man with severe DCM and acute fibrinoid organizing pneumonia, highlighting a unique cardiopulmonary pathology. METHODS We conducted detailed gross, histopathologic, immunophenotypic, and exome-based DNA sequencing analysis in the workup of this case. We also included the patient's clinical and radiologic findings in our study. RESULTS With rapid clinical deterioration and complex comorbidities, including substance abuse and psychiatric conditions, which precluded transplantation, the patient's cardiac function progressively worsened. Autopsy findings included extreme cardiomegaly, biventricular hypertrophy, and acute and chronic pericarditis. Significant pulmonary pathology consistent with acute fibrinoid organizing pneumonia was also noted. Molecular testing confirmed a deleterious maternally inherited TTN variant that was absent in the sibling of the proband and the extant medical literature, highlighting its rarity and significance. CONCLUSIONS This case contributes to the ongoing body of work on the impact of TTN variants on DCM. It suggests a potential link between genetic variants and complex cardiac injury patterns, emphasizing the need for further investigation into the interplay between cardiomyopathy and pulmonary pathology.
Collapse
Affiliation(s)
- Weijie Ma
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Dana L Wright
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Ourania Parra
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Nidhi D Shah
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Candice C Black
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Michael L Baker
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Wahab A Khan
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| |
Collapse
|
5
|
Yang H, Wang Z, Xu Y, Du Y, Yang H, Lu Y. Prognostic signature and therapeutic drug identification for dilated cardiomyopathy based on necroptosis via bioinformatics and experimental validation. Sci Rep 2025; 15:319. [PMID: 39747333 PMCID: PMC11696111 DOI: 10.1038/s41598-024-83455-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Necroptosis, a type of programmed cell death, has been increasingly linked to cardiovascular disease development, yet its role in dilated cardiomyopathy (DCM) remains unclear. In this study, we analyzed the GSE5406 dataset from the GEO database to explore necroptosis-related prognostic signatures in DCM using LASSO regression. We identified five necroptosis-related genes (BID, CAMK2B, GLUL, HSP90AB1, CHMP5) that define a necroptosis-related signature with strong predictive value, evidenced by ROC curve areas of 0.852 and 0.957 in training and test sets, respectively. Our analyses, including GO and GSEA enrichment, focused on pathways associated with high necroptosis-related scores (NRS) and revealed significant immune cell infiltration. Notably, nTreg and iTreg cells were enriched in the high NRS group, while CD8 naive T cells and CD8 T cells positively correlated with NRS. Small molecule drugs fenofibrate, procyclidine, and tienilic acid emerged as potential therapeutic agents for high-risk patients, with fenofibrate showing efficacy in inhibiting DCM progression in an inflammatory animal model. These findings underscore the clinical relevance of necroptosis-related genes in assessing DCM progression and prognosis and highlight their potential for targeted therapeutic development.
Collapse
Affiliation(s)
- Han Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawei Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yimei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Haibo Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yang Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
6
|
Tang Q, Meng X, Tu X, Zhang J. Mendelian Randomization Study on the Associations Between Genetically Predicted Cardiovascular Disease Subtypes and the Risk of Developing Cardiomyopathies. Clin Appl Thromb Hemost 2025; 31:10760296251328011. [PMID: 40152048 PMCID: PMC11951890 DOI: 10.1177/10760296251328011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 02/15/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiomyopathies are commonly believed to have genetic origins; however, the connection between cardiomyopathies and cardiovascular diseases remains uncertain. Thus, we employed a Mendelian randomization (MR) approach to investigate the potential causal effects of specific cardiovascular disease subtypes on dilated and hypertrophic cardiomyopathies, focusing primarily on a European population. Summary-level data for cardiomyopathies and other cardiovascular diseases were obtained from public genome-wide association studies. Random-effects inverse-variance weighting was used as the primary analysis, whereas sensitivity analyses, including weighted median, MR-Egger, and multivariable MR methods, were also conducted. A genetic predisposition to atrial fibrillation [odds ratio (OR): 1.33; 95% confidence interval (CI): 1.18-1.50; P < 0.001], heart failure (OR: 3.22; 95% CI: 1.92-5.41; P < 0.001), and hypertension (OR: 1.50; 95% CI: 1.25-1.81; P < 0.001) were causally linked to an increased risk of developing dilated cardiomyopathy. However, there was no direct causal connection between genetically predicted coronary heart disease, pulmonary embolism, or ischemic stroke and the risk of developing dilated cardiomyopathy. In contrast, no significant associations were found between genetically predicted CVD subtypes and the risk of developing hypertrophic cardiomyopathy. Genetically predicted heart failure is significantly associated with the risk of developing dilated cardiomyopathy, underscoring the importance of effective heart failure management for risk prevention. Moreover, individuals with hypertension and atrial fibrillation might have an increased predisposition to dilated cardiomyopathy, highlighting crucial implications for management.
Collapse
Affiliation(s)
- Qiaolin Tang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiangzhu Meng
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiaowen Tu
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Jian Zhang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| |
Collapse
|
7
|
Croon PM, van Vugt M, Allaart CP, Ruijsink B, Elliott PM, Asselbergs FW, Khera R, Bezzina CR, Winter M, Schmidt AF. Cardiac magnetic resonance markers of pre-clinical hypertrophic and dilated cardiomyopathy in genetic variant carriers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.15.24317368. [PMID: 39606349 PMCID: PMC11601774 DOI: 10.1101/2024.11.15.24317368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Patients with hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) exhibit structural and functional cardiac abnormalities. We aimed to identify imaging biomarkers for pre-clinical cardiomyopathy in healthy individuals carrying cardiomyopathy-associated variants (G+). Methods We included 40,169 UK biobank participants without cardiac disease who had cardiac magnetic resonance imaging (CMR) measurements and whole exome sequencing. We first validated 22 CMR measurements by associating them with incident atrial fibrillation (AF) or heart failure (HF). We subsequently assessed associations of these measurements with HCM or DCM G+, or specific genes, utilising generalised linear models conditional on cardiac risk factors. Results Thirteen CMR measurements were associated with incident AF and fifteen with HF. These included left-ventricular (LV) ejection fraction (EF) (hazard ratio [HR] 0.61, 95% confidence interval [95%CI] 0.54; 0.69) for HF and indexed maximum left atrial volume (LA-Vi max; HR1.47, 95%CI 1.29; 1.67) for AF. Five measurements associated with HCM G+, amongst which right ventricular (RV) end-systolic volume (RV-ESV; OR 0.62, 95%CI 0.53; 0.74), RV-EF (OR 1.36, 95%CI 1.19; 1.55), and right atrial EF (OR 1.22, 95%CI 1.08; 1.39). All associations overlapping with incident disease and HCM had opposite effect directions, such as RV-ESV with HF (OR 1.22, 95%CI 1.07; 1.40). Two CMR measurements associated with DCM G+: LV-ESVi (OR 1.35, 95%CI 1.15; 1.58) and LV-EF (OR 0.75, 95%CI 0.64; 0.88). Due to heterogeneity, we explored associations with individual cardiomyopathy genes, finding MAPSE associated with TTN and TNNT2, and LA pump and RA-EF associated with MYH7. Conclusion We identified right heart CMR measurements associated with HCM G+ in healthy individuals, indicating early compensation of cardiac function. LV measurements were associated with DCM G+, where the CMR associations varied across individual DCM genes, suggesting distinct early pathophysiology.
Collapse
Affiliation(s)
- Philip M. Croon
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Marion van Vugt
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, United Kingdom
| | - Cornelis P. Allaart
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Bram Ruijsink
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Perry M. Elliott
- Barts Heart Centre, St. Bartholomew’s Hospital, London, United Kingdom
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Folkert W. Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, University College London, London, UK
- National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, University College London, London, UK
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT
| | - Connie R. Bezzina
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Michiel Winter
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Floriaan Schmidt
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, United Kingdom
- UCL British Heart Foundation Research Accelerator, London, United Kingdom
| |
Collapse
|
8
|
Ameer SF, Elsaka M, Kahtoon S, Kerzabi RI, Casu G, Giordo R, Zayed H, Pintus G. Exploring the role of exosomes in the pathogenesis and treatment of cardiomyopathies: A comprehensive literature review. Life Sci 2024; 357:123063. [PMID: 39299384 DOI: 10.1016/j.lfs.2024.123063] [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: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Exosomes, a subset of small extracellular vesicles that play a crucial role in intercellular communication, have garnered significant attention for their potential applications in the diagnosis and treatment of cardiomyopathies. Cardiomyopathies, which encompass a spectrum of heart muscle disorders, present complex challenges in diagnosis and management. Understanding the role of exosomes in the etiology of cardiomyopathies such as dilated cardiomyopathy (DCM), restrictive cardiomyopathy (RCM), arrhythmogenic cardiomyopathy (AC), and hypertrophic cardiomyopathy (HCM) may open new possibilities for therapeutic intervention and diagnosis. Exosomes have indeed demonstrated promise as diagnostic biomarkers, particularly in identifying cardiac conditions such as atrial fibrillation (AF) and in the timely classification of high-risk patients with different forms of cardiomyopathy. In DCM, exosomes have been implicated in mediating pathological responses in cardiomyocytes, potentially exacerbating disease progression. Moreover, in RCM, AC, and HCM, exosomes present significant potential as diagnostic biomarkers and therapeutic targets, offering insights into disease pathogenesis and potential avenues for intervention. Understanding the influence of exosomes on disease progression and identifying the specific molecular pathways involved in cardiomyopathy pathogenesis may significantly advance diagnostic and treatment strategies. While key findings highlight the multifaceted role of exosomes in cardiomyopathy, they also emphasize the need for further research to elucidate molecular mechanisms and translate findings into clinical practice. This review highlights the evolving landscape of exosome research in cardiomyopathies and underscores the importance of ongoing investigations to harness the full potential of exosomes in improving patient outcomes.
Collapse
Affiliation(s)
- Shadiya Fawzul Ameer
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Manar Elsaka
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Summaiya Kahtoon
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Rabia-Illhem Kerzabi
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Gavino Casu
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy
| | - Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy.
| |
Collapse
|
9
|
Bottillo I, Giordano C, Ciccone MP, Pignataro MG, Albi F, Parisi G, Formicola D, Grotta S, Ranocchi F, Giuli MV, Checquolo S, Masuelli L, Re F, Majore S, d'Amati G, Grammatico P. Dilated cardiomyopathy due to a novel combination of TTN and BAG3 genetic variants: From acute heart failure to subclinical phenotypes. Cardiovasc Pathol 2024; 73:107675. [PMID: 39059779 DOI: 10.1016/j.carpath.2024.107675] [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: 12/31/2023] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is defined as left ventricular enlargement accompanied by systolic dysfunction not explained by abnormal loading conditions or coronary heart disease. The DCM clinical spectrum is broad, ranging from subclinical to severe presentation with progression to end stage heart failure. To date, different genetic loci have been found to have moderate/definitive evidence for causality in DCM and pathogenic variants in the TTN gene represent the main genetic determinant. Here, we describe a family in which the co-occurrence of two genetic hits, one in the TTN and one in the BAG3 gene, was associated with heterogeneous clinical presentation ranging from subclinical phenotypes to acute cardiogenic shock mimicking fulminant myocarditis. We hypothesize that at least some specific BAG3 genotypes could be related to DCM presenting with acute heart failure and suggest that patients and relatives carrying BAG3 pathogenic variants should be addressed to a tertiary-level heart care center.
Collapse
Affiliation(s)
- Irene Bottillo
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy.
| | - Carla Giordano
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Maria Pia Ciccone
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Maria Gemma Pignataro
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Fiammetta Albi
- Cardiology Division, Cardiac Arrhythmia Center and Cardiomyopathies Unit, San Camillo Forlanini Hospital, Rome, Italy
| | - Gabriella Parisi
- Department of Clinical Microbiology and Virology, San Camillo-Forlanini Hospital, Rome, Italy
| | - Daniela Formicola
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Simona Grotta
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federico Ranocchi
- Cardiac Surgery and Heart Transplantation Unit, San Camillo Hospital, Rome, Italy
| | - Maria Valeria Giuli
- Department of Medico-surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| | - Saula Checquolo
- Department of Medico-surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Re
- Cardiology Division, Cardiac Arrhythmia Center and Cardiomyopathies Unit, San Camillo Forlanini Hospital, Rome, Italy
| | - Silvia Majore
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Giulia d'Amati
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Paola Grammatico
- Division of Medical Genetics, Department of Experimental Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
10
|
Bhide S, Chandran S, Rajasekaran NS, Melkani GC. Genetic and Pathophysiological Basis of Cardiac and Skeletal Muscle Laminopathies. Genes (Basel) 2024; 15:1095. [PMID: 39202453 PMCID: PMC11354015 DOI: 10.3390/genes15081095] [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: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Nuclear lamins, a type V intermediate filament, are crucial components of the nuclear envelope's inner layer, maintaining nuclear integrity and mediating interactions between the nucleus and cytoplasm. Research on human iPSC-derived cells and animal models has demonstrated the importance of lamins in cardiac and skeletal muscle development and function. Mutations in lamins result in laminopathies, a group of diseases including muscular dystrophies, Hutchison-Gilford progeria syndrome, and cardiomyopathies with conduction defects. These conditions have been linked to disrupted autophagy, mTOR, Nrf2-Keap, and proteostasis signaling pathways, indicating complex interactions between the nucleus and cytoplasm. Despite progress in understanding these pathways, many questions remain about the mechanisms driving lamin-induced pathologies, leading to limited therapeutic options. This review examines the current literature on dysregulated pathways in cardiac and skeletal muscle laminopathies and explores potential therapeutic strategies for these conditions.
Collapse
Affiliation(s)
- Shruti Bhide
- Department of Biology, Molecular Biology Institute, San Diego State University, San Diego, CA 92182, USA; (S.B.); (S.C.)
| | - Sahaana Chandran
- Department of Biology, Molecular Biology Institute, San Diego State University, San Diego, CA 92182, USA; (S.B.); (S.C.)
| | - Namakkal S. Rajasekaran
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35294, USA;
| | - Girish C. Melkani
- Department of Biology, Molecular Biology Institute, San Diego State University, San Diego, CA 92182, USA; (S.B.); (S.C.)
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35294, USA;
| |
Collapse
|
11
|
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: 1] [Impact Index Per Article: 1.0] [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
|
12
|
Arnautu DA, Cozma D, Lala IR, Arnautu SF, Tomescu MC, Andor M. Risk Assessment and Personalized Treatment Options in Inherited Dilated Cardiomyopathies: A Narrative Review. Biomedicines 2024; 12:1643. [PMID: 39200108 PMCID: PMC11351202 DOI: 10.3390/biomedicines12081643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Considering the worldwide impact of heart failure, it is crucial to develop approaches that can help us comprehend its root cause and make accurate predictions about its outcome. This is essential for lowering the suffering and death rates connected with this widespread illness. Cardiomyopathies frequently result from genetic factors, and the study of heart failure genetics is advancing quickly. Dilated cardiomyopathy (DCM) is the most prevalent kind of cardiomyopathy, encompassing both genetic and nongenetic abnormalities. It is distinguished by the enlargement of the left ventricle or both ventricles, accompanied by reduced contractility. The discovery of the molecular origins and subsequent awareness of the molecular mechanism is broadening our knowledge of DCM development. Additionally, it emphasizes the complicated nature of DCM and the necessity to formulate several different strategies to address the diverse underlying factors contributing to this disease. Genetic variants that can be transmitted from one generation to another can be a significant contributor to causing family or sporadic hereditary DCM. Genetic variants also play a significant role in determining susceptibility for acquired triggers for DCM. The genetic causes of DCM can have a large range of phenotypic expressions. It is crucial to select patients who are most probable to gain advantages from genetic testing. The purpose of this research is to emphasize the significance of identifying genetic DCM, the relationships between genotype and phenotype, risk assessment, and personalized therapy for both those affected and their relatives. This approach is expected to gain importance once treatment is guided by genotype-specific advice and disease-modifying medications.
Collapse
Affiliation(s)
- Diana-Aurora Arnautu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dragos Cozma
- Department of Cardiology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioan-Radu Lala
- Department of Cardiology, Western University Vasile Goldis, 310025 Arad, Romania
| | - Sergiu-Florin Arnautu
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mirela-Cleopatra Tomescu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Minodora Andor
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
13
|
Li Y, Ma K, Dong Z, Gao S, Zhang J, Huang S, Yang J, Fang G, Li Y, Li X, Welch C, Griffin EL, Ramaswamy P, Valivullah Z, Liu X, Dong J, Wang DW, Du, Chung WK, Li Y. Frameshift variants in C10orf71 cause dilated cardiomyopathy in human, mouse, and organoid models. J Clin Invest 2024; 134:e177172. [PMID: 38950288 PMCID: PMC11178530 DOI: 10.1172/jci177172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Ke Ma
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zhujun Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jing Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jie Yang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Guangming Fang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Yujie Li
- Novogene Co. Ltd., Beijing, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Carrie Welch
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Emily L. Griffin
- Department of Pediatrics, Columbia University, New York, New York, USA
| | | | | | | | - Jianzeng Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Wendy K. Chung
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
14
|
León P, Franco P, Hinojosa N, Torres K, Moreano A, Romero VI. TTN novel splice variant in familial dilated cardiomyopathy and splice variants review: a case report. Front Cardiovasc Med 2024; 11:1387063. [PMID: 38938651 PMCID: PMC11210389 DOI: 10.3389/fcvm.2024.1387063] [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: 02/16/2024] [Accepted: 05/03/2024] [Indexed: 06/29/2024] Open
Abstract
This case report details the identification of a novel likely pathogenic splicing variant in the TTN gene, associated with dilated cardiomyopathy (DCM), in a 42-year-old male patient presenting with early-onset heart failure and reduced ejection fraction. DCM is a nonischemic heart condition characterized by left biventricular dilation and systolic dysfunction, with approximately one-third of cases being familial and often linked to genetic mutations. The TTN gene, encoding the largest human protein essential for muscle contraction and sarcomere structure, is implicated in about 25% of DCM cases through mutations, especially truncating variants. Our investigation revealed a previously unreported G > C mutation at the splice acceptor site in intron 356 of TTN, confirmed by Sanger sequencing and not found in population databases, suggesting a novel contribution to the understanding of DCM etiology. The case emphasizes the critical role of the TTN gene in cardiac function and the genetic complexity underlying DCM. A comprehensive literature review highlighted the prevalence and significance of splice variants in the TTN gene, particularly those affecting the titin A-band, which is known for its role in muscle contraction and stability. This variant's identification underscores the importance of genetic screening in patients with DCM, offering insights into the disease's familial transmission and potential therapeutic targets. Our findings contribute to the expanding knowledge of genetic factors in DCM, demonstrating the necessity of integrating genetic diagnostics in cardiovascular medicine. This case supports the growing evidence linking splicing mutations in specific regions of the TTN gene to DCM development and underscores the importance of genetic counseling and testing in managing heart disease.
Collapse
Affiliation(s)
- Paul León
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, Quito, Ecuador
| | - Paula Franco
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Nicole Hinojosa
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Kevin Torres
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Andrés Moreano
- Department of Cardiology, Universidad de Sao Paulo, Sao Paulo, Brazil
| | - Vanessa I. Romero
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, Quito, Ecuador
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| |
Collapse
|
15
|
Fan Z, Wu C, Wang C, Liu C, Fang L, Ma L, Zou W, Yuan B, Ji Z, Cai B, Liu G. Impact of Concurrent Ischaemic Stroke on Unfavourable Outcomes in Men and Women with Dilated Cardiomyopathy. Rev Cardiovasc Med 2024; 25:215. [PMID: 39076319 PMCID: PMC11270057 DOI: 10.31083/j.rcm2506215] [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: 10/09/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 07/31/2024] Open
Abstract
Background Growing evidence suggests that concurrent ischaemic stroke (IS) exacerbates the prognosis of patients with dilated cardiomyopathy (DCM) and that this effect may be further influenced by sex. However, the exact effect of sex remains unclear. This study aimed to explore the effects of the relevant risk factors on the prognosis of patients with DCM and concurrent IS. Considering the sex differences in DCM, this study further investigated the impact of concurrent IS on the prognosis of men and women with DCM. Methods A total of 632 patients with DCM enrolled between 2016 and 2021 were included in this study. Clinical data were obtained from medical records, and all participants were followed up in the outpatient clinic or by telephone for at least 1 year. A Cox proportional hazards model and Kaplan-Meier curves were used to evaluate the effects of concurrent IS on the prognosis of patients with DCM. Results Patients with DCM complicated with IS (DCM-IS) had significantly lower cumulative survival rates than patients with DCM without IS (non-IS) (74.6% vs. 84.2%, χ 2 = 6.85, p = 0.009). Additionally, IS was associated with greater risks of death and heart transplantation (HTx) in men (75.8% vs. 85.1%, χ 2 = 5.02, p = 0.025), but not in women (71.0% vs. 81.5%, χ 2 = 1.91, p = 0.167). Conclusions This large-scale multicentre prospective cohort study demonstrated a poorer prognosis in patients with concurrent DCM and IS, particularly among men. Patients with DCM should not be overlooked in IS screening, emphasis should be placed on the occurrence of IS in patients with DCM. Early and proactive secondary prevention of cerebrovascular diseases might improve the prognosis of DCM patients. More intervention studies focusing on men with DCM complicated with IS should be prioritised.
Collapse
Affiliation(s)
- Zexin Fan
- Department of Neurology, The Second Hospital of Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Chao Wu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chaobin Wang
- Department of Neurology, Beijing Fangshan District Liangxiang Hospital, 102401 Beijing, China
| | - Chun Liu
- Department of Neurology, Mechinka Hospital, Dnipro State Medical University, 49044 Dnipro, Ukraine
| | - Libo Fang
- Department of Neurology, Beijing Fuxing Hospital, Capital Medical University, 100038 Beijing, China
| | - Lin Ma
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Wenlong Zou
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Boyi Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Zeyu Ji
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Bin Cai
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Guangzhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
16
|
Bougaran P, Bautch VL. Life at the crossroads: the nuclear LINC complex and vascular mechanotransduction. Front Physiol 2024; 15:1411995. [PMID: 38831796 PMCID: PMC11144885 DOI: 10.3389/fphys.2024.1411995] [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: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Vascular endothelial cells line the inner surface of all blood vessels, where they are exposed to polarized mechanical forces throughout their lifespan. Both basal substrate interactions and apical blood flow-induced shear stress regulate blood vessel development, remodeling, and maintenance of vascular homeostasis. Disruption of these interactions leads to dysfunction and vascular pathologies, although how forces are sensed and integrated to affect endothelial cell behaviors is incompletely understood. Recently the endothelial cell nucleus has emerged as a prominent force-transducing organelle that participates in vascular mechanotransduction, via communication to and from cell-cell and cell-matrix junctions. The LINC complex, composed of SUN and nesprin proteins, spans the nuclear membranes and connects the nuclear lamina, the nuclear envelope, and the cytoskeleton. Here we review LINC complex involvement in endothelial cell mechanotransduction, describe unique and overlapping functions of each LINC complex component, and consider emerging evidence that two major SUN proteins, SUN1 and SUN2, orchestrate a complex interplay that extends outward to cell-cell and cell-matrix junctions and inward to interactions within the nucleus and chromatin. We discuss these findings in relation to vascular pathologies such as Hutchinson-Gilford progeria syndrome, a premature aging disorder with cardiovascular impairment. More knowledge of LINC complex regulation and function will help to understand how the nucleus participates in endothelial cell force sensing and how dysfunction leads to cardiovascular disease.
Collapse
Affiliation(s)
- Pauline Bougaran
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
| | - Victoria L. Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Scheel PJ, Cartella I, Murray B, Gilotra NA, Ammirati E. Role of genetics in inflammatory cardiomyopathy. Int J Cardiol 2024; 400:131777. [PMID: 38218248 DOI: 10.1016/j.ijcard.2024.131777] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Traditional cardiomyopathy paradigms segregate inflammatory etiologies from those caused by genetic variants. An identified or presumed trigger is implicated in acute myocarditis or chronic inflammatory cardiomyopathy but growing evidence suggests a significant proportion of patients have an underlying cardiomyopathy-associated genetic variant often even when a clear inflammatory trigger is identified. Recognizing a possible genetic contribution to inflammatory cardiomyopathy may have major downstream implications for both the patient and family. The presenting features of myocarditis (i.e. chest pain, arrhythmia, and/or heart failure) may provide insight into diagnostic considerations. One example is isolated cardiac sarcoidosis, a distinct inflammatory cardiomyopathy that carries diagnostic challenges and clinical overlap; genetic testing has increasingly reclassified cases of isolated cardiac sarcoidosis as genetic cardiomyopathy, notably altering management. On the other side, inflammatory presentations of genetic cardiomyopathies are likewise underappreciated and a growing area of investigation. Inflammation plays an important role in the pathogenesis of several familial cardiomyopathies, especially arrhythmogenic phenotypes. Given these clinical scenarios, and the implications on clinical decision making such as initiation of immunosuppression, sudden cardiac death prevention, and family screening, it is important to recognize when genetics may be playing a role.
Collapse
Affiliation(s)
- Paul J Scheel
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA.
| | - Iside Cartella
- De Gasperis Cardio Center, Transplant Center, Niguarda Hospital, Milano, Italy; Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA
| | - Nisha A Gilotra
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA
| | - Enrico Ammirati
- De Gasperis Cardio Center, Transplant Center, Niguarda Hospital, Milano, Italy; Department of Health Sciences, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
18
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Yang H, Gong K, Luo Y, Wang L, Tan Z, Yao Y, Xie L. Case report: A new de novo mutation of the Troponin T2 gene in a Chinese patient with dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1288328. [PMID: 38054088 PMCID: PMC10694197 DOI: 10.3389/fcvm.2023.1288328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a cardiovascular disease characterized by persistent ventricular dilatation and systolic dysfunction. DCM has a variety of causes, including myocarditis; exposure to narcotics, alcohol, or other toxins; and metabolic or endocrine disorders. Genetic factors play a dominant role in 30%-40% of DCM cases. Here, we report a case of DCM with very severe heart failure. Because of the severity of heart failure, the patient underwent heart transplantation. We speculated that the patient's DCM might be due to a mutation; hence, we performed whole-exome sequencing of the patient and their parents, which showed a de novo heterozygous mutation (NM_001001431.2c.769G>A:p.E257K) in TNNT2, which was considered pathogenic according to the ACMG pathogenicity assessment. This finding expands the genetic map of DCM and TNNT2 and will be important for future studies on the genetic and disease relationships between DCM and TNNT2.
Collapse
Affiliation(s)
- Huan Yang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Ke Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yong Luo
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Lei Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Zhiping Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yao Yao
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| |
Collapse
|
20
|
Hawey C, Bourque K, Alim K, Derish I, Rody E, Khan K, Gendron N, Cecere R, Giannetti N, Hébert TE. Measuring Single-Cell Calcium Dynamics Using a Myofilament-Localized Optical Biosensor in hiPSC-CMs Derived from DCM Patients. Cells 2023; 12:2526. [PMID: 37947605 PMCID: PMC10647603 DOI: 10.3390/cells12212526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Synchronized contractions of cardiomyocytes within the heart are tightly coupled to electrical stimulation known as excitation-contraction coupling. Calcium plays a key role in this process and dysregulated calcium handling can significantly impair cardiac function and lead to the development of cardiomyopathies and heart failure. Here, we describe a method and analytical technique to study myofilament-localized calcium signaling using the intensity-based fluorescent biosensor, RGECO-TnT. Dilated cardiomyopathy is a heart muscle disease that negatively impacts the heart's contractile function following dilatation of the left ventricle. We demonstrate how this biosensor can be used to characterize 2D hiPSC-CMs monolayers generated from a healthy control subject compared to two patients diagnosed with dilated cardiomyopathy. Lastly, we provide a step-by-step guide for single-cell data analysis and describe a custom Transient Analysis application, specifically designed to quantify features of calcium transients. All in all, we explain how this analytical approach can be applied to phenotype hiPSC-CM behaviours and stratify patient responses to identify perturbations in calcium signaling.
Collapse
Affiliation(s)
- Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Karima Alim
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Ida Derish
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Elise Rody
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Kashif Khan
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Natalie Gendron
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Renzo Cecere
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Nadia Giannetti
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| |
Collapse
|
21
|
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
|
22
|
Bui QM, Ding J, Hong KN, Adler EA. The Genetic Evaluation of Dilated Cardiomyopathy. STRUCTURAL HEART : THE JOURNAL OF THE HEART TEAM 2023; 7:100200. [PMID: 37745678 PMCID: PMC10512006 DOI: 10.1016/j.shj.2023.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 09/26/2023]
Abstract
Dilated cardiomyopathy (DCM) is a common cause of heart failure and is the primary indication for heart transplantation. A genetic etiology can be found in 20-35% of patients with DCM, especially in those with a family history of cardiomyopathy or sudden cardiac death at an early age. With advancements in genome sequencing, the understanding of genotype-phenotype relationships in DCM has expanded with over 60 genes implicated in the disease. Subsequently, these findings have increased adoption of genetic testing in the management of DCM, which has allowed for improved risk stratification and identification of at risk family members. In this review, we discuss the genetic evaluation of DCM with a focus on practical genetic testing considerations, genotype-phenotype associations, and insights into upcoming personalized therapies.
Collapse
Affiliation(s)
- Quan M. Bui
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Jeffrey Ding
- University of California San Diego School of Medicine, La Jolla, California, USA
| | - Kimberly N. Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eric A. Adler
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
23
|
Mansur ADP, Pereira-Barretto AC, del Carlo CH, Avakian SD, Nakagawa NK, Cesar LAM, Bocchi EA. Sex Differences in Prognosis of Heart Failure Due to Ischemic and Nonischemic Cardiomyopathy. J Clin Med 2023; 12:5323. [PMID: 37629365 PMCID: PMC10456101 DOI: 10.3390/jcm12165323] [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: 06/26/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Limited research has explored sex-specific differences in death predictors of HF patients with ischemic (iCMP) and nonischemic (niCMP) cardiomyopathy. This study assessed sex differences in niCMP and iCMP prognosis. METHODS We studied 7487 patients with HF between February 2017 and September 2020. Clinical features and echocardiographic findings were collected. We used Kaplan-Meier, Cox proportional hazard models, and chi-square scores of Cox regression to determine death predictors in women and men. RESULTS The mean age was 64.3 ± 14.2 years, with 4417 (59%) males. Women with iCMP and niCMP exhibited a significantly higher mean age, higher mean left ventricular ejection fraction, and smaller left ventricular diastolic diameter than men. Over 2.26 years of follow-up, 325 (14.7%) women and 420 (15.7%) men, and 211 women (24.5%) and 519 men (29.8%) with niCMP (p = NS) and iCMP (p = 0.004), respectively, died. The cumulative incidence of death was higher in men with iCMP (log-rank p < 0.0001) but similar with niCMP. Cox regression showed chronic kidney disease, diabetes, stroke, atrial fibrillation, age, and myocardial infarction as the main predictors of death for iCMP in women and men. CONCLUSIONS Women exhibited a better prognosis than men with iCMP, but similar for niCMP. Nevertheless, sex was not an independent predictor of death for both CMP.
Collapse
Affiliation(s)
- Antonio de Padua Mansur
- Serviço de Prevencao, Cardiopatia na Mulher e Reabilitação Cardiovascular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Antonio Carlos Pereira-Barretto
- Serviço de Prevencao, Cardiopatia na Mulher e Reabilitação Cardiovascular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Carlos Henrique del Carlo
- Hospital Dia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Solange Desirée Avakian
- Unidade Clinica de Valvopatias, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Naomi Kondo Nakagawa
- Departamento de Fisioterapia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-903, SP, Brazil;
| | - Luiz Antonio Machado Cesar
- Unidade Clinica de Coronariopatias Crônicas, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Edimar Alcides Bocchi
- Unidade Clinica de Insuficiencia Cardiaca, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| |
Collapse
|
24
|
Saha S, Singh P, Dutta A, Vaidya H, Negi PC, Sengupta S, Seth S, Basak T. A Comprehensive Insight and Mechanistic Understanding of the Lipidomic Alterations Associated With DCM. JACC. ASIA 2023; 3:539-555. [PMID: 37614533 PMCID: PMC10442885 DOI: 10.1016/j.jacasi.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 08/25/2023]
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure characterized by the enlargement of the left ventricular cavity and contractile dysfunction of the myocardium. Lipids are the major sources of energy for the myocardium. Impairment of lipid homeostasis has a potential role in the pathogenesis of DCM. In this review, we have summarized the role of different lipids in the progression of DCM that can be considered as potential biomarkers. Further, we have also explained the mechanistic pathways followed by the lipid molecules in disease progression along with the cardioprotective role of certain lipids. As the global epidemiological status of DCM is alarming, it is high time to define some disease-specific biomarkers with greater prognostic value. We are proposing an adaptation of a system lipidomics-based approach to profile DCM patients in order to achieve a better diagnosis and prognosis of the disease.
Collapse
Affiliation(s)
- Shubham Saha
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Abhi Dutta
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Hiteshi Vaidya
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Prakash Chand Negi
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| |
Collapse
|
25
|
Chen K, Shi Y, Zhu H. Analysis of the role of glucose metabolism-related genes in dilated cardiomyopathy based on bioinformatics. J Thorac Dis 2023; 15:3870-3884. [PMID: 37559624 PMCID: PMC10407475 DOI: 10.21037/jtd-23-906] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a prevalent condition with diverse etiologies, including viral infection, autoimmune response, and genetic factors. Despite the crucial role of energy metabolism in cardiac function, therapeutic targets for key genes in DCM's energy metabolism remain scarce. METHODS Our study employed the GSE79962 and GSE42955 datasets from the Gene Expression Omnibus (GEO) database for myocardial tissue sample collection and target gene identification via differential gene expression screening. Using various R packages, GSEA software, and the STRING database, we conducted data analysis, gene set enrichment, and protein-protein interaction predictions. The least absolute shrinkage and selection operator (LASSO) and Support Vector Machine (SVM) algorithms aided in feature gene selection, while the predictive model's efficiency was evaluated via the receiver operating characteristic (ROC) curve analysis. We used the non-negative matrix factorization (NMF) method for molecular typing and the cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm for predicting immune cell infiltration. RESULTS The DLAT and LDHA genes may regulate the immune microenvironment of DCM by influencing activated dendritic cells, activated mast cells, and M0 macrophages, respectively. The BPGM, DLAT, PGM2, ADH1A, ADH1C, LDHA, and PFKM genes may regulate m6A methylation in DCM by affecting the ZC3H13, ALKBH5, RBMX, HNRNPC, METTL3, and YTHDC1 genes. Further regulatory mechanism analysis suggested that PFKM, DLAT, PKLR, PGM2, LDHA, BPGM, ADH1A, and ADH1C could be involved in the development of cardiomyopathy by regulating the Toll-like receptor signaling pathway. CONCLUSIONS PFKM, DLAT, PKLR, PGM2, LDHA, BPGM, ADH1A, and ADH1C may serve as potential targets for guiding the diagnosis, treatment, and follow-up of DCM.
Collapse
Affiliation(s)
- Keping Chen
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Shi
- Operating Room, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Haijie Zhu
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
26
|
Kornienko J, Rodríguez-Martínez M, Fenzl K, Hinze F, Schraivogel D, Grosch M, Tunaj B, Lindenhofer D, Schraft L, Kueblbeck M, Smith E, Mao C, Brown E, Owens A, Saguner AM, Meder B, Parikh V, Gotthardt M, Steinmetz LM. Mislocalization of pathogenic RBM20 variants in dilated cardiomyopathy is caused by loss-of-interaction with Transportin-3. Nat Commun 2023; 14:4312. [PMID: 37463913 DOI: 10.1038/s41467-023-39965-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
Severe forms of dilated cardiomyopathy (DCM) are associated with point mutations in the alternative splicing regulator RBM20 that are frequently located in the arginine/serine-rich domain (RS-domain). Such mutations can cause defective splicing and cytoplasmic mislocalization, which leads to the formation of detrimental cytoplasmic granules. Successful development of personalized therapies requires identifying the direct mechanisms of pathogenic RBM20 variants. Here, we decipher the molecular mechanism of RBM20 mislocalization and its specific role in DCM pathogenesis. We demonstrate that mislocalized RBM20 RS-domain variants retain their splice regulatory activity, which reveals that aberrant cellular localization is the main driver of their pathological phenotype. A genome-wide CRISPR knockout screen combined with image-enabled cell sorting identified Transportin-3 (TNPO3) as the main nuclear importer of RBM20. We show that the direct RBM20-TNPO3 interaction involves the RS-domain, and is disrupted by pathogenic variants. Relocalization of pathogenic RBM20 variants to the nucleus restores alternative splicing and dissolves cytoplasmic granules in cell culture and animal models. These findings provide proof-of-principle for developing therapeutic strategies to restore RBM20's nuclear localization in RBM20-DCM patients.
Collapse
Affiliation(s)
- Julia Kornienko
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | | | - Kai Fenzl
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Hinze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Schraivogel
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Markus Grosch
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brigit Tunaj
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dominik Lindenhofer
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Laura Schraft
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Moritz Kueblbeck
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Eric Smith
- University of Michigan, Ann Arbor, MI, USA
| | - Chad Mao
- Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | | | - Anjali Owens
- University of Pennsylvania, Philadelphia, PA, USA
| | - Ardan M Saguner
- Department of Cardiology, University Heart Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Benjamin Meder
- Cardiogenetics Center Heidelberg, Department of Cardiology, Angiology and Pulmology, University Hospital Heidelberg, Heidelberg, Germany
| | - Victoria Parikh
- Stanford Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Gotthardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lars M Steinmetz
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Genome Technology Center, Palo Alto, CA, USA.
| |
Collapse
|
27
|
Koslow M, Mondaca-Ruff D, Xu X. Transcriptome studies of inherited dilated cardiomyopathies. Mamm Genome 2023; 34:312-322. [PMID: 36749382 PMCID: PMC10426000 DOI: 10.1007/s00335-023-09978-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023]
Abstract
Dilated cardiomyopathy (DCM) is a group of heart muscle diseases that often lead to heart failure, with more than 50 causative genes have being linked to DCM. The heterogenous nature of the inherited DCMs suggest the need of precision medicine. Consistent with this emerging concept, transcriptome studies in human patients with DCM indicated distinct molecular signature for DCMs of different genetic etiology. To facilitate this line of research, we reviewed the status of transcriptome studies of inherited DCMs by focusing on three predominant DCM causative genes, TTN, LMNA, and BAG3. Besides studies in human patients, we summarized transcriptomic analysis of these inherited DCMs in a variety of model systems ranging from iPSCs to rodents and zebrafish. We concluded that the RNA-seq technology is a powerful genomic tool that has already led to the discovery of new modifying genes, signaling pathways, and related therapeutic avenues. We also pointed out that both temporal (different pathological stages) and spatial (different cell types) information need to be considered for future transcriptome studies. While an important bottle neck is the low throughput in experimentally testing differentially expressed genes, new technologies in efficient animal models such as zebrafish starts to be developed. It is anticipated that the RNA-seq technology will continue to uncover both unique and common pathological events, aiding the development of precision medicine for inherited DCMs.
Collapse
Affiliation(s)
- Matthew Koslow
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David Mondaca-Ruff
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
28
|
Wang Y, Dobreva G. Epigenetics in LMNA-Related Cardiomyopathy. Cells 2023; 12:cells12050783. [PMID: 36899919 PMCID: PMC10001118 DOI: 10.3390/cells12050783] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Mutations in the gene for lamin A/C (LMNA) cause a diverse range of diseases known as laminopathies. LMNA-related cardiomyopathy is a common inherited heart disease and is highly penetrant with a poor prognosis. In the past years, numerous investigations using mouse models, stem cell technologies, and patient samples have characterized the phenotypic diversity caused by specific LMNA variants and contributed to understanding the molecular mechanisms underlying the pathogenesis of heart disease. As a component of the nuclear envelope, LMNA regulates nuclear mechanostability and function, chromatin organization, and gene transcription. This review will focus on the different cardiomyopathies caused by LMNA mutations, address the role of LMNA in chromatin organization and gene regulation, and discuss how these processes go awry in heart disease.
Collapse
Affiliation(s)
- Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| |
Collapse
|
29
|
Guevara-Ramírez P, Cadena-Ullauri S, Ibarra-Castillo R, Laso-Bayas JL, Paz-Cruz E, Tamayo-Trujillo R, Ruiz-Pozo VA, Doménech N, Ibarra-Rodríguez AA, Zambrano AK. Genomic analysis of a novel pathogenic variant in the gene LMNA associated with cardiac laminopathies found in Ecuadorian siblings: A case report. Front Cardiovasc Med 2023; 10:1141083. [PMID: 37025686 PMCID: PMC10070725 DOI: 10.3389/fcvm.2023.1141083] [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: 01/09/2023] [Accepted: 03/03/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction Cardiac laminopathies are caused by mutations in the LMNA gene and include a wide range of clinical manifestations involving electrical and mechanical changes in cardiomyocytes. In Ecuador, cardiovascular diseases were the primary cause of death in 2019, accounting for 26.5% of total deaths. Cardiac laminopathy-associated mutations involve genes coding for structural proteins with functions related to heart development and physiology. Family description Two Ecuadorian siblings, self-identified as mestizos, were diagnosed with cardiac laminopathies and suffered embolic strokes. Moreover, by performing Next-Generation Sequencing, a pathogenic variant (NM_170707.3:c.1526del) was found in the gene LMNA. Discussion and conclusion Currently, genetic tests are an essential step for disease genetic counseling, including cardiovascular disease diagnosis. Identification of a genetic cause that may explain the risk of cardiac laminopathies in a family can help the post-test counseling and recommendations from the cardiologist. In the present report, a pathogenic variant ((NM_170707.3:c.1526del) has been identified in two Ecuadorian siblings with cardiac laminopathies. The LMNA gene codes for A-type laminar proteins that are associated with gene transcription regulation. Mutations in the LMNA gene cause laminopathies, disorders with diverse phenotypic manifestations. Moreover, understanding the molecular biology of the disease-causing mutations is essential in deciding the correct type of treatment.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rita Ibarra-Castillo
- Department of Hemodynamics, Clinical Cardiac Electrophysiologist, Quito-Ecuador, Ecuador
| | - José Luis Laso-Bayas
- Department of Hemodynamics, Clinical Cardiac Electrophysiologist, Quito-Ecuador, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Nieves Doménech
- Instituto de Investigación Biomédica de A Coruña (INIBIC)-CIBERCV, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidad da Coruña (UDC), La Coruña-Spain, Spain
| | | | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- Correspondence: Ana Karina Zambrano
| |
Collapse
|
30
|
Filipovic N, Sustersic T, Milosevic M, Milicevic B, Simic V, Prodanovic M, Mijailovic S, Kojic M. SILICOFCM platform, multiscale modeling of left ventricle from echocardiographic images and drug influence for cardiomyopathy disease. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 227:107194. [PMID: 36368295 DOI: 10.1016/j.cmpb.2022.107194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVE In silico clinical trials are the future of medicine and virtual testing and simulation are the future of medical engineering. The use of a computational platform can reduce costs and time required for developing new models of medical devices and drugs. The computational platform, which is one of the main results of the SILICOFCM project, was developed using state-of-the-art finite element modeling for macro simulation of fluid-structure interaction with micro modeling at the molecular level for drug interaction with the cardiac cells. SILICOFCM platform is using for risk prediction and optimal drug therapy of familial cardiomyopathy in a specific patient. METHODS In order to obtain 3D image reconstruction, the U-net architecture was used to determine geometric parameters for the left ventricle which were extracted from the echocardiographic apical and M-mode views. A micro-mechanics cellular model which includes three kinetic processes of sarcomeric proteins interactions was developed. It allows simulation of the drugs which are divided into three major groups defined by the principal action of each drug. Fluid-solid coupling for the left ventricle was presented. A nonlinear material model of the heart wall that was developed by using constitutive curves which include the stress-strain relationship was used. RESULTS The results obtained with the parametric model of the left ventricle where pressure-volume (PV) diagrams depend on the change of Ca2+ were presented. It directly affects the ejection fraction. The presented approach with the variation of the left ventricle (LV) geometry and simulations which include the influence of different parameters on the PV diagrams are directly interlinked with drug effects on the heart function. It includes different drugs such as Entresto and Digoxin that directly affect the cardiac PV diagrams and ejection fraction. CONCLUSIONS Computational platforms such as the SILICOFCM platform are novel tools for risk prediction of cardiac disease in a specific patient that will certainly open a new avenue for in silico clinical trials in the future.
Collapse
Affiliation(s)
- Nenad Filipovic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia.
| | - Tijana Sustersic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| | - Miljan Milosevic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| | - Bogdan Milicevic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| | - Vladimir Simic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| | - Momcilo Prodanovic
- BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| | | | - Milos Kojic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia; BioIRC Bioengineering Research and Development center, Kragujevac, Serbia
| |
Collapse
|
31
|
Ghasemi S, Mahdavi M, Maleki M, Salahshourifar I, Kalayinia S. A novel likely pathogenic variant in the FBXO32 gene associated with dilated cardiomyopathy according to whole‑exome sequencing. BMC Med Genomics 2022; 15:234. [PMID: 36344977 PMCID: PMC9641816 DOI: 10.1186/s12920-022-01388-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Background Familial dilated cardiomyopathy (DCM) is a genetic heart disorder characterized by progressive heart failure and sudden cardiac death. Over 250 genes have been reported in association with DCM; nonetheless, the genetic cause of most DCM patients has been unknown. The goal of the present study was to determine the genetic etiology of familial DCM in an Iranian family. Methods Whole-exome sequencing was performed to identify the underlying variants in an Iranian consanguineous family with DCM. The presence of the candidate variant was confirmed and screened in available relatives by PCR and Sanger sequencing. The pathogenic effect of the candidate variant was assessed by bioinformatics analysis, homology modeling, and docking. Results One novel likely pathogenic deletion, c.884_886del: p.Lys295del, in F-box only protein 32 (muscle-specific ubiquitin-E3 ligase protein; FBXO32) was identified. Based on bioinformatics and modeling analysis, c.884_886del was the most probable cause of DCM in the studied family. Conclusions Our findings indicate that variants in FBXO32 play a role in recessive DCM. Variants in FBXO32 may disturb the degradation of target proteins in the ubiquitin–proteasome system and lead to severe DCM. We suggest considering this gene variants in patients with recessively inherited DCM.
Collapse
|
32
|
Balasooriya GI, Spector DL. Allele-specific differential regulation of monoallelically expressed autosomal genes in the cardiac lineage. Nat Commun 2022; 13:5984. [PMID: 36216821 PMCID: PMC9550772 DOI: 10.1038/s41467-022-33722-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Each mammalian autosomal gene is represented by two alleles in diploid cells. To our knowledge, no insights have been made in regard to allele-specific regulatory mechanisms of autosomes. Here we use allele-specific single cell transcriptomic analysis to elucidate the establishment of monoallelic gene expression in the cardiac lineage. We find that monoallelically expressed autosomal genes in mESCs and mouse blastocyst cells are differentially regulated based on the genetic background of the parental alleles. However, the genetic background of the allele does not affect the establishment of monoallelic genes in differentiated cardiomyocytes. Additionally, we observe epigenetic differences between deterministic and random autosomal monoallelic genes. Moreover, we also find a greater contribution of the maternal versus paternal allele to the development and homeostasis of cardiac tissue and in cardiac health, highlighting the importance of maternal influence in male cardiac tissue homeostasis. Our findings emphasize the significance of allele-specific insights into gene regulation in development, homeostasis and disease.
Collapse
|
33
|
Spurrell CH, Barozzi I, Kosicki M, Mannion BJ, Blow MJ, Fukuda-Yuzawa Y, Slaven N, Afzal SY, Akiyama JA, Afzal V, Tran S, Plajzer-Frick I, Novak CS, Kato M, Lee EA, Garvin TH, Pham QT, Kronshage AN, Lisgo S, Bristow J, Cappola TP, Morley MP, Margulies KB, Pennacchio LA, Dickel DE, Visel A. Genome-wide fetalization of enhancer architecture in heart disease. Cell Rep 2022; 40:111400. [PMID: 36130500 PMCID: PMC9534044 DOI: 10.1016/j.celrep.2022.111400] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/10/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Heart disease is associated with re-expression of key transcription factors normally active only during prenatal development of the heart. However, the impact of this reactivation on the regulatory landscape in heart disease is unclear. Here, we use RNA-seq and ChIP-seq targeting a histone modification associated with active transcriptional enhancers to generate genome-wide enhancer maps from left ventricle tissue from up to 26 healthy controls, 18 individuals with idiopathic dilated cardiomyopathy (DCM), and five fetal hearts. Healthy individuals have a highly reproducible epigenomic landscape, consisting of more than 33,000 predicted heart enhancers. In contrast, we observe reproducible disease-associated changes in activity at 6,850 predicted heart enhancers. Combined analysis of adult and fetal samples reveals that the heart disease epigenome and transcriptome both acquire fetal-like characteristics, with 3,400 individual enhancers sharing fetal regulatory properties. We also provide a comprehensive data resource (http://heart.lbl.gov) for the mechanistic exploration of DCM etiology.
Collapse
Affiliation(s)
- Cailyn H Spurrell
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Iros Barozzi
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Brandon J Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Matthew J Blow
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Yoko Fukuda-Yuzawa
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Neil Slaven
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Sarah Y Afzal
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jennifer A Akiyama
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Veena Afzal
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Stella Tran
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Ingrid Plajzer-Frick
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Catherine S Novak
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Momoe Kato
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Elizabeth A Lee
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Tyler H Garvin
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Quan T Pham
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anne N Kronshage
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Steven Lisgo
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James Bristow
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Thomas P Cappola
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA; Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Diane E Dickel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA; School of Natural Sciences, University of California, Merced, Merced, CA 95343, USA.
| |
Collapse
|
34
|
Luo K, Zheng C, Luo R, Cao X, Sun H, Ma H, Huang J, Yang X, Wu X, Li X. Identification and functional characterization of BICD2 as a candidate disease gene in an consanguineous family with dilated cardiomyopathy. BMC Med Genomics 2022; 15:189. [PMID: 36068540 PMCID: PMC9446846 DOI: 10.1186/s12920-022-01349-y] [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: 05/25/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022] Open
Abstract
Background Familial dilated cardiomyopathy (DCM) is a genetic cardiomyopathy that is associated with reduced left ventricle function or systolic function. Fifty-one DCM-causative genes have been reported, most of which are inherited in an autosomal dominant manner. However, recessive DCM-causative gene is rarely observed. Methods Whole-exome sequencing (WES) was performed in a consanguineous family with DCM to identify candidate variants. Sanger sequencing was utilized to confirm the variant. We then checked the DCM candidate gene in 210 sporadic DCM cases. We next explored BICD2 function in both embryonic and adult bicd2-knockout zebrafish models. In vivo cardiac function of bicd2-knockout fish was detected by echocardiography and RNA-seq. Results We identified an autosomal recessive and evolutionarily conserved missense variant, NM_001003800.1:c.2429G > A, in BICD2, which segregated with the disease phenotype in a consanguineous family with DCM. Furthermore, we confirmed the presence of BICD2 variants in 3 sporadic cases. Knockout of bicd2 resulted in partial embryonic lethality in homozygotes, suggesting a vital role for bicd2 in embryogenesis. Heart dilation and decreased ejection fraction, cardiac output and stroke volume were observed in bicd2-knockout zebrafish, suggesting a phenotype similar to human DCM. Furthermore, RNA-seq confirmed a larger transcriptome shift in in bicd2 homozygotes than in heterozygotes. Gene set enrichment analysis of bicd2-deficient fish showed the enrichment of altered gene expression in cardiac pathways and mitochondrial energy metabolism. Conclusions Our study first shows that BICD2 is a novel candidate gene associated with familial DCM, and our findings will facilitate further insights into the molecular pathological mechanisms of DCM. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01349-y.
Collapse
Affiliation(s)
- Kai Luo
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, People's Republic of China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China
| | - Chenqing Zheng
- Shenzhen Aone Medical Laboratory Co., Ltd., Shenzhen, People's Republic of China
| | - Rong Luo
- Institute of Geriatric Cardiovascular Disease, Chengdu Medical College, Chengdu, People's Republic of China
| | - Xin Cao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Huajun Sun
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China.,Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, People's Republic of China
| | - Huihui Ma
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, People's Republic of China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China
| | - Jichang Huang
- Institute of Geriatric Cardiovascular Disease, Chengdu Medical College, Chengdu, People's Republic of China
| | - Xu Yang
- Shenzhen Aone Medical Laboratory Co., Ltd., Shenzhen, People's Republic of China
| | - Xiushan Wu
- The Center for Heart Development, Hunan Normal University, Changsha, People's Republic of China. .,Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou, Guangdong, People's Republic of China.
| | - Xiaoping Li
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, People's Republic of China. .,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China.
| |
Collapse
|
35
|
Yousaf M, Khan WA, Shahzad K, Khan HN, Ali B, Hussain M, Awan FR, Mustafa H, Sheikh FN. Genetic Association of Beta-Myosin Heavy-Chain Gene (MYH7) with Cardiac Dysfunction. Genes (Basel) 2022; 13:genes13091554. [PMID: 36140722 PMCID: PMC9498774 DOI: 10.3390/genes13091554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiac dysfunction accelerates the risk of heart failure, and its pathogenesis involves a complex interaction between genetic and environmental factors. Variations in myosin affect contractile abilities of cardiomyocytes and cause structural and functional abnormalities in myocardium. The study aims to find the association of MYH7 rs121913642 (c.1594 T>C) and rs121913645 (c.667G>A) variants with cardiac dysfunction in the Punjabi Pakistani population. Patients with heart failure (n = 232) and healthy controls (n = 205) were enrolled in this study. MYH7 variant genotyping was performed using tetra ARMS-PCR. MYH7 rs121913642 TC genotype was significantly more prevalent in the patient group (p < 0.001). However, MYH7 rs121913645 genotype frequencies were not significantly different between the patient and control groups (p < 0.666). Regression analysis also revealed that the rs121913642 C allele increases the risk of cardiac failure by ~2 [OR:1.98, CI: 1.31−2.98, p < 0.001] in comparison to the T allele. High levels of the cardiac enzymes cardiac troponin I (cTnI) and CK-MB were observed in patients. There was also an increase in total cholesterol, LDL cholesterol, and uric acid in patients compared to the healthy control group (p < 0.001). In conclusion, the MYH7 gene variant rs121913642 is genetically associated with cardiac dysfunction and involved in the pathogenesis of HF.
Collapse
Affiliation(s)
- Memoona Yousaf
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
| | - Waqas Ahmed Khan
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
- Correspondence: (W.A.K.); (H.M.); Tel.: +92-321-9331563 (W.A.K.)
| | - Khurrum Shahzad
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
- Institute of Clinical Chemistry, University Hospital Leipzig Institute of Clinical Chemistry Liebigstraße27, D-04103 Leipzig, Germany
| | - Haq Nawaz Khan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan
| | - Basharat Ali
- Department of Family Medicine, University of Health Sciences, Lahore 42000, Pakistan
| | - Misbah Hussain
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan
| | - Fazli Rabbi Awan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan
| | - Hamid Mustafa
- Department of Animal Breeding & Genetics, University of Veterinary and Animal Sciences, Lahore 42000, Pakistan
- Correspondence: (W.A.K.); (H.M.); Tel.: +92-321-9331563 (W.A.K.)
| | | |
Collapse
|
36
|
Reaney M, Addepalli P, Allen V, Spertus JA, Dolan C, Sehnert AJ, Fine JT. Longitudinal Psychometric Analysis of the Hypertrophic Cardiomyopathy Symptom Questionnaire (HCMSQ) Using Outcomes from the Phase III EXPLORER-HCM Trial. PHARMACOECONOMICS - OPEN 2022; 6:575-586. [PMID: 35718845 PMCID: PMC9283589 DOI: 10.1007/s41669-022-00340-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) symptoms include shortness of breath (SOB), fatigue, chest pain, palpitations, dizziness, and fainting. The HCM Symptom Questionnaire (HCMSQ), the only patient-reported outcome instrument designed to specifically measure HCM symptoms, yields four domain scores (SOB, tiredness, cardiovascular symptoms, syncope) and a total score. We evaluated the longitudinal psychometric properties of the HCMSQ using baseline to week 30 data from the phase III EXPLORER-HCM trial (NCT03470545). METHODS Test-retest reliability was assessed via intraclass correlation of patients with stable Patient Global Impression of Change (PGIC) and Patient Global Impression of Severity (PGIS) responses. Sensitivity to change was assessed via Spearman correlations with the Kansas City Cardiomyopathy Questionnaire (KCCQ-23) and the EuroQoL visual analogue scale (EQ VAS), and via one-way ANOVA comparing change groups defined on clinical (New York Heart Association [NYHA] class, left ventricular outflow tract [LVOT] gradient, peak oxygen consumption [pVO2]) and patient-reported (PGIS, PGIC) variables. Meaningful change thresholds were established via PGIC/PGIS. RESULTS All HCMSQ scores showed strong evidence of test-retest reliability (intraclass correlation coefficient > 0.70). Sensitivity to change was demonstrated with mostly strong/moderate correlations with KCCQ-23 and EQ VAS, and significant differences (p ≤ 0.05) in PGIS, PGIC, pVO2, and NYHA (except tiredness domain) change categories, but not LVOT gradient. Clinically meaningful score reductions were ≥1 point for tiredness and cardiovascular symptoms domains, ≥ 2.5 points for SOB domain, and ≥2 points for total score. CONCLUSIONS Results suggest that HCMSQ is fit for purpose in capturing HCM symptoms and may provide evidence of treatment benefit from the patients' perspectives.
Collapse
Affiliation(s)
- Matthew Reaney
- IQVIA, 3 Forbury Place, 23 Forbury Road, Reading, RG1 3JH, UK.
| | | | | | - John A Spertus
- Saint Luke's Mid America Heart Institute and the University of Missouri-Kansas City, Kansas City, MO, USA
| | | | - Amy J Sehnert
- MyoKardia, Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA, USA
| | - Jennifer T Fine
- MyoKardia, Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA, USA
| |
Collapse
|
37
|
Genetic Basis of Dilated Cardiomyopathy in Dogs and Its Potential as a Bidirectional Model. Animals (Basel) 2022; 12:ani12131679. [PMID: 35804579 PMCID: PMC9265105 DOI: 10.3390/ani12131679] [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: 04/07/2022] [Revised: 06/16/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Heart disease is a leading cause of death for both humans and dogs. Inherited heart diseases, including dilated cardiomyopathy (DCM), account for a proportion of these cases. Human and canine patients with DCM suffer from an enlarged heart that can no longer pump efficiently, resulting in heart failure. This causes symptoms or clinical signs like difficulty breathing, irregular heartbeat, and eventually death. The symptoms or clinical signs of this disease vary in age of onset at the beginning of symptoms, sex predisposition, and overall disease progression. Despite the many similarities in DCM in both species, only a few candidate genes so far have been linked to this disease in dogs versus tens of genes identified in human DCM. Additionally, the use of induced pluripotent stem cells, or engineered stem cells, has been widely used in the study of human genetic heart disease but has not yet been fully adapted to study heart disease in dogs. This review describes the current knowledge on the genetics and subtypes of naturally occurring DCM in dogs, and how advances in research might benefit the dog but also the human patient. Additionally, a novel method using canine engineered stem cells to uncover unknown contributions of mistakes in DNA to the progression of DCM will be introduced along with its applications for human DCM disease modeling and treatment. Abstract Cardiac disease is a leading cause of death for both humans and dogs. Genetic cardiomyopathies, including dilated cardiomyopathy (DCM), account for a proportion of these cases in both species. Patients may suffer from ventricular enlargement and systolic dysfunction resulting in congestive heart failure and ventricular arrhythmias with high risk for sudden cardiac death. Although canine DCM has similar disease progression and subtypes as in humans, only a few candidate genes have been found to be associated with DCM while the genetic background of human DCM has been more thoroughly studied. Additionally, experimental disease models using induced pluripotent stem cells have been widely adopted in the study of human genetic cardiomyopathy but have not yet been fully adapted for the in-depth study of canine genetic cardiomyopathies. The clinical presentation of DCM is extremely heterogeneous for both species with differences occurring based on sex predisposition, age of onset, and the rate of disease progression. Both genetic predisposition and environmental factors play a role in disease development which are identical in dogs and humans in contrast to other experimental animals. Interestingly, different dog breeds have been shown to develop distinct DCM phenotypes, and this presents a unique opportunity for modeling as there are multiple breed-specific models for DCM with less genetic variance than human DCM. A better understanding of DCM in dogs has the potential for improved selection for breeding and could lead to better overall care and treatment for human and canine DCM patients. At the same time, progress in research made for human DCM can have a positive impact on the care given to dogs affected by DCM. Therefore, this review will analyze the feasibility of canines as a naturally occurring bidirectional disease model for DCM in both species. The histopathology of the myocardium in canine DCM will be evaluated in three different breeds compared to control tissue, and the known genetics that contributes to both canine and human DCM will be summarized. Lastly, the prospect of canine iPSCs as a novel method to uncover the contributions of genetic variants to the pathogenesis of canine DCM will be introduced along with the applications for disease modeling and treatment.
Collapse
|
38
|
Sarohi V, Srivastava S, Basak T. A Comprehensive Outlook on Dilated Cardiomyopathy (DCM): State-Of-The-Art Developments with Special Emphasis on OMICS-Based Approaches. J Cardiovasc Dev Dis 2022; 9:jcdd9060174. [PMID: 35735803 PMCID: PMC9225617 DOI: 10.3390/jcdd9060174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Dilated cardiomyopathy (DCM) remains an enigmatic cardiovascular disease (CVD) condition characterized by contractile dysfunction of the myocardium due to dilation of the ventricles. DCM is one of the major forms of CVD contributing to heart failure. Dilation of the left or both ventricles with systolic dysfunction, not explained by known causes, is a hallmark of DCM. Progression of DCM leads to heart failure. Genetic and various other factors greatly contribute to the development of DCM, but the etiology has still remained elusive in a large number of cases. A significant number of studies have been carried out to identify the genetic causes of DCM. These candidate-gene studies revealed that mutations in the genes of the fibrous, cytoskeletal, and sarcomeric proteins of cardiomyocytes result in the development of DCM. However, a significant proportion of DCM patients are idiopathic in nature. In this review, we holistically described the symptoms, causes (in adults and newborns), genetic basis, and mechanistic progression of DCM. Further, we also summarized the state-of-the-art diagnosis, available biomarkers, treatments, and ongoing clinical trials of potential drug regimens. DCM-mediated heart failure is on the rise worldwide including in India. The discovery of biomarkers with a better prognostic value is the need of the hour for better management of DCM-mediated heart failure patients. With the advent of next-generation omics-based technologies, it is now possible to probe systems-level alterations in DCM patients pertaining to the identification of novel proteomic and lipidomic biomarkers. Here, we also highlight the onset of a systems-level study in Indian DCM patients by applying state-of-the-art mass-spectrometry-based “clinical proteomics” and “clinical lipidomics”.
Collapse
Affiliation(s)
- Vivek Sarohi
- Indian Institute of Technology (IIT)-Mandi, School of Basic Sciences (SBS), Mandi 175075, HP, India; (V.S.); (S.S.)
- BioX Centre, Indian Institute of Technology (IIT)-Mandi, Mandi 175075, HP, India
| | - Shriya Srivastava
- Indian Institute of Technology (IIT)-Mandi, School of Basic Sciences (SBS), Mandi 175075, HP, India; (V.S.); (S.S.)
| | - Trayambak Basak
- Indian Institute of Technology (IIT)-Mandi, School of Basic Sciences (SBS), Mandi 175075, HP, India; (V.S.); (S.S.)
- BioX Centre, Indian Institute of Technology (IIT)-Mandi, Mandi 175075, HP, India
- Correspondence: ; Tel.: +91-1905-267826
| |
Collapse
|
39
|
Jia H, Sun Y, Yao W, Chen Z, Yang S, Wang C, Lu S. A novel deletion mutation accompanied by a point mutation in Lamin A/C gene: Screened from a dilated cardiomyopathy family. Perfusion 2022; 38:826-836. [PMID: 35514053 DOI: 10.1177/02676591221090587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND There are 30%-40% of patients with dilated cardiomyopathy (DCM) having genetic causes, among which Lamin A and C gene (LMNA) mutation is the second most frequent DCM-related mutation, and Lamin A/C may be involved in the pathogenesis of DCM through the regulation of gene transcription or the direct effect of cell structure. Methods: Echocardiography and electrocardiogram were used to diagnose DCM and arrhythmia in a DCM family. Then, linked mutations on LMNA were screened out by high-throughput sequencing and verified by Sanger sequencing in all research individuals. Meanwhile, Human Genome Variation Society (HGVS) and Integrative Genomics Viewer (IGV) were used to analyse the characteristics of the mutated Lamin A/C protein. Finally, mutated-type and wild-type LMNA plasmid was transfected into AC-16 cardiomyocytes with the form of a lentivirus vector, and its effect on nucleus and actin was studied by immunofluorescence detection. RESULTS In this study, we found a new frame-shifted mutation of LMNA (p.Ser414Alafs*66) linked with another point mutation from a DCM family by using High-throughput sequencing, and this deletion mutation led to a truncation of Lamin A/C. By analysing the clinical characteristics of this DCM family, we found that all DCM patients with arrhythmia were carriers of this co-segregation mutation. In the cytological experiment, we found that the mutated-type transfections showed weaker fluorescent intensities on both actin and cell nucleus. CONCLUSIONS A co-segregation mutation of LMNA (Point mutation chr1 156107548 c.1712 G>A and truncated frame-shifted mutation chr1 156106086 c.1240delA) was found from a DCM family, and this type of mutation could participate in the pathogenesis of DCM by affecting the expression of actin.
Collapse
Affiliation(s)
- Hao Jia
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongxin Sun
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wangchao Yao
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenhang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shouguo Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyang Lu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Trachtenberg BH, Jimenez J, Morris AA, Kransdorf E, Owens A, Fishbein DP, Jordan E, Kinnamon DD, Mead JO, Huggins GS, Hershberger RE. TTR variants in patients with dilated cardiomyopathy: An investigation of the DCM Precision Medicine Study. Genet Med 2022; 24:1495-1502. [PMID: 35438637 DOI: 10.1016/j.gim.2022.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022] Open
Abstract
PURPOSE The cardiac phenotype of hereditary transthyretin amyloidosis (hTTR) usually presents as a restrictive or hypertrophic cardiomyopathy, and, although rarely observed as dilated cardiomyopathy (DCM), TTR is routinely included in DCM genetic testing panels. However, the prevalence and phenotypes of TTR variants in patients with DCM have not been reported. METHODS Exome sequences of 729 probands with idiopathic DCM were analyzed for TTR and 35 DCM genes. RESULTS Rare TTR variants were identified in 2 (0.5%; 95% CI = 0.1%-1.8%) of 404 non-Hispanic White DCM probands; neither of them had features of hTTR. In 1 proband, a TTR His110Asn variant and a variant of uncertain significance in DSP were identified, and in the other proband, a TTR Val50Met variant known to cause hTTR and a likely pathogenic variant in FLNC were identified. The TTR Val142Ile variant was identified in 8 (3.0%) non-Hispanic Black probands, comparable with African/African American Genome Aggregation Database controls (OR = 1.01; 95% CI = 0.46-1.99). CONCLUSION Among the 729 DCM probands, 2 had rare TTR variants identified without the features of hTTR, and both had other plausible genetic causes of DCM. Moreover, the frequency of TTR Val142Ile was comparable to a control sample. These findings suggest that hTTR variants may have a limited role in patients with DCM without TTR-specific findings.
Collapse
Affiliation(s)
- Barry H Trachtenberg
- Houston Methodist DeBakey Heart & Vascular Center, J.C. Walter Jr Transplant Center, Houston Methodist, Houston, TX.
| | - Javier Jimenez
- Miami Cardiac & Vascular Institute, Baptist Health South Florida, Miami, FL
| | - Alanna A Morris
- Emory University School of Medicine, Emory University, Atlanta, GA
| | - Evan Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Anjali Owens
- Center for Inherited Cardiovascular Disease, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Elizabeth Jordan
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH
| | - Daniel D Kinnamon
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH
| | - Jonathan O Mead
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH
| | | | - Ray E Hershberger
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH; Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH.
| | | |
Collapse
|
41
|
Haas GJ, Zareba KM, Ni H, Bello-Pardo E, Huggins GS, Hershberger RE. Validating an Idiopathic Dilated Cardiomyopathy Diagnosis Using Cardiovascular Magnetic Resonance: The Dilated Cardiomyopathy Precision Medicine Study. Circ Heart Fail 2022; 15:e008877. [PMID: 35240856 PMCID: PMC9117485 DOI: 10.1161/circheartfailure.121.008877] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Coronary angiography to identify coronary artery disease has been foundational to distinguish the cause of dilated cardiomyopathy (DCM), including the assignment of idiopathic or ischemic cardiomyopathy. Late gadolinium enhancement (LGE) with cardiovascular magnetic resonance (CMR) has emerged as an approach to identify myocardial scar and identify etiology. METHODS The DCM Precision Medicine Study included patients with left ventricular dilation and dysfunction attributed to idiopathic DCM, after expert clinical review excluded ischemic or other cardiomyopathies. Ischemic cardiomyopathy was defined as coronary artery disease with >50% narrowing at angiography of ≥1 epicardial coronary artery. CMR was not required for study inclusion, but in a post hoc analysis of available CMR reports, patterns of LGE were classified as (1) no LGE, (2) ischemic-pattern LGE: subendocardial/transmural, (3) nonischemic LGE: midmyocardial/epicardial. RESULTS Of 1204 idiopathic DCM patients evaluated, 396 (32.9%) had a prior CMR study; of these, 327 (82.6% of 396) had LGE imaging (mean age 46 years; 53.2% male; 55.4% White); 178 of the 327 (54.4%) exhibited LGE, and 156 of the 178 had LGE consistent with idiopathic DCM. The remaining 22 had transmural or subendocardial LGE. Of these 22, coronary angiography was normal (13), showed luminal irregularities (3), a distant thrombus (1), coronary artery disease with <50% coronary artery narrowing (1), or was not available (4). CONCLUSIONS Of 327 probands enrolled in the DCM Precision Medicine Study cohort who had LGE-CMR data available, an ischemic-pattern of LGE was identified in 22 (6.7%), all of whom had idiopathic DCM as adjudicated by expert clinical review. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03037632.
Collapse
Affiliation(s)
- Garrie J Haas
- Advanced Heart Failure and Cardiac Transplant Program, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., R.E.H.).,Division of Cardiovascular Medicine, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., R.E.H.).,Dorothy M. Davis Heart and Lung Research Institute, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., H.N., E.B.-P., R.E.H.)
| | - Karolina M Zareba
- Division of Cardiovascular Medicine, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., R.E.H.).,Dorothy M. Davis Heart and Lung Research Institute, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., H.N., E.B.-P., R.E.H.)
| | - Hanyu Ni
- Dorothy M. Davis Heart and Lung Research Institute, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., H.N., E.B.-P., R.E.H.).,Division of Human Genetics, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (H.N., E.B.-P., R.E.H.)
| | - Erika Bello-Pardo
- Dorothy M. Davis Heart and Lung Research Institute, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., H.N., E.B.-P., R.E.H.).,Division of Human Genetics, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (H.N., E.B.-P., R.E.H.)
| | - Gordon S Huggins
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (G.S.H.)
| | - Ray E Hershberger
- Advanced Heart Failure and Cardiac Transplant Program, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., R.E.H.).,Division of Cardiovascular Medicine, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., R.E.H.).,Dorothy M. Davis Heart and Lung Research Institute, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (G.J.H., K.M.Z., H.N., E.B.-P., R.E.H.).,Division of Human Genetics, all in the Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (H.N., E.B.-P., R.E.H.)
| | | |
Collapse
|
42
|
Man Y, Yi C, Fan M, Yang T, Liu P, Liu S, Wang G. Identification of a novel missense mutation in the TPM1 gene via exome sequencing in a Chinese family with dilated cardiomyopathy: A case report and literature review. Medicine (Baltimore) 2022; 101:e28551. [PMID: 35029218 PMCID: PMC8758022 DOI: 10.1097/md.0000000000028551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/22/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Dilated cardiomyopathy (DCM) is a cardiovascular disorder characterized by consecutive ventricular dilation and contractile dysfunction, often leading to congestive heart failure. DCM type 1Y (DCM1Y) is caused by a mutation in the TPM1 (tropomyosin 1) gene. To date, about thirty TPM1 gene mutations have been reported to be related to DCM1Y. However, mutational screening of the TPM1 gene is still far from being complete. Identification of TPM1 mutation is particularly important in the diagnosis of DCM1Y and will give more insights into the molecular pathogenesis of DCM1Y. PATIENT CONCERNS A Chinese Han family with DCM phenotypes was examined. DIAGNOSIS A novel missense mutation, c.340G > C in exon 3 of the TPM1 gene, was identified. INTERVENTIONS Next-generation sequencing (NGS) of DNA samples was performed to detect the gene mutation in the proband, which was confirmed by Sanger sequencing. OUTCOMES This novel heterozygous mutation results in the substitution of glutamic acid with glutamine (p.E114Q). Based on this finding and clinical manifestations, a final diagnosis of DCM1Y was made. LESSONS We present evidence that p.E114Q mutation represents a novel TPM1 mutation in a Chinese Han family with DCM. Our data expand the mutation spectrum of the TPM1 gene and may facilitate the clinical diagnosis of DCM1Y.
Collapse
Affiliation(s)
- Yilong Man
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Innoviation Center of Intelligent Diagnosis, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Changying Yi
- Department of Clinical Laboratory, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Meili Fan
- Department of Acupuncture and Massage, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Tianyu Yang
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peng Liu
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shiguang Liu
- Department of Cardiology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guangxin Wang
- Shandong Innoviation Center of Intelligent Diagnosis, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Cardiology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
43
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
44
|
Asher C, Puyol-Antón E, Rizvi M, Ruijsink B, Chiribiri A, Razavi R, Carr-White G. The Role of AI in Characterizing the DCM Phenotype. Front Cardiovasc Med 2021; 8:787614. [PMID: 34993240 PMCID: PMC8724536 DOI: 10.3389/fcvm.2021.787614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Dilated Cardiomyopathy is conventionally defined by left ventricular dilatation and dysfunction in the absence of coronary disease. Emerging evidence suggests many patients remain vulnerable to major adverse outcomes despite clear therapeutic success of modern evidence-based heart failure therapy. In this era of personalized medical care, the conventional assessment of left ventricular ejection fraction falls short in fully predicting evolution and risk of outcomes in this heterogenous group of heart muscle disease, as such, a more refined means of phenotyping this disease appears essential. Cardiac MRI (CMR) is well-placed in this respect, not only for its diagnostic utility, but the wealth of information captured in global and regional function assessment with the addition of unique tissue characterization across different disease states and patient cohorts. Advanced tools are needed to leverage these sensitive metrics and integrate with clinical, genetic and biochemical information for personalized, and more clinically useful characterization of the dilated cardiomyopathy phenotype. Recent advances in artificial intelligence offers the unique opportunity to impact clinical decision making through enhanced precision image-analysis tasks, multi-source extraction of relevant features and seamless integration to enhance understanding, improve diagnosis, and subsequently clinical outcomes. Focusing particularly on deep learning, a subfield of artificial intelligence, that has garnered significant interest in the imaging community, this paper reviews the main developments that could offer more robust disease characterization and risk stratification in the Dilated Cardiomyopathy phenotype. Given its promising utility in the non-invasive assessment of cardiac diseases, we firstly highlight the key applications in CMR, set to enable comprehensive quantitative measures of function beyond the standard of care assessment. Concurrently, we revisit the added value of tissue characterization techniques for risk stratification, showcasing the deep learning platforms that overcome limitations in current clinical workflows and discuss how they could be utilized to better differentiate at-risk subgroups of this phenotype. The final section of this paper is dedicated to the allied clinical applications to imaging, that incorporate artificial intelligence and have harnessed the comprehensive abundance of data from genetics and relevant clinical variables to facilitate better classification and enable enhanced risk prediction for relevant outcomes.
Collapse
Affiliation(s)
- Clint Asher
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
| | - Esther Puyol-Antón
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Maleeha Rizvi
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
| | - Bram Ruijsink
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Amedeo Chiribiri
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
| | - Reza Razavi
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
| | - Gerry Carr-White
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Cardiology, Guys and St Thomas' NHS Trust, London, United Kingdom
| |
Collapse
|
45
|
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: 8] [Impact Index Per Article: 2.0] [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
|
46
|
Farooqi N, Metherell LA, Schrauwen I, Acharya A, Khan Q, Nouel Saied LM, Ali Y, El-Serehy HA, Jalil F, Leal SM. Exome Sequencing Identifies a Novel FBN1 Variant in a Pakistani Family with Marfan Syndrome That Includes Left Ventricle Diastolic Dysfunction. Genes (Basel) 2021; 12:1915. [PMID: 34946863 PMCID: PMC8700962 DOI: 10.3390/genes12121915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/12/2021] [Accepted: 09/18/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Cardiomyopathies are diseases of the heart muscle and are important causes of heart failure. Dilated cardiomyopathy (DCM) is a common form of cardiomyopathy that can be acquired, syndromic or non-syndromic. The current study was conducted to explore the genetic defects in a Pakistani family with cardiac disease and features of Marfan's syndrome (MFS). METHODS A family with left ventricle (LV) diastolic dysfunction and MFS phenotype was assessed in Pakistan. The clinical information and blood samples from the patients were collected after physical, cardiovascular, and ophthalmologic examinations. An affected individual (proband) was subjected to whole-exome sequencing (WES). The findings were further validated through Sanger sequencing in the family. RESULTS Through WES and sanger validation, we identified a novel variant NM_000138.4; c.1402A>G in the Fibrillin-1 (FBN1) gene that segregates with LV diastolic dysfunction and MFS. Furthermore, bioinformatic evaluation suggested that the novel variant is deleterious and disease-causing. CONCLUSIONS This study identified for the first time a novel FBN1 variant in a family with LV diastolic dysfunction and MFS in Pakistan.
Collapse
Affiliation(s)
- Nadia Farooqi
- Department of Biotechnology, Faculty of Chemical and Life Sciences, Abdul Wali Khan University, Mardan 23200, Pakistan; (N.F.); (Q.K.); (Y.A.)
| | - Louise A. Metherell
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square Campus, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; (I.S.); (A.A.); (L.M.N.S.)
| | - Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; (I.S.); (A.A.); (L.M.N.S.)
| | - Qayum Khan
- Department of Biotechnology, Faculty of Chemical and Life Sciences, Abdul Wali Khan University, Mardan 23200, Pakistan; (N.F.); (Q.K.); (Y.A.)
| | - Liz M. Nouel Saied
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; (I.S.); (A.A.); (L.M.N.S.)
| | - Yasir Ali
- Department of Biotechnology, Faculty of Chemical and Life Sciences, Abdul Wali Khan University, Mardan 23200, Pakistan; (N.F.); (Q.K.); (Y.A.)
| | - Hamed A. El-Serehy
- Department of Zoology, College of Science, King Saud University, Riyadh I1451, Saudi Arabia;
| | - Fazal Jalil
- Department of Biotechnology, Faculty of Chemical and Life Sciences, Abdul Wali Khan University, Mardan 23200, Pakistan; (N.F.); (Q.K.); (Y.A.)
| | - Suzanne M. Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; (I.S.); (A.A.); (L.M.N.S.)
- Taub Institute for Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
47
|
Diagnostic biomarkers of dilated cardiomyopathy. Immunobiology 2021; 226:152153. [PMID: 34784575 DOI: 10.1016/j.imbio.2021.152153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a condition involving dilation of cardiac chambers, which results in contraction impairment. Besides invasive and non-invasive diagnostic procedures, cardiac biomarkers are of great importance in both diagnosis and prognosis of the disease. These biomarkers are categorized into three groups based on their site; cardiomyocyte biomarkers, microenvironmental biomarkers and macroenvironmental biomarkers. AIMS In this review, an overview of characteristics, epidemiology, etiology and clinical manifestations of DCM is provided. In addition, the most important biomarkers, of all three categories, and their diagnostic and prognostic values are discussed. CONCLUSION Considering the association of DCM with conditions such as infections and autoimmunity, which are prevalent among the population, introducing efficient diagnostic tools is of high value for the early detection of DCM to prevent its severe complications. The three discussed classes of biomarkers are potential candidates for the detection of DCM. However, further studies are necessary in this regard.
Collapse
|
48
|
Whole-Exome Sequencing Identified a Novel Variant (C.405_422+39del) in DSP Gene in an Iranian Pedigree with Familial Dilated Cardiomyopathy. Rep Biochem Mol Biol 2021; 10:280-287. [PMID: 34604417 DOI: 10.52547/rbmb.10.2.280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/13/2021] [Indexed: 11/18/2022]
Abstract
Background Dilated cardiomyopathy (DCM) is a progressive heart condition characterized by left ventricular chamber enlargement associated with systolic heart failure and prolonged action potential duration. Genetic variations in genes that encode cytoskeleton, sarcomere, and nuclear envelope proteins are responsible for 45% of cases. In our study, we focused on a pedigree with familial DCM to decipher the potential genetic cause(s) in affected members developing arrhythmia, end-stage heart failure, and sudden death. Methods Whole-exome sequencing (WES) was exploited for a 27-year-old heart-transplanted female as the proband, and the derived data were filtered using the standard pipelines. Results A 57-nucleotide deletion (c.405_422+39del) in the desmoplakin gene (DSP) (NM_004415.4) was identified as a novel pathogenic variant. Familial segregation analysis indicated that this variant is present in clinically affected members and absent in unaffected members. Conclusion It seems that the detected variant induces intron retention, resulting in a premature stop codon in intron 3 of DSP leading to production of a truncated, nonfunctional protein. Additionally, it can trigger a nonsense-mediated mRNA decay pathway associated with inhibition of protein production. The present study results illustrated that a novel deletion in DSP can cause DCM in an Iranian family.
Collapse
|
49
|
El Mathari B, Briand P, Corbier A, Poirier B, Briand V, Raffenne-Devillers A, Harnist MP, Guillot E, Guilbert F, Janiak P. Apelin improves cardiac function mainly through peripheral vasodilation in a mouse model of dilated cardiomyopathy. Peptides 2021; 142:170568. [PMID: 33965442 DOI: 10.1016/j.peptides.2021.170568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/13/2021] [Accepted: 04/19/2021] [Indexed: 12/23/2022]
Abstract
There is growing evidence that apelin plays a role in the regulation of the cardiovascular system by increasing myocardial contractility and acting as a vasodilator. However, it remains unclear whether apelin improves cardiac contractility in a load-dependent or independent manner in pathological conditions. For this purpose we investigated the cardiovascular effects of apelin in α-actin transgenic mice (mActin-Tg mice), a model of cardiomyopathy. [Pyr1]apelin-13 was administered by continuous infusion at 2 mg/kg/d for 3 weeks. Effects on cardiac function were determined by echocardiography and a Pressure-Volume (PV) analysis. mActin-Tg mice showed a dilated cardiomyopathy (DCM) phenotype similar to that encountered in patients expressing the same mutation. Compared to WT animals, mActin-Tg mice displayed cardiac systolic impairment [significant decrease in ejection fraction (EF), cardiac output (CO), and stroke volume (SV)] associated with cardiac ventricular dilation and diastolic dysfunction, characterized by an impairment in mitral flow velocity (E/A) and in deceleration time (DT). Load-independent myocardial contractility was strongly decreased in mActin-Tg mice while total peripheral vascular resistance (TPR) was significantly increased. As compared to vehicle-treated animals, a 3-week treatment with [Pyr1]apelin-13 significantly improved EF%, SV, E/A, DT and corrected TPR, with no significant effect on load-independent indices of myocardial contractility, blood pressure and heart rate. In conclusion [Pyr1]apelin-13 displayed no intrinsic contractile effect but improved cardiac function in dilated cardiomyopathy mainly by reducing peripheral vascular resistance, with no change in blood pressure.
Collapse
Affiliation(s)
- Brahim El Mathari
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Pascale Briand
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Alain Corbier
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Bruno Poirier
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Véronique Briand
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Alice Raffenne-Devillers
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Marie-Pierre Harnist
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Etienne Guillot
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Frederique Guilbert
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Philip Janiak
- Cardiovascular & Metabolism Therapeutic Area, Sanofi R&D, 1 avenue Pierre Brossolette, 91385, Chilly-Mazarin, France.
| |
Collapse
|
50
|
Jordan E, Peterson L, Ai T, Asatryan B, Bronicki L, Brown E, Celeghin R, Edwards M, Fan J, Ingles J, James CA, Jarinova O, Johnson R, Judge DP, Lahrouchi N, Lekanne Deprez RH, Lumbers RT, Mazzarotto F, Medeiros Domingo A, Miller RL, Morales A, Murray B, Peters S, Pilichou K, Protonotarios A, Semsarian C, Shah P, Syrris P, Thaxton C, van Tintelen JP, Walsh R, Wang J, Ware J, Hershberger RE. Evidence-Based Assessment of Genes in Dilated Cardiomyopathy. Circulation 2021; 144:7-19. [PMID: 33947203 PMCID: PMC8247549 DOI: 10.1161/circulationaha.120.053033] [Citation(s) in RCA: 310] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/13/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Each of the cardiomyopathies, classically categorized as hypertrophic cardiomyopathy, dilated cardiomyopathy (DCM), and arrhythmogenic right ventricular cardiomyopathy, has a signature genetic theme. Hypertrophic cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy are largely understood as genetic diseases of sarcomere or desmosome proteins, respectively. In contrast, >250 genes spanning >10 gene ontologies have been implicated in DCM, representing a complex and diverse genetic architecture. To clarify this, a systematic curation of evidence to establish the relationship of genes with DCM was conducted. METHODS An international panel with clinical and scientific expertise in DCM genetics evaluated evidence supporting monogenic relationships of genes with idiopathic DCM. The panel used the Clinical Genome Resource semiquantitative gene-disease clinical validity classification framework with modifications for DCM genetics to classify genes into categories on the basis of the strength of currently available evidence. Representation of DCM genes on clinically available genetic testing panels was evaluated. RESULTS Fifty-one genes with human genetic evidence were curated. Twelve genes (23%) from 8 gene ontologies were classified as having definitive (BAG3, DES, FLNC, LMNA, MYH7, PLN, RBM20, SCN5A, TNNC1, TNNT2, TTN) or strong (DSP) evidence. Seven genes (14%; ACTC1, ACTN2, JPH2, NEXN, TNNI3, TPM1, VCL) including 2 additional ontologies were classified as moderate evidence; these genes are likely to emerge as strong or definitive with additional evidence. Of these 19 genes, 6 were similarly classified for hypertrophic cardiomyopathy and 3 for arrhythmogenic right ventricular cardiomyopathy. Of the remaining 32 genes (63%), 25 (49%) had limited evidence, 4 (8%) were disputed, 2 (4%) had no disease relationship, and 1 (2%) was supported by animal model data only. Of the 16 evaluated clinical genetic testing panels, most definitive genes were included, but panels also included numerous genes with minimal human evidence. CONCLUSIONS In the curation of 51 genes, 19 had high evidence (12 definitive/strong, 7 moderate). It is notable that these 19 genes explain only a minority of cases, leaving the remainder of DCM genetic architecture incompletely addressed. Clinical genetic testing panels include most high-evidence genes; however, genes lacking robust evidence are also commonly included. We recommend that high-evidence DCM genes be used for clinical practice and that caution be exercised in the interpretation of variants in variable-evidence DCM genes.
Collapse
Affiliation(s)
- Elizabeth Jordan
- Division of Human Genetics (E.J., L.P., T.A., R.E.H.), Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus
| | - Laiken Peterson
- Division of Human Genetics (E.J., L.P., T.A., R.E.H.), Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus
| | - Tomohiko Ai
- Division of Human Genetics (E.J., L.P., T.A., R.E.H.), Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus
| | - Babken Asatryan
- Department for Cardiology, Inselspital, Bern University Hospital, University of Bern, Switzerland (B.A.)
| | - Lucas Bronicki
- Department of Genetics, Children’s Hospital of Eastern Ontario, Ottawa, Canada (L.B., O.J.)
- Department of Laboratory and Pathology Medicine, University of Ottawa, Ontario, Canada (L.B., O.J.)
| | - Emily Brown
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (E.B., C.A.J., B.M.)
| | - Rudy Celeghin
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padua, Italy (R.C., K.P.)
| | - Matthew Edwards
- Clinical Genetics and Genomics Laboratory, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom (M.E.)
| | - Judy Fan
- Department of Medicine, University of California, Los Angeles (J.F., J. Wang)
| | - Jodie Ingles
- Cardio Genomics Program at Centenary Institute, University of Sydney, Australia (J.I.)
| | - Cynthia A. James
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (E.B., C.A.J., B.M.)
| | - Olga Jarinova
- Department of Genetics, Children’s Hospital of Eastern Ontario, Ottawa, Canada (L.B., O.J.)
- Department of Laboratory and Pathology Medicine, University of Ottawa, Ontario, Canada (L.B., O.J.)
| | - Renee Johnson
- Victor Chang Cardiac Research Institute, Sydney, Australia (R.J.)
- Department of Medicine, University of New South Wales, Sydney, Australia (R.J.)
| | - Daniel P. Judge
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (D.P.J.)
| | - Najim Lahrouchi
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, University of Amsterdam, the Netherlands (N.L., R.W.)
| | - Ronald H. Lekanne Deprez
- Department of Clinical Genetics, Amsterdam University Medical Center location Academic Medical Center, the Netherlands (R.H.L.D.)
| | - R. Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK (R.T.L.)
- Health Data Research UK London, University College London, UK (R.T.L.)
- University College London British Heart Foundation Research Accelerator, London, United Kingdom (R.T.L.)
| | - Francesco Mazzarotto
- Cardiovascular Research Center, Royal Brompton and Harefield Hospitals, National Health Service Foundation Trust, London, United Kingdom (F.M., J. Ware)
- National Heart and Lung Institute, Imperial College London, United Kingdom (F.M., J. Ware)
- Department of Clinical and Experimental Medicine, University of Florence, Italy (F.M.)
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (F.M.)
| | | | - Rebecca L. Miller
- Cardiovascular Genomics Center, Inova Heart and Vascular Institute, Falls Church, VA (R.L.M., P. Shah)
| | | | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (E.B., C.A.J., B.M.)
| | - Stacey Peters
- Department of Cardiology and Genomic Medicine, Royal Melbourne Hospital, Australia (S.P.)
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padua, Italy (R.C., K.P.)
| | - Alexandros Protonotarios
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom (A.P., P. Syrris)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, University of Sydney, Australia (C.S.)
| | - Palak Shah
- Cardiovascular Genomics Center, Inova Heart and Vascular Institute, Falls Church, VA (R.L.M., P. Shah)
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom (A.P., P. Syrris)
| | - Courtney Thaxton
- Department of Genetics, University of North Carolina, Chapel Hill (C.T.)
| | - J. Peter van Tintelen
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, The Netherlands (J.P.v.T.)
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, University of Amsterdam, the Netherlands (N.L., R.W.)
| | - Jessica Wang
- Department of Medicine, University of California, Los Angeles (J.F., J. Wang)
| | - James Ware
- Cardiovascular Research Center, Royal Brompton and Harefield Hospitals, National Health Service Foundation Trust, London, United Kingdom (F.M., J. Ware)
- National Heart and Lung Institute, Imperial College London, United Kingdom (F.M., J. Ware)
- Medical Research Council London Institute for Medical Sciences, Imperial College London, United Kingdom (J. Ware)
| | - Ray E. Hershberger
- Division of Human Genetics (E.J., L.P., T.A., R.E.H.), Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus
- Division of Cardiovascular Medicine (R.E.H.), Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus
| |
Collapse
|