1
|
Khattab E, Myrianthefs MM, Sakellaropoulos S, Alexandrou K, Mitsis A. Precision medicine applications in dilated cardiomyopathy: Advancing personalized care. Curr Probl Cardiol 2025; 50:103076. [PMID: 40381754 DOI: 10.1016/j.cpcardiol.2025.103076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Dilated cardiomyopathy (DCM) is a prevalent cardiac disorder affecting 1 in 250-500 individuals, characterized by ventricular dilation and impaired systolic function, leading to heart failure and increased mortality, including sudden cardiac death. DCM arises from genetic and environmental factors, such as drug-induced, inflammatory, and viral causes, resulting in diverse yet overlapping phenotypes. Advances in precision medicine are revolutionizing DCM management by leveraging genetic and molecular profiling for tailored diagnostic and therapeutic approaches. This review highlights comprehensive diagnostic evaluations, genetic discoveries, and multi-omics approaches integrating genomic, transcriptomic, proteomic, and metabolomic data to enhance understanding of DCM pathophysiology. Innovative risk stratification methods, including machine learning, are improving predictions of disease progression. Despite these advancements, the current one-size-fits-all management strategy contributes to persistently high morbidity and mortality. Emerging targeted therapies, such as CRISPR/Cas9 genome editing, aetiology-specific interventions, and pharmacogenomics, are reshaping treatment paradigms. Precision medicine holds promise for optimizing DCM diagnosis, treatment, and outcomes, aiming to reduce the burden of this debilitating condition.
Collapse
Affiliation(s)
- Elina Khattab
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Michael M Myrianthefs
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Stefanos Sakellaropoulos
- Department of Internal Medicine, Cardiology Clinic, Kantonsspital Baden, Baden 5404, Switzerland
| | - Kyriakos Alexandrou
- Department of Nursing, School of Health Sciences, Cyprus University of Technology; Archiepiskopou Kyprianou 30, Limassol 3036, Cyprus
| | - Andreas Mitsis
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus.
| |
Collapse
|
2
|
Li J, Xuan H, Kuang X, Li Y, Lian H, Yu N. Cas13b-mediated RNA targeted therapy alleviates genetic dilated cardiomyopathy in mice. Cell Biosci 2024; 14:4. [PMID: 38178244 PMCID: PMC10768345 DOI: 10.1186/s13578-023-01143-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/09/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Recent advances in gene editing technology have opened up new avenues for in vivo gene therapy, which holds great promise as a potential treatment method for dilated cardiomyopathy (DCM). The CRISPR-Cas13 system has been shown to be an effective tool for knocking down RNA expression in mammalian cells. PspCas13b, a type VI-B effector that can be packed into adeno-associated viruses and improve RNA knockdown efficiency, is a potential treatment for diseases characterized by abnormal gene expression. RESULTS Using PspCas13b, we were able to efficiently and specifically knockdown the mutant transcripts in the AC16 cell line carrying the heterozygous human TNNT2R141W (hTNNT2R141W) mutation. We used adeno-associated virus vector serotype 9 to deliver PspCas13b with specific single guide RNA into the hTNNT2R141W transgenic DCM mouse model, effectively knocking down hTNNT2R141W transcript expression. PspCas13b-mediated knockdown significantly increased myofilament sensitivity to Ca2+, improved cardiac function, and reduced myocardial fibrosis in hTNNT2R141W DCM mice. CONCLUSIONS These findings suggest that targeting genes through Cas13b is a promising approach for in vivo gene therapy for genetic diseases caused by aberrant gene expression. Our study provides further evidence of Cas13b's application in genetic disease therapy and paves the way for future applicability of genetic therapies for cardiomyopathy.
Collapse
Affiliation(s)
- Jiacheng Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - He Xuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xin Kuang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yahuan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Nie Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, 450046, China.
| |
Collapse
|
3
|
Saracino AM, Kelberman D, Otto GW, Gagunashvili A, Abraham DJ, Denton CP. Unravelling morphoea aetiopathogenesis by next-generation sequencing of paired skin biopsies. Arch Dermatol Res 2023; 315:2035-2056. [PMID: 36912952 PMCID: PMC10366313 DOI: 10.1007/s00403-023-02541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 11/24/2022] [Accepted: 01/16/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Morphoea can have a significant disease burden. Aetiopathogenesis remains poorly understood, with very limited existing genetic studies. Linear morphoea (LM) may follow Blascho's lines of epidermal development, providing potential pathogenic clues. OBJECTIVE The first objective of this study was to identify the presence of primary somatic epidermal mosaicism in LM. The second objective was tTo explore differential gene expression in morphoea epidermis and dermis to identify potential pathogenic molecular pathways and tissue layer cross-talk. METHODOLOGY Skin biopsies from paired affected and contralateral unaffected skin were taken from 16 patients with LM. Epidermis and dermis were isolated using a 2-step chemical-physical separation protocol. Whole Genome Sequencing (WGS; n = 4 epidermal) and RNA-seq (n = 5-epidermal, n = 5-dermal) with gene expression analysis via GSEA-MSigDBv6.3 and PANTHER-v14.1 pathway analyses, were performed. RTqPCR and immunohistochemistry were used to replicate key results. RESULTS Sixteen participants (93.8% female, mean age 27.7 yrs disease-onset) were included. Epidermal WGS identified no single affected gene or SNV. However, many potential disease-relevant pathogenic variants were present, including ADAMTSL1 and ADAMTS16. A highly proliferative, inflammatory and profibrotic epidermis was seen, with significantly-overexpressed TNFα-via-NFkB, TGFβ, IL6/JAKSTAT and IFN-signaling, apoptosis, p53 and KRAS-responses. Upregulated IFI27 and downregulated LAMA4 potentially represent initiating epidermal 'damage' signals and enhanced epidermal-dermal communication. Morphoea dermis exhibited significant profibrotic, B-cell and IFN-signatures, and upregulated morphogenic patterning pathways such as Wnt. CONCLUSION This study supports the absence of somatic epidermal mosaicism in LM, and identifies potential disease-driving epidermal mechanisms, epidermal-dermal interactions and disease-specific dermal differential-gene-expression in morphoea. We propose a potential molecular narrative for morphoea aetiopathogenesis which could help guide future targeted studies and therapies.
Collapse
Affiliation(s)
- Amanda M Saracino
- Division of Medicine, Centre for Rheumatology and Connective Tissues Diseases, University College London, London, UK.
- Department of Dermatology, Royal Free NHS Foundation Trust, London, UK.
- Melbourne Dermatology Clinic, 258 Park Street, South Melbourne, VIC, 3205, Australia.
| | - Daniel Kelberman
- GOSgene, Genetics and Genomic Medicine, Great Ormand Street Institute of Child Health, University College London, London, UK
| | - Georg W Otto
- GOSgene, Genetics and Genomic Medicine, Great Ormand Street Institute of Child Health, University College London, London, UK
| | - Andrey Gagunashvili
- GOSgene, Genetics and Genomic Medicine, Great Ormand Street Institute of Child Health, University College London, London, UK
| | - David J Abraham
- Division of Medicine, Centre for Rheumatology and Connective Tissues Diseases, University College London, London, UK
| | - Christopher P Denton
- Division of Medicine, Centre for Rheumatology and Connective Tissues Diseases, University College London, London, UK
- Department of Rheumatology, Royal Free NHS Foundation Trust, London, UK
| |
Collapse
|
4
|
Clinical Implication of Genetic Testing in Dilated Cardiomyopathy. INTERNATIONAL JOURNAL OF HEART FAILURE 2022; 4:1-11. [PMID: 36262197 PMCID: PMC9383343 DOI: 10.36628/ijhf.2021.0024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/29/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022]
Abstract
Dilated cardiomyopathy (DCM) is one of the important causes of heart failure (HF). With the rapidly evolving technologies for gene analysis and tremendous advances in knowledge of HF genetics, the importance of genetic testing in DCM is currently highlighted. Several genetic variants causing DCM have been identified and this information is used for diagnosis, risk stratification and family screening of DCM patients. However, there are still several challenges in applying genetic testing to real clinical practice. In this review, we will summarize recent understandings in DCM genetics and provide an evidence-based practical guide to the use of genetic testing for DCM patients.
Collapse
|
5
|
Grassi S, Campuzano O, Coll M, Cazzato F, Sarquella-Brugada G, Rossi R, Arena V, Brugada J, Brugada R, Oliva A. Update on the Diagnostic Pitfalls of Autopsy and Post-Mortem Genetic Testing in Cardiomyopathies. Int J Mol Sci 2021; 22:4124. [PMID: 33923560 PMCID: PMC8074148 DOI: 10.3390/ijms22084124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 02/08/2023] Open
Abstract
Inherited cardiomyopathies are frequent causes of sudden cardiac death (SCD), especially in young patients. Despite at the autopsy they usually have distinctive microscopic and/or macroscopic diagnostic features, their phenotypes may be mild or ambiguous, possibly leading to misdiagnoses or missed diagnoses. In this review, the main differential diagnoses of hypertrophic cardiomyopathy (e.g., athlete's heart, idiopathic left ventricular hypertrophy), arrhythmogenic cardiomyopathy (e.g., adipositas cordis, myocarditis) and dilated cardiomyopathy (e.g., acquired forms of dilated cardiomyopathy, left ventricular noncompaction) are discussed. Moreover, the diagnostic issues in SCD victims affected by phenotype-negative hypertrophic cardiomyopathy and the relationship between myocardial bridging and hypertrophic cardiomyopathy are analyzed. Finally, the applications/limits of virtopsy and post-mortem genetic testing in this field are discussed, with particular attention to the issues related to the assessment of the significance of the genetic variants.
Collapse
Affiliation(s)
- Simone Grassi
- Department of Health Surveillance and Bioethics, Section of Legal Medicine, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.C.); (R.R.); (A.O.)
| | - Oscar Campuzano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (O.C.); (M.C.); (J.B.); (R.B.)
- Cardiovascular Genetics Center, Institut d’Investigació Biomèdica Girona (IDIBGI), University of Girona, 17190 Girona, Spain
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
| | - Mònica Coll
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (O.C.); (M.C.); (J.B.); (R.B.)
- Cardiovascular Genetics Center, Institut d’Investigació Biomèdica Girona (IDIBGI), University of Girona, 17190 Girona, Spain
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
| | - Francesca Cazzato
- Department of Health Surveillance and Bioethics, Section of Legal Medicine, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.C.); (R.R.); (A.O.)
| | - Georgia Sarquella-Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
| | - Riccardo Rossi
- Department of Health Surveillance and Bioethics, Section of Legal Medicine, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.C.); (R.R.); (A.O.)
| | - Vincenzo Arena
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00147 Rome, Italy;
- Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Josep Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (O.C.); (M.C.); (J.B.); (R.B.)
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, Universitat de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ramon Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (O.C.); (M.C.); (J.B.); (R.B.)
- Cardiovascular Genetics Center, Institut d’Investigació Biomèdica Girona (IDIBGI), University of Girona, 17190 Girona, Spain
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
| | - Antonio Oliva
- Department of Health Surveillance and Bioethics, Section of Legal Medicine, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.C.); (R.R.); (A.O.)
| |
Collapse
|
6
|
Grassi S, Campuzano O, Coll M, Brión M, Arena V, Iglesias A, Carracedo Á, Brugada R, Oliva A. Genetic variants of uncertain significance: How to match scientific rigour and standard of proof in sudden cardiac death? Leg Med (Tokyo) 2020; 45:101712. [PMID: 32361481 DOI: 10.1016/j.legalmed.2020.101712] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/17/2020] [Accepted: 04/21/2020] [Indexed: 02/08/2023]
Abstract
In many SCD cases, in particular in pediatric age, autopsy can be completely negative and then a post-mortem genetic testing (molecular autopsy) is indicated. In NGS era finding new/rare variants is extremely frequent and, when only variants of unknown significance are found, molecular autopsy fails to find a cause of death. We describe the emblematic case of the sudden death of a 7-year-old girl. We performed a full-body micro-CT analysis, an accurate autopsy, a serum tryptase test and toxicological tests. Since the only macroscopic abnormality we found was a myocardial bridging (length: 1,1 cm, thickness: 0,5 cm) of the left anterior descending coronary artery, a molecular autopsy has been performed. NGS analysis on victim DNA detected rare variants in DPP6, MYH7, SCN2B and NOTCH1 and segregation analysis was then achieved. On the basis of ACMG/AMP (clinical) guidelines, all the found variants were classified as of unknown significance. In other words, both the macroscopic and genetic anomalies we found were of uncertain significance and then the autopsy failed to find the cause of the death. Our case raises three main discussion points: (a) economical, ethical and legal limitations of genetic investigation; (b) risk that genetic testing does not succeed in finding a certain cause of the death; (c) absence of specific guidelines to face the problem of VUS in forensic cases.
Collapse
Affiliation(s)
- Simone Grassi
- Institute of Public Health, Section of Legal Medicine, Catholic University, Rome, Italy
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Centro Investigación Biomédica Red Enfermedades Cardiovasculares, Madrid, Spain; Department of Biochemistry and Molecular Genetics, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Mònica Coll
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - María Brión
- Genetics of Cardiovascular and Ophthalmological Diseases, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Genomic Medicine, University of Santiago de Compostela, IDIS, CIBERER, Santiago de Compostela, Spain
| | - Vincenzo Arena
- Institute of Anatomical Pathology, Catholic University, Rome, Italy
| | - Anna Iglesias
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Ángel Carracedo
- Genomic Medicine, University of Santiago de Compostela, IDIS, CIBERER, Santiago de Compostela, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Centro Investigación Biomédica Red Enfermedades Cardiovasculares, Madrid, Spain; Cardiology Service, Hospital Josep Trueta, Girona, Spain
| | - Antonio Oliva
- Institute of Public Health, Section of Legal Medicine, Catholic University, Rome, Italy.
| |
Collapse
|
7
|
Dilated Cardiomyopathy Due to BLC2-Associated Athanogene 3 (BAG3) Mutations. J Am Coll Cardiol 2019; 72:2471-2481. [PMID: 30442290 DOI: 10.1016/j.jacc.2018.08.2181] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/12/2018] [Accepted: 08/13/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND The BAG3 (BLC2-associated athanogene 3) gene codes for an antiapoptotic protein located on the sarcomere Z-disc. Mutations in BAG3 are associated with dilated cardiomyopathy (DCM), but only a small number of cases have been reported to date, and the natural history of BAG3 cardiomyopathy is poorly understood. OBJECTIVES This study sought to describe the phenotype and prognosis of BAG3 mutations in a large multicenter DCM cohort. METHODS The study cohort comprised 129 individuals with a BAG3 mutation (62% males, 35.1 ± 15.0 years of age) followed at 18 European centers. Localization of BAG3 in cardiac tissue was analyzed in patients with truncating BAG3 mutations using immunohistochemistry. RESULTS At first evaluation, 57.4% of patients had DCM. After a median follow-up of 38 months (interquartile range: 7 to 95 months), 68.4% of patients had DCM and 26.1% who were initially phenotype-negative developed DCM. Disease penetrance in individuals >40 years of age was 80% at last evaluation, and there was a trend towards an earlier onset of DCM in men (age 34.6 ± 13.2 years vs. 40.7 ± 12.2 years; p = 0.053). The incidence of adverse cardiac events (death, left ventricular assist device, heart transplantation, and sustained ventricular arrhythmia) was 5.1% per year among individuals with DCM. Male sex, decreased left ventricular ejection fraction. and increased left ventricular end-diastolic diameter were associated with adverse cardiac events. Myocardial tissue from patients with a BAG3 mutation showed myofibril disarray and a relocation of BAG3 protein in the sarcomeric Z-disc. CONCLUSIONS DCM caused by mutations in BAG3 is characterized by high penetrance in carriers >40 years of age and a high risk of progressive heart failure. Male sex, decreased left ventricular ejection fraction, and enlarged left ventricular end-diastolic diameter are associated with adverse outcomes in patients with BAG3 mutations.
Collapse
|
8
|
Garcia-Pavia P, Kim Y, Restrepo-Cordoba MA, Lunde IG, Wakimoto H, Smith AM, Toepfer CN, Getz K, Gorham J, Patel P, Ito K, Willcox JA, Arany Z, Li J, Owens AT, Govind R, Nuñez B, Mazaika E, Bayes-Genis A, Walsh R, Finkelman B, Lupon J, Whiffin N, Serrano I, Midwinter W, Wilk A, Bardaji A, Ingold N, Buchan R, Tayal U, Pascual-Figal DA, de Marvao A, Ahmad M, Garcia-Pinilla JM, Pantazis A, Dominguez F, John Baksi A, O’Regan DP, Rosen SD, Prasad SK, Lara-Pezzi E, Provencio M, Lyon AR, Alonso-Pulpon L, Cook SA, DePalma SR, Barton PJ, Aplenc R, Seidman JG, Ky B, Ware JS, Seidman CE. Genetic Variants Associated With Cancer Therapy-Induced Cardiomyopathy. Circulation 2019; 140:31-41. [PMID: 30987448 PMCID: PMC6613726 DOI: 10.1161/circulationaha.118.037934] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cancer therapy-induced cardiomyopathy (CCM) is associated with cumulative drug exposures and preexisting cardiovascular disorders. These parameters incompletely account for substantial interindividual susceptibility to CCM. We hypothesized that rare variants in cardiomyopathy genes contribute to CCM. METHODS We studied 213 patients with CCM from 3 cohorts: retrospectively recruited adults with diverse cancers (n=99), prospectively phenotyped adults with breast cancer (n=73), and prospectively phenotyped children with acute myeloid leukemia (n=41). Cardiomyopathy genes, including 9 prespecified genes, were sequenced. The prevalence of rare variants was compared between CCM cohorts and The Cancer Genome Atlas participants (n=2053), healthy volunteers (n=445), and an ancestry-matched reference population. Clinical characteristics and outcomes were assessed and stratified by genotypes. A prevalent CCM genotype was modeled in anthracycline-treated mice. RESULTS CCM was diagnosed 0.4 to 9 years after chemotherapy; 90% of these patients received anthracyclines. Adult patients with CCM had cardiovascular risk factors similar to the US population. Among 9 prioritized genes, patients with CCM had more rare protein-altering variants than comparative cohorts ( P≤1.98e-04). Titin-truncating variants (TTNtvs) predominated, occurring in 7.5% of patients with CCM versus 1.1% of The Cancer Genome Atlas participants ( P=7.36e-08), 0.7% of healthy volunteers ( P=3.42e-06), and 0.6% of the reference population ( P=5.87e-14). Adult patients who had CCM with TTNtvs experienced more heart failure and atrial fibrillation ( P=0.003) and impaired myocardial recovery ( P=0.03) than those without. Consistent with human data, anthracycline-treated TTNtv mice and isolated TTNtv cardiomyocytes showed sustained contractile dysfunction unlike wild-type ( P=0.0004 and P<0.002, respectively). CONCLUSIONS Unrecognized rare variants in cardiomyopathy-associated genes, particularly TTNtvs, increased the risk for CCM in children and adults, and adverse cardiac events in adults. Genotype, along with cumulative chemotherapy dosage and traditional cardiovascular risk factors, improves the identification of patients who have cancer at highest risk for CCM. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov . Unique identifiers: NCT01173341; AAML1031; NCT01371981.
Collapse
Affiliation(s)
- Pablo Garcia-Pavia
- Hospital Universitario Puerta de Hierro, Madrid, Spain (P.G.-P., M.A.R.-C., F.D., L.A.-P.)
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- University Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain (P.G.-P.)
| | - Yuri Kim
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- Massachusetts General Hospital, Boston (Y.K.)
| | - Maria Alejandra Restrepo-Cordoba
- Hospital Universitario Puerta de Hierro, Madrid, Spain (P.G.-P., M.A.R.-C., F.D., L.A.-P.)
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
| | - Ida G. Lunde
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- Oslo University Hospital and University of Oslo, Norway (I.G.L.)
| | - Hiroko Wakimoto
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
| | - Amanda M. Smith
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Christopher N. Toepfer
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- University of Oxford (C.N.T.)
| | - Kelly Getz
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Joshua Gorham
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
| | - Parth Patel
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- Brigham and Women's Hospital, Boston MA (P.P., C.E.S.)
| | - Kaoru Ito
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
| | - Jonathan A. Willcox
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
| | - Zoltan Arany
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Jian Li
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Anjali T. Owens
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Risha Govind
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Beatriz Nuñez
- Hospital Universitario Puerta de Hierro, Universidad Autónoma de Madrid, Spain (B.N., M.P.)
| | - Erica Mazaika
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Antoni Bayes-Genis
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- Hospital Universitario Germans Trias i Pujol, Badalona, Spain (A.B.-G., J. Lupon)
| | - Roddy Walsh
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Brian Finkelman
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Josep Lupon
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- Hospital Universitario Germans Trias i Pujol, Badalona, Spain (A.B.-G., J. Lupon)
| | - Nicola Whiffin
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
| | - Isabel Serrano
- Hospital Universitario de Tarragona Joan XXIII. IISPV, Rovira Virgili University, Spain (I.S., A.B.)
| | - William Midwinter
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Alicja Wilk
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Alfredo Bardaji
- Hospital Universitario de Tarragona Joan XXIII. IISPV, Rovira Virgili University, Spain (I.S., A.B.)
| | - Nathan Ingold
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Rachel Buchan
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Upasana Tayal
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Domingo A. Pascual-Figal
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- Hospital Universitario Virgen de la Arrixaca, University of Murcia. Spain (D.A.P.-F.)
| | - Antonio de Marvao
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
| | - Mian Ahmad
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Jose Manuel Garcia-Pinilla
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- Hospital Universitario Virgen de la Victoria, IBIMA, Malaga, Spain (J.M.G.-P.)
| | - Antonis Pantazis
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Fernando Dominguez
- Hospital Universitario Puerta de Hierro, Madrid, Spain (P.G.-P., M.A.R.-C., F.D., L.A.-P.)
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
| | - A. John Baksi
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Declan P. O’Regan
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
| | - Stuart D. Rosen
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Sanjay K. Prasad
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Enrique Lara-Pezzi
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (E.L.-P.)
| | - Mariano Provencio
- Hospital Universitario Puerta de Hierro, Universidad Autónoma de Madrid, Spain (B.N., M.P.)
| | - Alexander R. Lyon
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Luis Alonso-Pulpon
- Hospital Universitario Puerta de Hierro, Madrid, Spain (P.G.-P., M.A.R.-C., F.D., L.A.-P.)
- Centro de Investigación Biomédica en Red Enfermedades in Cardiovascular Diseases (CIBERCV), Madrid, Spain (P.G.-P., M.A.R.-C., A.B.-G., J. Lupon, D.A.P.-F., J.M.G.-P., F.D., E.L.-P., L.A.-P.)
| | - Stuart A. Cook
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
- National Heart Centre Singapore and Duke-National University of Singapore (S.A.C.)
| | - Steven R. DePalma
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (S.R.D., C.E.S.)
| | - Paul J.R. Barton
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
| | - Richard Aplenc
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - Jonathan G. Seidman
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
| | - Bonnie Ky
- Perelman School of Medicine and University of Pennsylvania Health System, Philadelphia (A.M.S., K.G., Z.A., J. Li, A.T.O., B.F., R.A., B.K.)
| | - James S. Ware
- National Heart & Lung Institute, Imperial College London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., E.L.-P., A.R.L., S.A.C., P.J.R.B., J.S.W.)
- Royal Brompton & Harefield NHS Foundation Trust, London, UK (R.G., E.M., R.W., N.W., W.M., A.W., N.I., R.B., U.T., A.d.M., M.A., A.P., A.J.B., S.D.R., S.K.P., A.R.L., P.J.R.B., J.S.W.)
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
| | - Christine E. Seidman
- Harvard Medical School, Boston, MA (Y.K., I.G.L., H.W., C.N.T., J.G., P.P., K.I., J.A.W., S.R.D., J.G.S., C.E.S.)
- MRC London Institute of Medical Sciences, Imperial College UK (N.W., D.P.O., S.A.C., J.S.W., C.E.S., A.d.M.)
- Howard Hughes Medical Institute, Chevy Chase, MD (S.R.D., C.E.S.)
- Brigham and Women's Hospital, Boston MA (P.P., C.E.S.)
| |
Collapse
|
9
|
Fang HJ, Liu BP. Prevalence of TTN mutations in patients with dilated cardiomyopathy : A meta-analysis. Herz 2019; 45:29-36. [PMID: 31209521 DOI: 10.1007/s00059-019-4825-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/05/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022]
Abstract
A meta-analysis was performed to assess the prevalence of TTN mutations in patients with dilated cardiomyopathy (DCM). Prevalence point estimates and 95% confidence intervals were computed using the logit transformation formula. The prevalence of TTN mutations in patient with DCM, familial dilated cardiomyopathy (FDCM), and sporadic dilated cardiomyopathy (SDCM) was 0.17 (95% CI: 0.14-0.19), 0.23 (95% CI: 0.20-0.26), and 0.16 (95% CI: 0.12-0.21), respectively. No individual study had a marked influence on the pooled prevalence in the meta-analysis. Meta-regression analysis between the logit event for prevalence and sample size explained 32% of between-study variance (p < 0.05). Cumulative meta-analysis confirmed the influence of sample size on the reported prevalence among the different studies. In conclusion, the present analysis suggests that TTN mutations are familial in DCM patients. More attention should be paid to TTN mutations in clinical examinations.
Collapse
Affiliation(s)
- H-J Fang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Jingwuweiqi Rd, 250021, Jinan, China
| | - B-P Liu
- Department of Epidemiology, Shandong University School of Public Health, No.44, Wenhuaxi Rd, 250012, Jinan, China.
| |
Collapse
|
10
|
Hong L, Lai H, Fang Y, Tao Y, Qiu Y. Silencing CTGF/CCN2 inactivates the MAPK signaling pathway to alleviate myocardial fibrosis and left ventricular hypertrophy in rats with dilated cardiomyopathy. J Cell Biochem 2018; 119:9519-9531. [PMID: 30129221 DOI: 10.1002/jcb.27268] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Lang Hong
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Heng‐Li Lai
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yan Fang
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yu Tao
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yun Qiu
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| |
Collapse
|
11
|
Mak TSH, Lee YK, Tang CS, Hai JSH, Ran X, Sham PC, Tse HF. Coverage and diagnostic yield of Whole Exome Sequencing for the Evaluation of Cases with Dilated and Hypertrophic Cardiomyopathy. Sci Rep 2018; 8:10846. [PMID: 30022097 PMCID: PMC6052112 DOI: 10.1038/s41598-018-29263-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
Targeted next generation sequencing of gene panels has become a popular tool for the genetic diagnosis of hypertrophic (HCM) and dilated cardiomyopathy (DCM). However, it is uncertain whether the use of Whole Exome Sequencing (WES) represents a more effective approach for diagnosis of cases with HCM and DCM. In this study, we performed indirect comparisons of the coverage and diagnostic yield of WES on genes and variants related to HCM and DCM versus 4 different commercial gene panels using 40 HCM and DCM patients, assuming perfect coverage in those panels. We identified 6 pathogenic or likely pathogenic among 14 HCM patients (diagnostic yield 43%). 3 pathogenic or likely pathogenic were found among the 26 DCM patients (diagnostic yield 12%). The coverage was similar to that of four existing commercial gene panels due to the clustering of mutation within MYH7, MYBPC3, TPM1, TNT2, and TTN. Moreover, the coverage of WES appeared inadequate for TNNI3 and PLN. We conclude that most of the pathogenic variants for HCM and DCM can be found within a small number of genes which were covered by all the commercial gene panels, and the application of WES did not increase diagnostic yield.
Collapse
Affiliation(s)
- Timothy Shin Heng Mak
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yee-Ki Lee
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Clara S Tang
- Department of Surgery, the University of Hong Kong, Hong Kong, China
- The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong, China
| | - JoJo S H Hai
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Xinru Ran
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Pak-Chung Sham
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.
| | - Hung-Fat Tse
- Department of Medicine, the University of Hong Kong, Hong Kong, China.
- Department of Medicine, Shenzhen Hong Kong University Hospital, Shenzhen, China.
- Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, the University of Hong Kong, Hong Kong, China.
- Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
12
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
13
|
Dookhun MN, Sun Y, Zou H, Cao X, Lu X. Classification of New Biomarkers of Dilated Cardiomyopathy Based on Pathogenesis—An Update. Health (London) 2018. [DOI: 10.4236/health.2018.103024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
14
|
Xu JH, Gu JY, Guo YH, Zhang H, Qiu XB, Li RG, Shi HY, Liu H, Yang XX, Xu YJ, Qu XK, Yang YQ. Prevalence and Spectrum of NKX2-5 Mutations Associated With Sporadic Adult-Onset Dilated Cardiomyopathy. Int Heart J 2017; 58:521-529. [PMID: 28690296 DOI: 10.1536/ihj.16-440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dilated cardiomyopathy (DCM), the most common form of primary myocardial disease, is a leading cause of congestive heart failure and the most common indication for heart transplantation. Recently, NKX2-5 mutations have been involved in the pathogenesis of familial DCM. However, the prevalence and spectrum of NKX2-5 mutations associated with sporadic DCM remain to be evaluated. In this study, the coding regions and flanking introns of the NKX2-5 gene, which encodes a cardiac transcription factor pivotal for cardiac development and structural remodeling, were sequenced in 210 unrelated patients with sporadic adult-onset DCM. A total of 300 unrelated healthy individuals used as controls were also genotyped for NKX2-5. The functional effect of the mutant NKX2-5 was investigated using a dual-luciferase reporter assay system. As a result, two novel heterozygous NKX2-5 mutations, p.R139W and p.E167X, were identified in 2 unrelated patients with sporadic adult-onset DCM, with a mutational prevalence of approximately 0.95%. The mutations were absent in 600 referential chromosomes and the altered amino acids were completely conserved evolutionarily across species. Functional assays revealed that the NKX2-5 mutants were associated with significantly reduced transcriptional activity. Furthermore, the mutations abrogated the synergistic activation between NKX2-5 and GATA4 as well as TBX20, two other cardiac key transcription factors that have been causally linked to adult-onset DCM. This study is the first to associate NKX2-5 loss-of-function mutations with enhanced susceptibility to sporadic DCM, which provides novel insight into the molecular etiology underpinning DCM, and suggests the potential implications for the genetic counseling and personalized treatment of the DCM patients.
Collapse
Affiliation(s)
- Jia-Hong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Jian-Yun Gu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Yu-Han Guo
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Hong Zhang
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hong-Yu Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hua Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xiao-Xiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Cardiovascular Research Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University
| |
Collapse
|
15
|
Chebib FT, Hogan MC, El-Zoghby ZM, Irazabal MV, Senum SR, Heyer CM, Madsen CD, Cornec-Le Gall E, Behfar A, Harris PC, Torres VE. Autosomal Dominant Polycystic Kidney Patients May Be Predisposed to Various Cardiomyopathies. Kidney Int Rep 2017; 2:913-923. [PMID: 29270497 PMCID: PMC5733883 DOI: 10.1016/j.ekir.2017.05.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/11/2017] [Accepted: 05/28/2017] [Indexed: 01/18/2023] Open
Abstract
Introduction Mutations in PKD1 and PKD2 cause autosomal dominant polycystic kidney disease (ADPKD). Experimental evidence suggests an important role of the polycystins in cardiac development and myocardial function. To determine whether ADPKD may predispose to the development of cardiomyopathy, we have evaluated the coexistence of diagnoses of ADPKD and primary cardiomyopathy in our patients. Methods Clinical data were retrieved from medical records for patients with a coexisting diagnosis of ADPKD and cardiomyopathies evaluated at the Mayo Clinic (1984-2015). Results Among the 58 of 667 patients with available echocardiography data, 39 (5.8%) had idiopathic dilated cardiomyopathy (IDCM), 17 (2.5%) had hypertrophic obstructive cardiomyopathy, and 2 (0.3%) had left ventricular noncompaction. Genetic data were available for 19, 8, and 2 cases of IDCM, hypertrophic obstructive cardiomyopathy, and left ventricular noncompaction, respectively. PKD1 mutations were detected in 42.1%, 62.5%, and 100% of IDCM, hypertrophic obstructive cardiomyopathy, and left ventricular noncompaction cases, respectively. PKD2 mutations were detected only in IDCM cases and were overrepresented (36.8%) relative to the expected frequency in ADPKD (15%). In at least 1 patient from 3 IDMC families and 1 patient from a hypertrophic obstructive cardiomyopathy family, the cardiomyopathy did not segregate with ADPKD, suggesting that the PKD mutations may be predisposing factors rather than solely responsible for the development of cardiomyopathy. Discussion Coexistence of ADPKD and cardiomyopathy in our tertiary referral center cohort appears to be higher than expected by chance. We suggest that PKD1 and PKD2 mutations may predispose to primary cardiomyopathies and that genetic interactions may account for the observed coexistence of ADPKD and cardiomyopathies.
Collapse
Affiliation(s)
- Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Marie C Hogan
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Ziad M El-Zoghby
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Maria V Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Christina M Heyer
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Charles D Madsen
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Emilie Cornec-Le Gall
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Atta Behfar
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Park HY. Hereditary Dilated Cardiomyopathy: Recent Advances in Genetic Diagnostics. Korean Circ J 2017; 47:291-298. [PMID: 28567076 PMCID: PMC5449520 DOI: 10.4070/kcj.2016.0017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 10/28/2016] [Accepted: 11/11/2016] [Indexed: 01/07/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is the most common cause of heart failure in young adults and up to 50% of idiopathic DCM is thought to be caused by genetic mutations in candidate genes. Although a genetic diagnosis can confirm a clinical diagnosis of hereditary DCM, genetic testing has not been easily accessible due to genetic heterogeneity and complexity. Next-generation sequencing (NGS) technologies have recently been introduced, and genetic testing for multiple genes is currently available and more than 40 different genes have been associated with DCM. In Korea, the government has supported genetic diagnosis for patients with idiopathic DCM. When a targeted gene panel with NGS technology was used, the detection rate was about 40%. MYBPC3, LMNA, and MYH7 were the most frequently identified genes, and the pattern of causative genes was different from previous reports. In the analysis, a significant number of subjects (42.0%) had rare or novel unspecified variants in DCM candidate genes, which should be assessed as potential causative mutations. Developing a more comprehensive test panel with additional DCM genes and whole exome sequencing will improve the detection rate, and allow genetic testing to be an option for patients with idiopathic DCM. However, all genetic variations are not pathogenic mutations, and the majority of reported mutations in DCM are unique to a single family, which makes genetic data interpretation more difficult. Therefore, clinical features and familial history integration are needed to improve clinical decision making.
Collapse
Affiliation(s)
- Hyun-Young Park
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Science, Korea National Institute of Health, Cheongju, Korea
| |
Collapse
|
17
|
de Gonzalo-Calvo D, Quezada M, Campuzano O, Perez-Serra A, Broncano J, Ayala R, Ramos M, Llorente-Cortes V, Blasco-Turrión S, Morales FJ, Gonzalez P, Brugada R, Mangas A, Toro R. Familial dilated cardiomyopathy: A multidisciplinary entity, from basic screening to novel circulating biomarkers. Int J Cardiol 2017; 228:870-880. [PMID: 27889554 DOI: 10.1016/j.ijcard.2016.11.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/23/2016] [Accepted: 11/05/2016] [Indexed: 12/11/2022]
Abstract
Idiopathic dilated cardiomyopathy has become one of the most prevalent inherited cardiomyopathies over the past decades. Genetic screening of first-degree relatives has revealed that 30-50% of the cases have a familial origin. Similar to other heart diseases, familial dilated cardiomyopathy is characterized by a high genetic heterogeneity that complicates family studies. Cli'nical screening, 12-lead electrocardiogram and transthoracic echocardiogram are recommended for patients and first-degree family members. Magnetic resonance also needs to be considered. Genetic technologies have become fundamental for the clinical management of this disease. New generation sequencing methods have made genetic testing feasible for extensive panels of genes related to the disease. Recently, new imaging modalities such as speckle-tracking, strain and strain rate or magnetic resonance, and circulating biomarkers such as non-coding RNAs, have emerged as potential strategies to help cardiologists in their clinical practice. Imaging, genetic and blood-based techniques should be considered together in the evaluation and testing of familial dilated cardiomyopathy. Here, we discuss the current procedures and novel approaches for the clinical management of familial dilated cardiomyopathy.
Collapse
Affiliation(s)
- D de Gonzalo-Calvo
- Cardiovascular Research Center, CSIC-ICCC, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - M Quezada
- Hospital Universitario de la Cruz Roja, Madrid, Spain
| | - O Campuzano
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain; Medical Science Department, School of Medicine, Girona, Spain
| | - A Perez-Serra
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain
| | - J Broncano
- Cardiothoracic Imaging Section, Hospital Cruz Roja, Hospital San Juan de Dios. Ressalta, Health Time Group, Córdoba, Spain
| | - R Ayala
- Hospital Universitario de la Cruz Roja, Madrid, Spain
| | - M Ramos
- Hospital Universitario de la Cruz Roja, Madrid, Spain
| | - V Llorente-Cortes
- Cardiovascular Research Center, CSIC-ICCC, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - S Blasco-Turrión
- Cardiology Department, Puerto Real Universitary Hospital, Cádiz, Spain
| | - F J Morales
- Cardiology Department, Puerto Real Universitary Hospital, Cádiz, Spain
| | - P Gonzalez
- Cardiology Department, Puerto Real Universitary Hospital, Cádiz, Spain
| | - R Brugada
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain; Medical Science Department, School of Medicine, Girona, Spain; Cardiology Department, Hospital Josep Trueta, Girona, Spain
| | - A Mangas
- Medicine Department, School of Medicine, University of Cadiz, Cádiz, Spain
| | - R Toro
- Medicine Department, School of Medicine, University of Cadiz, Cádiz, Spain.
| |
Collapse
|
18
|
Qiu XB, Qu XK, Li RG, Liu H, Xu YJ, Zhang M, Shi HY, Hou XM, Liu X, Yuan F, Sun YM, Wang J, Huang RT, Xue S, Yang YQ. CASZ1 loss-of-function mutation contributes to familial dilated cardiomyopathy. ACTA ACUST UNITED AC 2017; 55:1417-1425. [DOI: 10.1515/cclm-2016-0612] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 12/09/2016] [Indexed: 02/06/2023]
Abstract
AbstractBackground:The zinc finger transcription factor CASZ1 plays a key role in cardiac development and postnatal adaptation, and in mice, deletion of theMethods:The coding exons and splicing junction sites of theResults:A novel heterozygous CASZ1 mutation, p.K351X, was identified in an index patient with DCM. Genetic analysis of the mutation carrier’s family showed that the mutation co-segregated with DCM, which was transmitted in an autosomal dominant pattern with complete penetrance. The nonsense mutation, which was absent in 400 referential chromosomes, altered the amino acid that was highly conserved evolutionarily. Biological investigations revealed that the mutant CASZ1 had no transcriptional activity.Conclusions:The current study reveals
Collapse
|
19
|
Pérez-Serra A, Toro R, Sarquella-Brugada G, de Gonzalo-Calvo D, Cesar S, Carro E, Llorente-Cortes V, Iglesias A, Brugada J, Brugada R, Campuzano O. Genetic basis of dilated cardiomyopathy. Int J Cardiol 2016; 224:461-472. [PMID: 27736720 DOI: 10.1016/j.ijcard.2016.09.068] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/15/2016] [Accepted: 09/17/2016] [Indexed: 01/19/2023]
Abstract
Dilated cardiomyopathy is a rare cardiac disease characterized by left ventricular dilatation and systolic dysfunction leading to heart failure and sudden cardiac death. Currently, despite several conditions have been reported as aetiologies of the disease, a large number of cases remain classified as idiopathic. Recent studies determine that nearly 60% of cases are inherited, therefore due to a genetic cause. Progressive technological advances in genetic analysis have identified over 60 genes associated with this entity, being TTN the main gene, so far. All these genes encode a wide variety of myocyte proteins, mainly sarcomeric and desmosomal, but physiopathologic pathways are not yet completely unraveled. We review the recent published data about genetics of familial dilated cardiomyopathy.
Collapse
Affiliation(s)
| | - Rocio Toro
- Medicine Department, School of Medicine, Cadiz, Spain
| | | | - David de Gonzalo-Calvo
- Cardiovascular Research Center (CSIC-ICCC), Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sergi Cesar
- Arrhythmias Unit, Sant Joan de Deu Hospital, University of Barcelona, Barcelona, Spain
| | - Esther Carro
- Arrhythmias Unit, Sant Joan de Deu Hospital, University of Barcelona, Barcelona, Spain
| | - Vicenta Llorente-Cortes
- Cardiovascular Research Center (CSIC-ICCC), Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Anna Iglesias
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain
| | - Josep Brugada
- Arrhythmias Unit, Sant Joan de Deu Hospital, University of Barcelona, Barcelona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Cardiomyopathy Unit, Hospital Josep Trueta, University of Girona, Girona, Spain.
| | - Oscar Campuzano
- Cardiovascular Genetics Center, IDIBGI, University of Girona, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain
| |
Collapse
|
20
|
Zhou YM, Dai XY, Huang RT, Xue S, Xu YJ, Qiu XB, Yang YQ. A novel TBX20 loss-of-function mutation contributes to adult-onset dilated cardiomyopathy or congenital atrial septal defect. Mol Med Rep 2016; 14:3307-14. [DOI: 10.3892/mmr.2016.5609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
|
21
|
Cuenca S, Ruiz-Cano MJ, Gimeno-Blanes JR, Jurado A, Salas C, Gomez-Diaz I, Padron-Barthe L, Grillo JJ, Vilches C, Segovia J, Pascual-Figal D, Lara-Pezzi E, Monserrat L, Alonso-Pulpon L, Garcia-Pavia P. Genetic basis of familial dilated cardiomyopathy patients undergoing heart transplantation. J Heart Lung Transplant 2016; 35:625-35. [PMID: 26899768 DOI: 10.1016/j.healun.2015.12.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/26/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is the most frequent cause of heart transplantation (HTx). The genetic basis of DCM among patients undergoing HTx has been poorly characterized. We sought to determine the genetic basis of familial DCM HTx and to establish the yield of modern next generation sequencing (NGS) technologies in this setting. METHODS Fifty-two heart-transplanted patients due to familial DCM underwent NGS genetic evaluation with a panel of 126 genes related to cardiac conditions (59 associated with DCM). Genetic variants were initially classified as pathogenic mutations or as variants of uncertain significance (VUS). Final pathogenicity status was determined by familial cosegregation studies. RESULTS Initially, 24 pathogenic mutations were found in 21 patients (40%); 25 patients (48%) carried 19 VUS and 6 (12%) did not show any genetic variant. Familial evaluation of 220 relatives from 36 of the 46 families with genetic variants confirmed pathogenicity in 14 patients and allowed reclassification of VUS as pathogenic in 17 patients, and as non-pathogenic in 3 cases. At the end of the study, the DCM-causing mutation was identified in 38 patients (73%) and 5 patients (10%) harbored only VUS. No genetic variants were identified in 9 cases (17%). CONCLUSIONS The genetic spectrum of familial DCM patients undergoing HTx is heterogeneous and involves multiple genes. NGS technology plus detailed familial studies allow identification of causative mutations in the vast majority of familial DCM cases. Detailed familial studies remain critical to determine the pathogenicity of underlying genetic defects in a substantial number of cases.
Collapse
Affiliation(s)
- Sofia Cuenca
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Maria J Ruiz-Cano
- Heart Failure and Heart Transplantation Unit, Department of Cardiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Alfonso Jurado
- Heart Failure and Heart Transplantation Unit, Department of Cardiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Clara Salas
- Department of Pathology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Laura Padron-Barthe
- Myocardial Biology Programme, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jose Javier Grillo
- Department of Cardiology, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Carlos Vilches
- Department of Immunology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Javier Segovia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Domingo Pascual-Figal
- Department of Cardiology, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Enrique Lara-Pezzi
- Myocardial Biology Programme, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Luis Alonso-Pulpon
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain; Myocardial Biology Programme, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| | | |
Collapse
|
22
|
Aherrahrou Z, Schlossarek S, Stoelting S, Klinger M, Geertz B, Weinberger F, Kessler T, Aherrahrou R, Moreth K, Bekeredjian R, Hrabě de Angelis M, Just S, Rottbauer W, Eschenhagen T, Schunkert H, Carrier L, Erdmann J. Knock-out of nexilin in mice leads to dilated cardiomyopathy and endomyocardial fibroelastosis. Basic Res Cardiol 2015; 111:6. [PMID: 26659360 DOI: 10.1007/s00395-015-0522-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 11/19/2015] [Indexed: 12/01/2022]
Abstract
Cardiomyopathy is one of the most common causes of chronic heart failure worldwide. Mutations in the gene encoding nexilin (NEXN) occur in patients with both hypertrophic and dilated cardiomyopathy (DCM); however, little is known about the pathophysiological mechanisms and relevance of NEXN to these disorders. Here, we evaluated the functional role of NEXN using a constitutive Nexn knock-out (KO) mouse model. Heterozygous (Het) mice were inter-crossed to produce wild-type (WT), Het, and homozygous KO mice. At birth, 32, 46, and 22 % of the mice were WT, Het, and KO, respectively, which is close to the expected Mendelian ratio. After postnatal day 6, the survival of the Nexn KO mice decreased dramatically and all of the animals died by day 8. Phenotypic characterizations of the WT and KO mice were performed at postnatal days 1, 2, 4, and 6. At birth, the relative heart weights of the WT and KO mice were similar; however, at day 4, the relative heart weight of the KO group was 2.3-fold higher than of the WT group. In addition, the KO mice developed rapidly progressive cardiomyopathy with left ventricular dilation and wall thinning and decreased cardiac function. At day 6, the KO mice developed a fulminant DCM phenotype characterized by dilated ventricular chambers and systolic dysfunction. At this stage, collagen deposits and some elastin deposits were observed within the left ventricle cavity, which resembles the features of endomyocardial fibroelastosis (EFE). Overall, these results further emphasize the role of NEXN in DCM and suggest a novel role in EFE.
Collapse
Affiliation(s)
- Zouhair Aherrahrou
- Institute for Integrative and Experimental Genomics, University of Lübeck, 23562, Lübeck, Germany. .,DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany. .,University Heart Center Luebeck, 23562, Lübeck, Germany.
| | - Saskia Schlossarek
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephanie Stoelting
- Institute for Integrative and Experimental Genomics, University of Lübeck, 23562, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany
| | | | - Birgit Geertz
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Weinberger
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany
| | - Redouane Aherrahrou
- Institute for Integrative and Experimental Genomics, University of Lübeck, 23562, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany
| | - Kristin Moreth
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Raffi Bekeredjian
- Department of Cardiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany.,German Center for Diabetes Research (DZD), Ingostädter Landstr. 1, 85764, Neuherberg, Germany
| | - Steffen Just
- Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Wolfgang Rottbauer
- Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Thomas Eschenhagen
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,DZHK (German Research Centre for Cardiovascular Research), partner site Munich Heart Alliance (MHA), Munich, Germany
| | - Lucie Carrier
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jeanette Erdmann
- Institute for Integrative and Experimental Genomics, University of Lübeck, 23562, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck/Hamburg, Germany.,University Heart Center Luebeck, 23562, Lübeck, Germany
| |
Collapse
|
23
|
Association of Nicotinamide Phosphoribosyltransferase (NAMPT) Gene Polymorphisms and of Serum NAMPT Levels with Dilated Cardiomyopathy in a Chinese Population. Int J Mol Sci 2015; 16:22299-318. [PMID: 26389889 PMCID: PMC4613309 DOI: 10.3390/ijms160922299] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 02/05/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) has crucial roles for myocardial development, cardiomyocyte energy metabolism and cell death/survival by regulating NAD+-dependent sirtuin-1 (SIRT1) deacetylase. This study aimed to determine if the single nucleotide polymorphisms (SNPs) of the NAMPT gene may affect the susceptibility and prognosis for patients with dilated cardiomyopathy (DCM) and to describe the association of serum NAMPT levels with clinical features of DCM. Three SNPs (rs61330082, rs2505568, and rs9034) were analyzed by the polymerase chain reaction-restriction fragment length polymorphism method in a case-control study of 394 DCM patients and 395 controls from China. Serum NAMPT levels were measured by enzyme-linked immunosorbent assay kits. The homozygote for the minor allele at rs2505568 and rs9034 could not be detected in this study. Rs9034 T allele and CT genotype were associated with increased DCM risk (OR: 1.63, 95% CI = 1.16–2.27, p = 0.005 and OR: 1.72, 95% CI = 1.20–2.50, p = 0.0027, respectively). Nominally significant decreased DCM risk was found to be associated with the A allele and AT genotype of rs2505568 (OR: 0.48, 95% CI = 0.35–0.67, p < 0.0001 and OR: 0.44, 95% CI = 0.31–0.62, p < 0.0001, respectively), but it should be interpreted with caution because of Hardy-Weinberg disequilibrium in the control group. Of five haplotypes constructed, TAC (rs61330082-rs2505568-rs9034) was a protective haplotype to DCM (OR: 0.22, 95% CI = 0.13–0.39, p = 1.84 × 10−8). The Cox multivariate survival analysis indicated that the rs9034 CT genotype (hazard ratio (HR): 0.59, 95% CI = 0.37–0.96, p = 0.03) was an independently multivariate predictor for longer overall survival in DCM patients. Serum NAMPT levels were significantly higher in the DCM group than controls (p < 0.0001) and gradually increased with the increase of New York Heart Association grade in DCM patients. However, there was a lack of association of the three SNPs with serum NAMPT levels. Spearman correlation test revealed that the NAMPT level was positively associated with brain natriuretic peptide (r = 0.56, p = 0.001), left ventricular end-diastolic diameter (r = 0.293, p = 0.011) and left ventricular end-diastolic volume (r = 0.294, p = 0.011). Our study suggested that NAMPT may play an important role in the development of DCM.
Collapse
|
24
|
Coats CJ, Heywood WE, Mills K, Elliott PM. Current applications of biomarkers in cardiomyopathies. Expert Rev Cardiovasc Ther 2015; 13:825-37. [DOI: 10.1586/14779072.2015.1053873] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Zhou W, Zhao L, Jiang JQ, Jiang WF, Yang YQ, Qiu XB. A novel TBX5 loss-of-function mutation associated with sporadic dilated cardiomyopathy. Int J Mol Med 2015; 36:282-8. [PMID: 25963046 DOI: 10.3892/ijmm.2015.2206] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 05/07/2015] [Indexed: 01/08/2023] Open
Abstract
Dilated cardiomyopathy (DCM) represents the most prevalent form of primary cardiomyopathy, and is the most common reason for heart transplantation and a major cause of congestive heart failure. Aggregating evidence demonstrates that genetic defects are associated with DCM, and a great number of mutations in >50 genes have been linked to DCM. However, DCM is a genetically heterogeneous disorder and the genetic components underpinning DCM in a significant proportion of patients remain unknown. In the present study, the coding exons and flanking exon‑intron boundaries of the T-Box 5 (TBX5) gene, which encodes a T‑box transcription factor required for normal cardiac development, were sequenced in 146 unrelated patients with sporadic DCM. The functional characteristics of the mutant TBX5 were assayed in contrast to its wild‑type counterpart by using a dual‑luciferase reporter assay system. As a result, a novel heterozygous TBX5 mutation, p.A143T, was identified in a patient with sporadic DCM. The missense mutation, which was absent in 400 control chromosomes, altered the amino acid that was completely conserved evolutionarily among species. Biological analyses revealed that the A143T mutation of TBX5 was associated with significantly decreased transcriptional activity on the promoter of the target gene atrial natriuretic factor (ANF), when compared to its wild‑type counterpart. Furthermore, the A143T mutation abolished the synergistic activation of the ANF promoter between TBX5 and GATA binding protein 4 (GATA4), another crucial transcriptional factor for heart development. To the best of our knowledge, this is the first report on the association of a TBX5 loss‑of‑function mutation with an enhanced susceptibility to sporadic DCM, providing novel insight into the molecular mechanisms of the pathogenesis of DCM and suggesting potential implications for the prenatal prophylaxis and personalized treatment of this commonest primary myocardial disease.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Emergency Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Lan Zhao
- Department of Cardiology, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Jin-Qi Jiang
- Department of Emergency Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Wei-Feng Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
26
|
Yang J, Xu WW, Hu SJ. Heart failure: advanced development in genetics and epigenetics. BIOMED RESEARCH INTERNATIONAL 2015; 2015:352734. [PMID: 25949994 PMCID: PMC4407520 DOI: 10.1155/2015/352734] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 02/25/2015] [Accepted: 03/19/2015] [Indexed: 01/16/2023]
Abstract
Heart failure (HF) is a complex pathophysiological syndrome that arises from a primary defect in the ability of the heart to take in and/or eject sufficient blood. Genetic mutations associated with familial dilated cardiomyopathy, hypertrophic cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy can contribute to the various pathologies of HF. Therefore, genetic screening could be an approach for guiding individualized therapies and surveillance. In addition, epigenetic regulation occurs via key mechanisms, including ATP-dependent chromatin remodeling, DNA methylation, histone modification, and RNA-based mechanisms. MicroRNA is also a hot spot in HF research. This review gives an overview of genetic mutations associated with cardiomyopathy and the roles of some epigenetic mechanisms in HF.
Collapse
Affiliation(s)
- Jian Yang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qing-Chun Road, Hangzhou 310003, China
| | - Wei-wei Xu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qing-Chun Road, Hangzhou 310003, China
| | - Shen-jiang Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qing-Chun Road, Hangzhou 310003, China
| |
Collapse
|
27
|
Alcalde M, Campuzano O, Sarquella-Brugada G, Arbelo E, Allegue C, Partemi S, Iglesias A, Oliva A, Brugada J, Brugada R. Clinical interpretation of genetic variants in arrhythmogenic right ventricular cardiomyopathy. Clin Res Cardiol 2015; 104:288-303. [PMID: 25398255 DOI: 10.1007/s00392-014-0794-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/11/2014] [Indexed: 01/08/2023]
Abstract
Arrhythmogenic right ventricular cardiomyopathy is an inherited cardiac entity characterized by right ventricular, or biventricular, fibrofatty replacement of myocardium. Structural alterations may lead to sudden cardiac death, mainly in young males during exercise. Autosomal dominant pattern of inheritance is reported in most parts of pathogenic genetic variations identified. Currently, 13 genes have been associated with the disease but nearly 40 % of clinically diagnosed cases remain without a genetic diagnosis. New genetic technologies allow further genetic analysis, generating a significant amount of genetic data in novel genes, which is often classified as of ambiguous significance. We focus on genetic advances of arrhythmogenic right ventricular cardiomyopathy, helping clinicians to interpret and translate genetic data into clinical practice.
Collapse
Affiliation(s)
- Mireia Alcalde
- Cardiovascular Genetics Centre, IDIBGI-University of Girona, C/Pic de Peguera 11, 17003, Girona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yuan F, Qiu XB, Li RG, Qu XK, Wang J, Xu YJ, Liu X, Fang WY, Yang YQ, Liao DN. A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med 2014; 35:478-86. [PMID: 25503402 DOI: 10.3892/ijmm.2014.2029] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 12/05/2014] [Indexed: 01/08/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is the most prevalent type of primary myocardial disease, which is the third most common cause of heart failure and the most frequent reason for heart transplantation. Aggregating evidence demonstrates that genetic risk factors are involved in the pathogenesis of idiopathic DCM. Nevertheless, DCM is of remarkable genetic heterogeneity and the genetic defects underpinning DCM in an overwhelming majority of patients remain unknown. In the present study, the whole coding exons and splice junction sites of the NKX2-5 gene, which encodes a homeodomain transcription factor crucial for cardiac development and structural remodeling, were sequenced in 130 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped for the NKX2-5 gene. The functional effect of the mutant NKX2-5 was characterized in contrast to its wild-type counterpart using a dual-luciferase reporter assay system. As a result, a novel heterozygous NKX2-5 mutation, p.S146W, was identified in a family with DCM inherited as an autosomal dominant trait, which co-segregated with DCM in the family with complete penetrance. Notably, the mutation carriers also had arrhythmias, such as paroxysmal atrial fibrillation and atrioventricular block. The missense mutation was absent in 400 reference chromosomes and the altered amino acid was completely conserved evolutionarily among species. Functional analysis revealed that the NKX2-5 mutant was associated with a significantly reduced transcriptional activity. The findings expand the mutational spectrum of NKX2-5 linked to DCM and provide novel insight into the molecular mechanisms underlying DCM, contributing to the antenatal prophylaxis and allele-specific management of DCM.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Juan Wang
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xu Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Wei-Yi Fang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - De-Ning Liao
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
29
|
Guzzo-Merello G, Cobo-Marcos M, Gallego-Delgado M, Garcia-Pavia P. Alcoholic cardiomyopathy. World J Cardiol 2014; 6:771-781. [PMID: 25228956 PMCID: PMC4163706 DOI: 10.4330/wjc.v6.i8.771] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 05/15/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Alcohol is the most frequently consumed toxic substance in the world. Low to moderate daily intake of alcohol has been shown to have beneficial effects on the cardiovascular system. In contrast, exposure to high levels of alcohol for a long period could lead to progressive cardiac dysfunction and heart failure. Cardiac dysfunction associated with chronic and excessive alcohol intake is a specific cardiac disease known as alcoholic cardiomyopathy (ACM). In spite of its clinical importance, data on ACM and how alcohol damages the heart are limited. In this review, we evaluate available evidence linking excessive alcohol consumption with heart failure and dilated cardiomyopathy. Additionally, we discuss the clinical presentation, prognosis and treatment of ACM.
Collapse
|
30
|
Prevalence and spectrum of GATA4 mutations associated with sporadic dilated cardiomyopathy. Gene 2014; 548:174-81. [PMID: 25017055 DOI: 10.1016/j.gene.2014.07.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/12/2014] [Accepted: 07/09/2014] [Indexed: 01/14/2023]
Abstract
Dilated cardiomyopathy (DCM) is the most frequent type of primary myocardial disorder responsible for substantial morbidity and mortality. DCM is the third most common cause of heart failure and the most common reason for heart transplantation. A recent study has implicated GATA4 mutation in the pathogenesis of familial DCM. However, the prevalence and spectrum of GATA4 mutations associated with sporadic DCM remain unclear. In this study, the coding exons and exon-intron boundaries of the GATA4 gene, which encodes a cardiac transcription factor crucial for normal cardiogenesis, were sequenced in 220 unrelated patients with sporadic DCM. A total of 200 unrelated ethnically-matched healthy individuals used as controls were genotyped. The functional characteristics of the mutant GATA4 were assayed in contrast to its wild-type counterpart using a luciferase reporter assay system. As a result, 3 novel heterozygous GATA4 mutations, p.V39L, p.P226Q and p.T279S, were identified in 3 unrelated patients with sporadic DCM, with a mutational prevalence of approximately 1.36%. The missense mutations were absent in 400 control chromosomes and the altered amino acids were completely conserved evolutionarily across species. Functional analysis showed that the GATA4 mutants were consistently associated with significantly decreased transcriptional activity and markedly reduced the synergistic activation between GATA4 and NKX2-5. This study firstly links GATA4 mutations to increased susceptibility to sporadic DCM and provides novel insight into the molecular etiology underlying DCM, suggesting the potential implications for the early prophylaxis and allele-specific treatment of this common form of cardiomyopathy.
Collapse
|
31
|
Zhang M, Chen J, Si D, Zheng Y, Jiao H, Feng Z, Hu Z, Duan R. Whole exome sequencing identifies a novel EMD mutation in a Chinese family with dilated cardiomyopathy. BMC MEDICAL GENETICS 2014; 15:77. [PMID: 24997722 PMCID: PMC4105140 DOI: 10.1186/1471-2350-15-77] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/27/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Variants in the emerin gene (EMD) were implicated in X-linked recessive Emery-Dreifuss muscular dystrophy (EDMD), characterized by early-onset contractures of tendons, progressive muscular weakness and cardiomyopathy. To date, 223 mutations have been reported in EMD gene and the majority of them caused a predominant skeletal muscular phenotype. In this study, we identified a novel deletion mutation in EMD exon 1, which results in almost a complete loss of emerin protein in a large Chinese family. However, the patients suffered severe dilated cardiomyopathy (DCM) but very mild skeletal muscle disorder. CASE PRESENTATION Whole exome sequencing (WES) and linkage analysis were performed to identify the underlying mutation in a Chinese DCM family spanning five generations. A missense variation in the GPR50 gene was found co-segregated with the disease phenotype, whereas no functional alteration was detected in the variant GPR50 protein. When analyzing the failure sequences in the exome sequencing data, a novel deletion mutation (c.26_39delATACCGAGCTGACC) in EMD exon 1, was identified in this family. Different from the typical clinical features caused by most reported EMD mutations, patients in our study presented very mild skeletal muscle degeneration that had not been diagnosed until the mutation was found. CONCLUSION We described a family with rare clinical presentations caused by a novel EMD deletion mutation. Our findings broaden the heterogeneous spectrum of phenotypes attributed to EMD mutations and provide new insight to explain the genotype-phenotype correlations between EMD mutations and EDMD symptoms.
Collapse
Affiliation(s)
- Mingqiu Zhang
- China Japan Union Hospital, Jilin University, 130031 Changchun, Jilin province, China
| | - Jia Chen
- State Key Lab of Medical Genetics, Central South University, 410008 Changsha, Hunan province, China
| | - Dayong Si
- School of Life Science, Jilin University, 130031 Changchun, Jilin province, China
| | - Yu Zheng
- State Key Lab of Medical Genetics, Central South University, 410008 Changsha, Hunan province, China
| | - Haixu Jiao
- China Japan Union Hospital, Jilin University, 130031 Changchun, Jilin province, China
| | - Zhaohui Feng
- China Japan Union Hospital, Jilin University, 130031 Changchun, Jilin province, China
| | - Zhengmao Hu
- State Key Lab of Medical Genetics, Central South University, 410008 Changsha, Hunan province, China
| | - Ranhui Duan
- State Key Lab of Medical Genetics, Central South University, 410008 Changsha, Hunan province, China
| |
Collapse
|
32
|
|
33
|
Zhao L, Xu JH, Xu WJ, Yu H, Wang Q, Zheng HZ, Jiang WF, Jiang JF, Yang YQ. A novel GATA4 loss-of-function mutation responsible for familial dilated cardiomyopathy. Int J Mol Med 2013; 33:654-60. [PMID: 24366163 DOI: 10.3892/ijmm.2013.1600] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 12/20/2013] [Indexed: 11/06/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is the most common form of primary myocardial disorder and is associated with substantial morbidity and mortality. Increasing evidence suggests that genetic risk factors play an important role in the pathogenesis of idiopathic DCM. However, DCM is a genetically heterogeneous disease, and the genetic defects responsible for DCM in an overwhelming majority of cases remain to be identified. In the present study, the entire coding region and the splice junction sites of the GATA4 gene, which encodes a cardiac transcription factor essential for cardiogenesis, were sequenced in 150 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped. The functional characteristics of the mutant GATA4 were delineated in contrast to its wild-type counterpart using a luciferase reporter assay system. As a result, a novel heterozygous GATA4 mutation, p.V291L, was identified in a family with DCM inherited in an autosomal dominant pattern, which co-segregated with DCM in the family with complete penetrance. The missense mutation was absent in 400 control chromosomes, and the altered amino acid was completely conserved evolutionarily among species. Functional analysis revealed that the GATA4 mutant was associated with significantly diminished transcriptional activity. The findings expand the mutational spectrum of GATA4 linked to DCM and provide novel insight into the molecular etiology involved in DCM, suggesting the potential implications in the early prophylaxis and allele-specific treatment for this common type of cardiomyopathy.
Collapse
Affiliation(s)
- Lan Zhao
- Department of Cardiology, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Jia-Hong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Wen-Jun Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Hong Yu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Qian Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Hong-Zhen Zheng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Wei-Feng Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jin-Fa Jiang
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
34
|
Affiliation(s)
- Alison R Muir
- The Belfast Trust, First floor, West Wing, Royal Victoria Hospital, Grosvenor road, Belfast, BT12 6BA, UK
| | - Ian BA Menown
- Craigavon Cardiac Centre, Southern Trust, Craigavon, BT63 5QQ, UK
| |
Collapse
|