1
|
Figueiral M, Paldino A, Fazzini L, Pereira NL. Genetic Biomarkers in Heart Failure: From Gene Panels to Polygenic Risk Scores. Curr Heart Fail Rep 2024; 21:554-569. [PMID: 39405019 DOI: 10.1007/s11897-024-00687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide a comprehensive overview of the current understanding of genetic markers associated with heart failure (HF) and its underlying causative diseases, such as cardiomyopathies. It highlights the relevance of genetic biomarkers in diagnosing HF, predicting prognosis, potentially identifying its preclinical stages and identifying targets to enable the implementation of individualized medicine approaches. RECENT FINDINGS The prevalence of HF is increasing due to an aging population but with greater access to disease-modifying therapies. Advanced diagnostic tools such as cardiac magnetic resonance, nuclear imaging, and AI-enabled diagnostic testing are now being utilized to further characterize HF patients. Additionally, the importance of genetic testing in HF diagnosis and management is increasingly being recognized. Genetic biomarkers, including single nucleotide polymorphisms (SNPs) and rare genetic variants, are emerging as crucial tools for diagnosing HF substrates, determining prognosis and increasingly directing therapy. These genetic insights are key to optimizing HF management and delivering personalized treatment tailored to individual patients. HF is a complex syndrome affecting millions globally, characterized by high mortality and significant economic burden. Understanding the underlying etiologies of HF is essential for improving management and clinical outcomes. Recent advances highlight the use of multimodal assessments, including AI-enabled diagnostics and genetic testing, to better characterize and manage HF. Genetic biomarkers are particularly promising in identifying preclinical HF stages and providing personalized treatment options. The genetic contribution to HF is heterogeneous, with both monogenic and polygenic bases playing a role. These developments underscore the shift towards personalized medicine in HF management.
Collapse
Affiliation(s)
- Marta Figueiral
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alessia Paldino
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Luca Fazzini
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Mėlinytė-Ankudavičė K, Šukys M, Kasputytė G, Krikštolaitis R, Ereminienė E, Galnaitienė G, Mizarienė V, Šakalytė G, Krilavičius T, Jurkevičius R. Association of uncertain significance genetic variants with myocardial mechanics and morphometrics in patients with nonischemic dilated cardiomyopathy. BMC Cardiovasc Disord 2024; 24:224. [PMID: 38664609 PMCID: PMC11044472 DOI: 10.1186/s12872-024-03888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Careful interpretation of the relation between phenotype changes of the heart and gene variants detected in dilated cardiomyopathy (DCM) is important for patient care and monitoring. OBJECTIVE We sought to assess the association between cardiac-related genes and whole-heart myocardial mechanics or morphometrics in nonischemic dilated cardiomyopathy (NIDCM). METHODS It was a prospective study consisting of patients with NIDCM. All patients were referred for genetic testing and a genetic analysis was performed using Illumina NextSeq 550 and a commercial gene capture panel of 233 genes (Systems Genomics, Cardiac-GeneSGKit®). It was analyzed whether there are significant differences in clinical, two-dimensional (2D) echocardiographic, and magnetic resonance imaging (MRI) parameters between patients with the genes variants and those without. 2D echocardiography and MRI were used to analyze myocardial mechanics and morphometrics. RESULTS The study group consisted of 95 patients with NIDCM and the average age was 49.7 ± 10.5. All echocardiographic and MRI parameters of myocardial mechanics (left ventricular ejection fraction 28.4 ± 8.7 and 30.7 ± 11.2, respectively) were reduced and all values of cardiac chambers were increased (left ventricular end-diastolic diameter 64.5 ± 5.9 mm and 69.5 ± 10.7 mm, respectively) in this group. It was noticed that most cases of whole-heart myocardial mechanics and morphometrics differences between patients with and without gene variants were in the genes GATAD1, LOX, RASA1, KRAS, and KRIT1. These genes have not been previously linked to DCM. It has emerged that KRAS and KRIT1 genes were associated with worse whole-heart mechanics and enlargement of all heart chambers. GATAD1, LOX, and RASA1 genes variants showed an association with better cardiac function and morphometrics parameters. It might be that these variants alone do not influence disease development enough to be selective in human evolution. CONCLUSIONS Combined variants in previously unreported genes related to DCM might play a significant role in affecting clinical, morphometrics, or myocardial mechanics parameters.
Collapse
Affiliation(s)
- Karolina Mėlinytė-Ankudavičė
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania.
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania.
| | - Marius Šukys
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, Kaunas, LT-50161, Lithuania
| | - Gabrielė Kasputytė
- Faculty of Informatics, Vytautas Magnus University, Kaunas, LT-44248, Lithuania
| | | | - Eglė Ereminienė
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| | - Grytė Galnaitienė
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania
| | - Vaida Mizarienė
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania
| | - Gintarė Šakalytė
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| | - Tomas Krilavičius
- Faculty of Informatics, Vytautas Magnus University, Kaunas, LT-44248, Lithuania
| | - Renaldas Jurkevičius
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, LT-44307, Lithuania
| |
Collapse
|
3
|
Tan K, Foo R, Loh M. Cardiomyopathy in Asian Cohorts: Genetic and Epigenetic Insights. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:496-506. [PMID: 37589150 DOI: 10.1161/circgen.123.004079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Previous studies on cardiomyopathies have been particularly valuable for clarifying pathological mechanisms in heart failure, an etiologically heterogeneous disease. In this review, we specifically focus on cardiomyopathies in Asia, where heart failure is particularly pertinent. There has been an increase in prevalence of cardiomyopathies in Asia, in sharp contrast with the decline observed in Western countries. Indeed, important disparities in cardiomyopathy incidence, clinical characteristics, and prognosis have been reported in Asian versus White cohorts. These have been accompanied by emerging descriptions of a distinct rare and common genetic basis for disease among Asian cardiomyopathy patients marked by an increased burden of variants with uncertain significance, reclassification of variants deemed pathogenic based on evidence from predominantly White cohorts, and the discovery of Asian-specific cardiomyopathy-associated loci with underappreciated pathogenicity under conventional classification criteria. Findings from epigenetic studies of heart failure, particularly DNA methylation studies, have complemented genetic findings in accounting for the phenotypic variability in cardiomyopathy. Though extremely limited, findings from Asian ancestry-focused DNA methylation studies of cardiomyopathy have shown potential to contribute to general understanding of cardiomyopathy pathophysiology by proposing disease and cause-relevant pathophysiological mechanisms. We discuss the value of multiomics study designs incorporating genetic, methylation, and transcriptomic information for future DNA methylation studies in Asian cardiomyopathy cohorts to yield Asian ancestry-specific insights that will improve risk stratification in the Asian population.
Collapse
Affiliation(s)
- Konstanze Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore (R.F.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore (R.F.)
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
- Genome Institute of Singapore, Singapore (GIS), Agency for Science, Technology and Research (A*STAR) (M.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (M.L.)
- National Skin Centre, Singapore (M.L.)
| |
Collapse
|
4
|
Niskanen JE, Ohlsson Å, Ljungvall I, Drögemüller M, Ernst RF, Dooijes D, van Deutekom HWM, van Tintelen JP, Snijders Blok CJB, van Vugt M, van Setten J, Asselbergs FW, Petrič AD, Salonen M, Hundi S, Hörtenhuber M, Kere J, Pyle WG, Donner J, Postma AV, Leeb T, Andersson G, Hytönen MK, Häggström J, Wiberg M, Friederich J, Eberhard J, Harakalova M, van Steenbeek FG, Wess G, Lohi H. Identification of novel genetic risk factors of dilated cardiomyopathy: from canine to human. Genome Med 2023; 15:73. [PMID: 37723491 PMCID: PMC10506233 DOI: 10.1186/s13073-023-01221-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/17/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a life-threatening heart disease and a common cause of heart failure due to systolic dysfunction and subsequent left or biventricular dilatation. A significant number of cases have a genetic etiology; however, as a complex disease, the exact genetic risk factors are largely unknown, and many patients remain without a molecular diagnosis. METHODS We performed GWAS followed by whole-genome, transcriptome, and immunohistochemical analyses in a spontaneously occurring canine model of DCM. Canine gene discovery was followed up in three human DCM cohorts. RESULTS Our results revealed two independent additive loci associated with the typical DCM phenotype comprising left ventricular systolic dysfunction and dilatation. We highlight two novel candidate genes, RNF207 and PRKAA2, known for their involvement in cardiac action potentials, energy homeostasis, and morphology. We further illustrate the distinct genetic etiologies underlying the typical DCM phenotype and ventricular premature contractions. Finally, we followed up on the canine discoveries in human DCM patients and discovered candidate variants in our two novel genes. CONCLUSIONS Collectively, our study yields insight into the molecular pathophysiology of DCM and provides a large animal model for preclinical studies.
Collapse
Affiliation(s)
- Julia E Niskanen
- Department of Medical and Clinical Genetics, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin katu 2, 00790, Helsinki, Finland
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland
| | - Åsa Ohlsson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ingrid Ljungvall
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Michaela Drögemüller
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, 3001, Switzerland
| | - Robert F Ernst
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hanneke W M van Deutekom
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - J Peter van Tintelen
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christian J B Snijders Blok
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marion van Vugt
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Jessica van Setten
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | | | - Milla Salonen
- Department of Medical and Clinical Genetics, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin katu 2, 00790, Helsinki, Finland
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland
| | - Sruthi Hundi
- Department of Medical and Clinical Genetics, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin katu 2, 00790, Helsinki, Finland
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland
| | - Matthias Hörtenhuber
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Juha Kere
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Research Programs Unit, Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - W Glen Pyle
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Dalhousie Medicine, Saint John, NB, Canada
| | - Jonas Donner
- Wisdom Panel Research Team, Wisdom Panel, Kinship, Helsinki, Finland
| | - Alex V Postma
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, 3001, Switzerland
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Marjo K Hytönen
- Department of Medical and Clinical Genetics, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin katu 2, 00790, Helsinki, Finland
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland
| | - Jens Häggström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Maria Wiberg
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Jana Friederich
- LMU Small Animal Clinic, Ludwig Maximilians University of Munich, Munich, Germany
| | - Jenny Eberhard
- LMU Small Animal Clinic, Ludwig Maximilians University of Munich, Munich, Germany
| | - Magdalena Harakalova
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Frank G van Steenbeek
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, Utrecht, 3584 CM, The Netherlands
| | - Gerhard Wess
- LMU Small Animal Clinic, Ludwig Maximilians University of Munich, Munich, Germany
| | - Hannes Lohi
- Department of Medical and Clinical Genetics, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland.
- Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin katu 2, 00790, Helsinki, Finland.
- Folkhälsan Research Center, Haartmaninkatu 8, P.O.Box 63, 00290, Helsinki, Finland.
| |
Collapse
|
5
|
Gui H, Tang WHW, Francke S, Li J, She R, Bazeley P, Pereira NL, Adams K, Luzum JA, Connolly TM, Hernandez AF, McNaughton CD, Williams LK, Lanfear DE. Common Variants on FGD5 Increase Hazard of Mortality or Rehospitalization in Patients With Heart Failure From the ASCEND-HF Trial. Circ Heart Fail 2023; 16:e010438. [PMID: 37725680 PMCID: PMC10597552 DOI: 10.1161/circheartfailure.122.010438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/13/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Heart failure remains a global health burden, and patients hospitalized are particularly at risk, but genetic associates for subsequent death or rehospitalization are still lacking. METHODS The genetic substudy of the ASCEND-HF trial (Acute Study of Clinical Effectiveness of Nesiritide in Decompensated Heart Failure) was used to perform genome-wide association study and transethnic meta-analysis. The overall trial included the patients of self-reported European ancestry (n=2173) and African ancestry (n=507). The end point was death or heart failure rehospitalization within 180 days. Cox models adjusted for 11 a priori predictors of rehospitalization and 5 genetic principal components were used to test the association between single-nucleotide polymorphisms and outcome. Summary statistics from the 2 populations were combined via meta-analysis with the significance threshold considered P<5×10-8. RESULTS Common variants (rs2342882 and rs35850039 in complete linkage disequilibrium) located in FGD5 were significantly associated with the primary outcome in both ancestry groups (European Americans: hazard ratio [HR], 1.38; P=2.42×10-6; African ancestry: HR, 1.51; P=4.43×10-3; HR in meta-analysis, 1.41; P=4.25×10-8). FGD5 encodes a regulator of VEGF (vascular endothelial growth factor)-mediated angiogenesis, and in silico investigation revealed several previous genome-wide association study hits in this gene, among which rs748431 was associated with our outcome (HR, 1.20; meta P<0.01). Sensitivity analysis proved FGD5 common variants survival association did not appear to operate via coronary artery disease or nesiritide treatment (P>0.05); and the signal was still significant when changing the censoring time from 180 to 30 days (HR, 1.39; P=1.59×10-5). CONCLUSIONS In this multiethnic genome-wide association study of ASCEND-HF, single-nucleotide polymorphisms in FGD5 were associated with increased risk of death or rehospitalization. Additional investigation is required to examine biological mechanisms and whether FGD5 could be a therapeutic target. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT00475852.
Collapse
Affiliation(s)
- Hongsheng Gui
- Center for Individualized and Genomics Medicine Research (H.G., J.A.L., L.K.W., D.E.L.), Henry Ford Hospital, Detroit, MI
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic, OH (W.H.W.T., P.B.)
| | | | - Jia Li
- Department of Public Health Science (J.L., R.S.), Henry Ford Hospital, Detroit, MI
| | - Ruicong She
- Department of Public Health Science (J.L., R.S.), Henry Ford Hospital, Detroit, MI
| | - Peter Bazeley
- Department of Cardiovascular Medicine, Cleveland Clinic, OH (W.H.W.T., P.B.)
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (N.L.P.)
| | - Kirkwood Adams
- Department of Medicine, University of North Carolina, Chapel Hill (K.A.)
| | - Jasmine A Luzum
- Center for Individualized and Genomics Medicine Research (H.G., J.A.L., L.K.W., D.E.L.), Henry Ford Hospital, Detroit, MI
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor (J.A.L.)
| | - Thomas M Connolly
- Lansdale, PA, previously Janssen Research & Development LLC, Spring House, PA (T.M.C.)
| | | | - Candace D McNaughton
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN (C.D.M.)
| | - L Keoki Williams
- Center for Individualized and Genomics Medicine Research (H.G., J.A.L., L.K.W., D.E.L.), Henry Ford Hospital, Detroit, MI
| | - David E Lanfear
- Center for Individualized and Genomics Medicine Research (H.G., J.A.L., L.K.W., D.E.L.), Henry Ford Hospital, Detroit, MI
- Heart and Vascular Institute (D.E.L.), Henry Ford Hospital, Detroit, MI
| |
Collapse
|
6
|
Perez-Bermejo JA, Judge LM, Jensen CL, Wu K, Watry HL, Truong A, Ho JJ, Carter M, Runyon WV, Kaake RM, Pulido EH, Mandegar MA, Swaney DL, So PL, Krogan NJ, Conklin BR. Functional analysis of a common BAG3 allele associated with protection from heart failure. NATURE CARDIOVASCULAR RESEARCH 2023; 2:615-628. [PMID: 39195919 DOI: 10.1038/s44161-023-00288-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 05/18/2023] [Indexed: 08/29/2024]
Abstract
Multiple genetic association studies have correlated a common allelic block linked to the BAG3 gene with a decreased incidence of heart failure, but the molecular mechanism remains elusive. In this study, we used induced pluripotent stem cells to test if the only coding variant in this allele block, BAG3C151R, alters protein and cellular function in human cardiomyocytes. Quantitative protein interaction analysis identified changes in BAG3C151R protein partners specific to cardiomyocytes. Knockdown of genes encoding for BAG3-interacting factors in cardiomyocytes followed by myofibrillar analysis revealed that BAG3C151R associates more strongly with proteins involved in the maintenance of myofibrillar integrity. Finally, we demonstrate that cardiomyocytes expressing the BAG3C151R variant have improved response to proteotoxic stress in a dose-dependent manner. This study suggests that BAG3C151R could be responsible for the cardioprotective effect of the haplotype block, by increasing cardiomyocyte protection from stress. Preferential binding partners of BAG3C151R may reveal potential targets for cardioprotective therapies.
Collapse
Affiliation(s)
| | - Luke M Judge
- Gladstone Institutes, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kenneth Wu
- Gladstone Institutes, San Francisco, CA, USA
| | | | | | - Jaclyn J Ho
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | | | - Robyn M Kaake
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Danielle L Swaney
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Po-Lin So
- Gladstone Institutes, San Francisco, CA, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Innovative Genomics Institute, Berkeley, CA, USA.
| |
Collapse
|
7
|
Lau C, Gul U, Liu B, Captur G, Hothi SS. Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy. Medicina (B Aires) 2023; 59:medicina59030439. [PMID: 36984439 PMCID: PMC10057087 DOI: 10.3390/medicina59030439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of non-ischaemic heart failure, conferring high morbidity and mortality, including sudden cardiac death due to systolic dysfunction or arrhythmic sudden death. Within the DCM cohort exists a group of patients with familial disease. In this article we review the pathophysiology and cardiac imaging findings of familial DCM, with specific attention to known disease subtypes. The role of advanced cardiac imaging cardiovascular magnetic resonance is still accumulating, and there remains much to be elucidated. We discuss its potential clinical roles as currently known, with respect to diagnostic utility and risk stratification. Advances in such risk stratification may help target pharmacological and device therapies to those at highest risk.
Collapse
Affiliation(s)
- Clement Lau
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
| | - Uzma Gul
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
| | - Boyang Liu
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Gabriella Captur
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London WC1E 6BT, UK
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Centre for Inherited Heart Muscle Conditions, Cardiology Department, The Royal Free Hospital, London NW3 2QG, UK
| | - Sandeep S. Hothi
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Correspondence:
| |
Collapse
|
8
|
Spracklen TF, Keavney B, Laing N, Ntusi N, Shaboodien G. Modern genomic techniques in the identification of genetic causes of cardiomyopathy. Heart 2022; 108:1843-1850. [PMID: 35140110 DOI: 10.1136/heartjnl-2021-320424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/18/2022] [Indexed: 11/04/2022] Open
Abstract
Over the past three decades numerous disease-causing genes have been linked to the pathogenesis of heritable cardiomyopathies, but many causal genes are yet to be identified. Next-generation sequencing (NGS) platforms have revolutionised clinical testing capacity in familial cardiomyopathy. In this review, we summarise how NGS technologies have advanced our understanding of genetic non-syndromic cardiomyopathy over the last decade. First, 26 putative new disease-causing genes have been identified to date, mostly from whole-exome sequencing, and some of which (FLNC, MTO1, HCN4) have had a considerable clinical impact and are now included in routine diagnostic gene panels. Second, we consider challenges in variant interpretation and the importance of large-scale NGS population control cohorts for this purpose. Third, an emerging role of common variation in some forms of genetic cardiomyopathy is being elucidated through recent studies which have illustrated an additive effect of numerous polymorphic loci on cardiac parameters; this may explain phenotypic variability and low rates of genetic diagnosis from sequencing studies. Finally, we discuss the clinical utility of genetic testing in cardiomyopathy in Western settings, where NGS panel testing of core disease genes is currently recommended with possible implications for patient management. Given the findings of recent studies, whole-exome or whole-genome sequencing should be considered in patients of non-European ancestry with clearly familial disease, or severe paediatric disease, when no result is obtained on panel sequencing. The clinical utility of polygenic risk assessment needs to be investigated further in patients with unexplained dilated cardiomyopathy and hypertrophic cardiomyopathy in whom a pathogenic variant is not identified.
Collapse
Affiliation(s)
- Timothy F Spracklen
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Bernard Keavney
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | - Nakita Laing
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Ntobeko Ntusi
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Universities Body Imaging Centre, Cape Town, South Africa
| | - Gasnat Shaboodien
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
| |
Collapse
|
9
|
Nazarenko MS, Sleptcov AA, Puzyrev VP. “Mendelian Code” in the Genetic Structure of Common Multifactorial Diseases. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422100052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
10
|
Shah RA, Asatryan B, Sharaf Dabbagh G, Aung N, Khanji MY, Lopes LR, van Duijvenboden S, Holmes A, Muser D, Landstrom AP, Lee AM, Arora P, Semsarian C, Somers VK, Owens AT, Munroe PB, Petersen SE, Chahal CAA. Frequency, Penetrance, and Variable Expressivity of Dilated Cardiomyopathy-Associated Putative Pathogenic Gene Variants in UK Biobank Participants. Circulation 2022; 146:110-124. [PMID: 35708014 PMCID: PMC9375305 DOI: 10.1161/circulationaha.121.058143] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND There is a paucity of data regarding the phenotype of dilated cardiomyopathy (DCM) gene variants in the general population. We aimed to determine the frequency and penetrance of DCM-associated putative pathogenic gene variants in a general adult population, with a focus on the expression of clinical and subclinical phenotype, including structural, functional, and arrhythmic disease features. METHODS UK Biobank participants who had undergone whole exome sequencing, ECG, and cardiovascular magnetic resonance imaging were selected for study. Three variant-calling strategies (1 primary and 2 secondary) were used to identify participants with putative pathogenic variants in 44 DCM genes. The observed phenotype was graded DCM (clinical or cardiovascular magnetic resonance diagnosis); early DCM features, including arrhythmia or conduction disease, isolated ventricular dilation, and hypokinetic nondilated cardiomyopathy; or phenotype-negative. RESULTS Among 18 665 individuals included in the study, 1463 (7.8%) possessed ≥1 putative pathogenic variant in 44 DCM genes by the main variant calling strategy. A clinical diagnosis of DCM was present in 0.34% and early DCM features in 5.7% of individuals with putative pathogenic variants. ECG and cardiovascular magnetic resonance analysis revealed evidence of subclinical DCM in an additional 1.6% and early DCM features in an additional 15.9% of individuals with putative pathogenic variants. Arrhythmias or conduction disease (15.2%) were the most common early DCM features, followed by hypokinetic nondilated cardiomyopathy (4%). The combined clinical/subclinical penetrance was ≤30% with all 3 variant filtering strategies. Clinical DCM was slightly more prevalent among participants with putative pathogenic variants in definitive/strong evidence genes as compared with those with variants in moderate/limited evidence genes. CONCLUSIONS In the UK Biobank, ≈1 of 6 of adults with putative pathogenic variants in DCM genes exhibited early DCM features potentially associated with DCM genotype, most commonly manifesting with arrhythmias in the absence of substantial ventricular dilation or dysfunction.
Collapse
Affiliation(s)
- Ravi A Shah
- Imperial College Healthcare NHS Trust, London, United Kingdom (R.A.S.)
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Switzerland (B.A.)
| | - Ghaith Sharaf Dabbagh
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA (G.S.D., C.A.A.C.).,University of Michigan, Division of Cardiovascular Medicine, Ann Arbor (G.S.D.)
| | - Nay Aung
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (N.A., M.Y.K., L.R.L., A.M.L., S.E.P., C.A.A.C.).,NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Mohammed Y Khanji
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (N.A., M.Y.K., L.R.L., A.M.L., S.E.P., C.A.A.C.).,NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (L.R.L.)
| | - Stefan van Duijvenboden
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | | | - Daniele Muser
- Cardiac Electrophysiology, Cardiovascular Division, Hospital of the University of Pennsylvania, Philadelphia (D.M., C.A.A.C.)
| | - Andrew P Landstrom
- Departments of Pediatrics, Division of Cardiology, and Cell Biology, Duke University School of Medicine, Durham, NC (A.P.L.)
| | - Aaron Mark Lee
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Pankaj Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham (P.A.)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute (C.S.), The University of Sydney, New South Wales, Australia.,Sydney Medical School Faculty of Medicine and Health (C.S.), The University of Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (C.S.)
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (V.K.S., C.A.A.C.)
| | - Anjali T Owens
- Center for Inherited Cardiovascular Disease, Cardiovascular Division, University of Pennsylvania Perelman School of Medicine, Philadelphia (A.T.O.)
| | - Patricia B Munroe
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Steffen E Petersen
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - C Anwar A Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA (G.S.D., C.A.A.C.).,Cardiac Electrophysiology, Cardiovascular Division, Hospital of the University of Pennsylvania, Philadelphia (D.M., C.A.A.C.).,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (V.K.S., C.A.A.C.)
| | | |
Collapse
|
11
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
12
|
McLendon JM, Zhang X, Matasic DS, Kumar M, Koval OM, Grumbach IM, Sadayappan S, London B, Boudreau RL. Knockout of Sorbin And SH3 Domain Containing 2 (Sorbs2) in Cardiomyocytes Leads to Dilated Cardiomyopathy in Mice. J Am Heart Assoc 2022; 11:e025687. [PMID: 35730644 PMCID: PMC9333371 DOI: 10.1161/jaha.122.025687] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Sorbin and SH3 domain containing 2 (Sorbs2) protein is a cytoskeletal adaptor with an emerging role in cardiac biology and disease; yet, its potential relevance to adult‐onset cardiomyopathies remains underexplored. Sorbs2 global knockout mice display lethal arrhythmogenic cardiomyopathy; however, the causative mechanisms remain unclear. Herein, we examine Sorbs2 dysregulation in heart failure, characterize novel Sorbs2 cardiomyocyte‐specific knockout mice (Sorbs2‐cKO), and explore associations between Sorbs2 genetic variations and human cardiovascular disease. Methods and Results Bioinformatic analyses show myocardial Sorbs2 mRNA is consistently upregulated in humans with adult‐onset cardiomyopathies and in heart failure models. We generated Sorbs2‐cKO mice and report that they develop progressive systolic dysfunction and enlarged cardiac chambers, and they die with congestive heart failure at about 1 year old. After 3 months, Sorbs2‐cKO mice begin to show atrial enlargement and P‐wave anomalies, without dysregulation of action potential–associated ion channel and gap junction protein expressions. After 6 months, Sorbs2‐cKO mice exhibit impaired contractility in dobutamine‐treated hearts and skinned myofibers, without dysregulation of contractile protein expressions. From our comprehensive survey of potential mechanisms, we found that within 4 months, Sorbs2‐cKO hearts have defective microtubule polymerization and compensatory upregulation of structural cytoskeletal and adapter proteins, suggesting that this early intracellular structural remodeling is responsible for contractile dysfunction. Finally, we identified genetic variants that associate with decreased Sorbs2 expression and human cardiac phenotypes, including conduction abnormalities, atrial enlargement, and dilated cardiomyopathy, consistent with Sorbs2‐cKO mice phenotypes. Conclusions Our studies show that Sorbs2 is essential for maintaining structural integrity in cardiomyocytes, likely through strengthening the interactions between microtubules and other cytoskeletal proteins at cross‐link sites.
Collapse
Affiliation(s)
- Jared M McLendon
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Xiaoming Zhang
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Daniel S Matasic
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Department of Molecular Physiology and Biophysics University of Iowa Carver College of Medicine Iowa City IA
| | - Mohit Kumar
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Olha M Koval
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Isabella M Grumbach
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Sakthivel Sadayappan
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Barry London
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Ryan L Boudreau
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| |
Collapse
|
13
|
Aung N, Vargas JD, Yang C, Fung K, Sanghvi MM, Piechnik SK, Neubauer S, Manichaikul A, Rotter JI, Taylor KD, Lima JAC, Bluemke DA, Kawut SM, Petersen SE, Munroe PB. Genome-wide association analysis reveals insights into the genetic architecture of right ventricular structure and function. Nat Genet 2022; 54:783-791. [PMID: 35697868 DOI: 10.1038/s41588-022-01083-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/22/2022] [Indexed: 01/03/2023]
Abstract
Right ventricular (RV) structure and function influence the morbidity and mortality from coronary artery disease (CAD), dilated cardiomyopathy (DCM), pulmonary hypertension and heart failure. Little is known about the genetic basis of RV measurements. Here we perform genome-wide association analyses of four clinically relevant RV phenotypes (RV end-diastolic volume, RV end-systolic volume, RV stroke volume, RV ejection fraction) from cardiovascular magnetic resonance images, using a state-of-the-art deep learning algorithm in 29,506 UK Biobank participants. We identify 25 unique loci associated with at least one RV phenotype at P < 2.27 ×10-8, 17 of which are validated in a combined meta-analysis (n = 41,830). Several candidate genes overlap with Mendelian cardiomyopathy genes and are involved in cardiac muscle contraction and cellular adhesion. The RV polygenic risk scores (PRSs) are associated with DCM and CAD. The findings substantially advance our understanding of the genetic underpinning of RV measurements.
Collapse
Affiliation(s)
- Nay Aung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Jose D Vargas
- Veterans Affairs Medical Center, Washington, DC, USA.,Georgetown University, Washington, DC, USA
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Kenneth Fung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Mihir M Sanghvi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Steffen E Petersen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK. .,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK. .,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK.
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK. .,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
14
|
Kucher AN, Sleptcov AA, Nazarenko MS. Genetic Landscape of Dilated Cardiomyopathy. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422030085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
Winter MJ, Ono Y, Ball JS, Walentinsson A, Michaelsson E, Tochwin A, Scholpp S, Tyler CR, Rees S, Hetheridge MJ, Bohlooly-Y M. A Combined Human in Silico and CRISPR/Cas9-Mediated in Vivo Zebrafish Based Approach to Provide Phenotypic Data for Supporting Early Target Validation. Front Pharmacol 2022; 13:827686. [PMID: 35548346 PMCID: PMC9082939 DOI: 10.3389/fphar.2022.827686] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/16/2022] [Indexed: 12/29/2022] Open
Abstract
The clinical heterogeneity of heart failure has challenged our understanding of the underlying genetic mechanisms of this disease. In this respect, large-scale patient DNA sequencing studies have become an invaluable strategy for identifying potential genetic contributing factors. The complex aetiology of heart failure, however, also means that in vivo models are vital to understand the links between genetic perturbations and functional impacts as part of the process for validating potential new drug targets. Traditional approaches (e.g., genetically-modified mice) are optimal for assessing small numbers of genes, but less practical when multiple genes are identified. The zebrafish, in contrast, offers great potential for higher throughput in vivo gene functional assessment to aid target prioritisation, by providing more confidence in target relevance and facilitating gene selection for definitive loss of function studies undertaken in mice. Here we used whole-exome sequencing and bioinformatics on human patient data to identify 3 genes (API5, HSPB7, and LMO2) suggestively associated with heart failure that were also predicted to play a broader role in disease aetiology. The role of these genes in cardiovascular system development and function was then further investigated using in vivo CRISPR/Cas9-mediated gene mutation analysis in zebrafish. We observed multiple impacts in F0 knockout zebrafish embryos (crispants) following effective somatic mutation, including changes in ventricle size, pericardial oedema, and chamber malformation. In the case of lmo2, there was also a significant impact on cardiovascular function as well as an expected reduction in erythropoiesis. The data generated from both the human in silico and zebrafish in vivo assessments undertaken supports further investigation of the potential roles of API5, HSPB7, and LMO2 in human cardiovascular disease. The data presented also supports the use of human in silico genetic variant analysis, in combination with zebrafish crispant phenotyping, as a powerful approach for assessing gene function as part of an integrated multi-level drug target validation strategy.
Collapse
Affiliation(s)
- Matthew J Winter
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Yosuke Ono
- Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Jonathan S Ball
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Anna Walentinsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Erik Michaelsson
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Tochwin
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Steffen Scholpp
- Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Charles R Tyler
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Steve Rees
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Malcolm J Hetheridge
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Mohammad Bohlooly-Y
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
16
|
Xiao L, Wu D, Sun Y, Hu D, Dai J, Chen Y, Wang D. Whole-exome sequencing reveals genetic risks of early-onset sporadic dilated cardiomyopathy in the Chinese Han population. SCIENCE CHINA. LIFE SCIENCES 2022; 65:770-780. [PMID: 34302607 DOI: 10.1007/s11427-020-1951-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/01/2021] [Indexed: 06/13/2023]
Abstract
To reveal genetic risks of early-onset sporadic dilated cardiomyopathy (DCM) patients in the Chinese Han population, we enlisted 363 DCM cases and 414 healthy controls. Whole-exome sequencing and phenotypic characterization were conducted. In total, we identified 26 loss-of-function (LOF) candidates and 66 pathogenic variants from 33 genes, most of which were novel. The deleterious variants can account for 25.07% (91/363) of all patients. Furthermore, rare missense variants in 21 genes were found to be significantly associated with DCM in burden tests. Other than rare variants, twelve common SNPs were significantly associated with an increased risk of DCM in allele-based genetic model association analysis. Of note, in the cumulative risk model, high-risk subjects had a 3.113-fold higher risk of developing DCM than low-risk subjects. Also, DCM in the high-risk group had a younger age of onset than that in the low-risk group. In terms of cardiac function, the mean left ventricular ejection fraction of patients with the deleterious variants was lower than those without (27.73%±10.02% vs. 30.61%±10.85%, P=0.026). To conclude, we mapped a comprehensive atlas of genetic risks in Chinese patients with DCM that might lead to new insights into the mechanisms and risk stratification for DCM.
Collapse
Affiliation(s)
- Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dongyang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiaqi Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanghui Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Yan Y, Long T, Su Q, Wang Y, Chen K, Yang T, Zhao G, Ma Q, Hu X, Liu C, Liao X, Min W, Li S, Zhang D, Yang Y, Pu WT, Dong Y, Wang DZ, Chen Y, Huang ZP. Cardiac ISL1-Interacting Protein, a Cardioprotective Factor, Inhibits the Transition From Cardiac Hypertrophy to Heart Failure. Front Cardiovasc Med 2022; 9:857049. [PMID: 35369338 PMCID: PMC8970336 DOI: 10.3389/fcvm.2022.857049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure is characterized by the inability of the heart to pump effectively and generate proper blood circulation to meet the body’s needs; it is a devastating condition that affects more than 100 million people globally. In spite of this, little is known about the mechanisms regulating the transition from cardiac hypertrophy to heart failure. Previously, we identified a cardiomyocyte-enriched gene, CIP, which regulates cardiac homeostasis under pathological stimulation. Here, we show that the cardiac transcriptional factor GATA4 binds the promotor of CIP gene and regulates its expression. We further determined that both CIP mRNA and protein decrease in diseased human hearts. In a mouse model, induced cardiac-specific overexpression of CIP after the establishment of cardiac hypertrophy protects the heart by inhibiting disease progression toward heart failure. Transcriptome analyses revealed that the IGF, mTORC2 and TGFβ signaling pathways mediate the inhibitory function of CIP on pathologic cardiac remodeling. Our study demonstrates GATA4 as an upstream regulator of CIP gene expression in cardiomyocytes, as well as the clinical significance of CIP expression in human heart disease. More importantly, our investigation suggests CIP is a key regulator of the transition from cardiac hypertrophy to heart failure. The ability of CIP to intervene in the onset of heart failure suggests a novel therapeutic avenue of investigation for the prevention of heart disease progression.
Collapse
Affiliation(s)
- Youchen Yan
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Long
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Qiao Su
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ken Chen
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Machine Intelligence and Advanced Computing, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guangyin Zhao
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Chen Liu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shujuan Li
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Dihua Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuedong Yang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Machine Intelligence and Advanced Computing, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Da-Zhi Wang,
| | - Yili Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Yili Chen,
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- Zhan-Peng Huang,
| |
Collapse
|
18
|
Lesurf R, Said A, Akinrinade O, Breckpot J, Delfosse K, Liu T, Yao R, Persad G, McKenna F, Noche RR, Oliveros W, Mattioli K, Shah S, Miron A, Yang Q, Meng G, Yue MCS, Sung WWL, Thiruvahindrapuram B, Lougheed J, Oechslin E, Mondal T, Bergin L, Smythe J, Jayappa S, Rao VJ, Shenthar J, Dhandapany PS, Semsarian C, Weintraub RG, Bagnall RD, Ingles J, Melé M, Maass PG, Ellis J, Scherer SW, Mital S. Whole genome sequencing delineates regulatory, copy number, and cryptic splice variants in early onset cardiomyopathy. NPJ Genom Med 2022; 7:18. [PMID: 35288587 PMCID: PMC8921194 DOI: 10.1038/s41525-022-00288-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/04/2022] [Indexed: 11/08/2022] Open
Abstract
Cardiomyopathy (CMP) is a heritable disorder. Over 50% of cases are gene-elusive on clinical gene panel testing. The contribution of variants in non-coding DNA elements that result in cryptic splicing and regulate gene expression has not been explored. We analyzed whole-genome sequencing (WGS) data in a discovery cohort of 209 pediatric CMP patients and 1953 independent replication genomes and exomes. We searched for protein-coding variants, and non-coding variants predicted to affect the function or expression of genes. Thirty-nine percent of cases harbored pathogenic coding variants in known CMP genes, and 5% harbored high-risk loss-of-function (LoF) variants in additional candidate CMP genes. Fifteen percent harbored high-risk regulatory variants in promoters and enhancers of CMP genes (odds ratio 2.25, p = 6.70 × 10-7 versus controls). Genes involved in α-dystroglycan glycosylation (FKTN, DTNA) and desmosomal signaling (DSC2, DSG2) were most highly enriched for regulatory variants (odds ratio 6.7-58.1). Functional effects were confirmed in patient myocardium and reporter assays in human cardiomyocytes, and in zebrafish CRISPR knockouts. We provide strong evidence for the genomic contribution of functionally active variants in new genes and in regulatory elements of known CMP genes to early onset CMP.
Collapse
Affiliation(s)
- Robert Lesurf
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Abdelrahman Said
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Oyediran Akinrinade
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- St. George's University School of Medicine, Grenada, Grenada
| | | | - Kathleen Delfosse
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ting Liu
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Roderick Yao
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gabrielle Persad
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Fintan McKenna
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ramil R Noche
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Zebrafish Genetics and Disease Models Core, The Hospital for Sick Children, Toronto, ON, Canada
| | - Winona Oliveros
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, Spain
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shreya Shah
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anastasia Miron
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Qian Yang
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Guoliang Meng
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Wilson W L Sung
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Jane Lougheed
- Division of Cardiology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Erwin Oechslin
- Peter Munk Cardiac Centre, Division of Cardiology, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Tapas Mondal
- Department of Pediatrics, Hamilton Health Sciences Centre, Hamilton, ON, Canada
| | - Lynn Bergin
- Division of Cardiology, London Health Sciences Centre, London, ON, Canada
| | - John Smythe
- Department of Pediatrics, Kingston General Hospital, Kingston, ON, Canada
| | - Shashank Jayappa
- Cardiovascular Biology and Disease Theme, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (inStem), Bangalore, India
| | - Vinay J Rao
- Cardiovascular Biology and Disease Theme, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (inStem), Bangalore, India
| | - Jayaprakash Shenthar
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, India
| | - Perundurai S Dhandapany
- Cardiovascular Biology and Disease Theme, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (inStem), Bangalore, India
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Robert G Weintraub
- Cardiology Department, Royal Children's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Richard D Bagnall
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Jodie Ingles
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
- Cardio Genomics Program at Centenary Institute, The University of Sydney, Sydney, Australia
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, Spain
| | - Philipp G Maass
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - James Ellis
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen W Scherer
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Seema Mital
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada.
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
19
|
Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol 2022; 19:151-167. [PMID: 34526680 DOI: 10.1038/s41569-021-00608-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2021] [Indexed: 01/06/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) was traditionally described as an autosomal dominant Mendelian disease but is now increasingly recognized as having a complex genetic aetiology. Although eight core genes encoding sarcomeric proteins account for >90% of the pathogenic variants in patients with HCM, variants in several additional genes (ACTN2, ALPK3, CSRP3, FHOD3, FLNC, JPH2, KLHL24, PLN and TRIM63), encoding non-sarcomeric proteins with diverse functions, have been shown to be disease-causing in a small number of patients. Genome-wide association studies (GWAS) have identified numerous loci in cardiomyopathy case-control studies and biobank investigations of left ventricular functional traits. Genes associated with Mendelian cardiomyopathy are enriched in the putative causal gene lists at these loci. Intriguingly, many loci are associated with both HCM and dilated cardiomyopathy but with opposite directions of effect on left ventricular traits, highlighting a genetic basis underlying the contrasting pathophysiological effects observed in each condition. This overlap extends to rare Mendelian variants with distinct variant classes in several genes associated with HCM and dilated cardiomyopathy. In this Review, we appraise the complex contribution of the non-sarcomeric, HCM-associated genes to cardiomyopathies across a range of variant classes (from common non-coding variants of individually low effect size to complete gene knockouts), which provides insights into the genetic basis of cardiomyopathies, causal genes at GWAS loci and the application of clinical genetic testing.
Collapse
|
20
|
Tedesco B, Cristofani R, Ferrari V, Cozzi M, Rusmini P, Casarotto E, Chierichetti M, Mina F, Galbiati M, Piccolella M, Crippa V, Poletti A. Insights on Human Small Heat Shock Proteins and Their Alterations in Diseases. Front Mol Biosci 2022; 9:842149. [PMID: 35281256 PMCID: PMC8913478 DOI: 10.3389/fmolb.2022.842149] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The family of the human small Heat Shock Proteins (HSPBs) consists of ten members of chaperones (HSPB1-HSPB10), characterized by a low molecular weight and capable of dimerization and oligomerization forming large homo- or hetero-complexes. All HSPBs possess a highly conserved centrally located α-crystallin domain and poorly conserved N- and C-terminal domains. The main feature of HSPBs is to exert cytoprotective functions by preserving proteostasis, assuring the structural maintenance of the cytoskeleton and acting in response to cellular stresses and apoptosis. HSPBs take part in cell homeostasis by acting as holdases, which is the ability to interact with a substrate preventing its aggregation. In addition, HSPBs cooperate in substrates refolding driven by other chaperones or, alternatively, promote substrate routing to degradation. Notably, while some HSPBs are ubiquitously expressed, others show peculiar tissue-specific expression. Cardiac muscle, skeletal muscle and neurons show high expression levels for a wide variety of HSPBs. Indeed, most of the mutations identified in HSPBs are associated to cardiomyopathies, myopathies, and motor neuropathies. Instead, mutations in HSPB4 and HSPB5, which are also expressed in lens, have been associated with cataract. Mutations of HSPBs family members encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or in the generation of truncated or elongated proteins. This review will provide an updated overview of disease-related mutations in HSPBs focusing on the structural and biochemical effects of mutations and their functional consequences.
Collapse
Affiliation(s)
- B. Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - R. Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - P. Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - E. Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F. Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - A. Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- *Correspondence: A. Poletti,
| |
Collapse
|
21
|
He X, Liu J, Gu F, Chen J, Lu YW, Ding J, Guo H, Nie M, Kataoka M, Lin Z, Hu X, Chen H, Liao X, Dong Y, Min W, Deng ZL, Pu WT, Huang ZP, Wang DZ. Cardiac CIP protein regulates dystrophic cardiomyopathy. Mol Ther 2022; 30:898-914. [PMID: 34400329 PMCID: PMC8822131 DOI: 10.1016/j.ymthe.2021.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/24/2021] [Accepted: 08/08/2021] [Indexed: 02/04/2023] Open
Abstract
Heart failure is a leading cause of fatality in Duchenne muscular dystrophy (DMD) patients. Previously, we discovered that cardiac and skeletal-muscle-enriched CIP proteins play important roles in cardiac function. Here, we report that CIP, a striated muscle-specific protein, participates in the regulation of dystrophic cardiomyopathy. Using a mouse model of human DMD, we found that deletion of CIP leads to dilated cardiomyopathy and heart failure in young, non-syndromic mdx mice. Conversely, transgenic overexpression of CIP reduces pathological dystrophic cardiomyopathy in old, syndromic mdx mice. Genome-wide transcriptome analyses reveal that molecular pathways involving fibrogenesis and oxidative stress are affected in CIP-mediated dystrophic cardiomyopathy. Mechanistically, we found that CIP interacts with dystrophin and calcineurin (CnA) to suppress the CnA-Nuclear Factor of Activated T cells (NFAT) pathway, which results in decreased expression of Nox4, a key component of the oxidative stress pathway. Overexpression of Nox4 accelerates the development of dystrophic cardiomyopathy in mdx mice. Our study indicates CIP is a modifier of dystrophic cardiomyopathy and a potential therapeutic target for this devastating disease.
Collapse
Affiliation(s)
- Xin He
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Fei Gu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jinghai Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jian Ding
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Haipeng Guo
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Biology, Nanjing Normal University, Nanjing, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhong-Liang Deng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
22
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
23
|
Sergi CM. Point of care with serial N-terminal pro-B-type natriuretic peptide (NT-proBNP) for heart failure in patients with acute decompensation. An invited commentary. Contemp Clin Trials Commun 2022; 26:100889. [PMID: 36684692 PMCID: PMC9846449 DOI: 10.1016/j.conctc.2022.100889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/06/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Point-of-care (POC) Testing (POCT) is a medical diagnostic approach carried out outside the clinical laboratory and close to the patient. The results are used for the immediate clinical decision to improve patient care. Accreditation Canada and Diagnostic Accreditation Programs regulatory standards and the College of the American Pathologists for POCT have implemented several guidelines and certifications. POC in heart failure (POC-HF) will demonstrate that it is helpful to develop a preliminary understanding of the value of serial N-terminal pro-B-type natriuretic peptide (NT-proBNP) measurements in the diagnostic and therapeutic process in patients hospitalized with acute decompensation of heart failure. In addition, POC-HF will hopefully answer questions on the workability of this approach in clinical routine and the usefulness and justification to perform larger-scale studies investigating this strategy.
Collapse
Affiliation(s)
- Consolato M. Sergi
- Anatomy Pathology Division, Children's Hospital of Eastern Ontario, University of Ottawa, ON, Canada,Departments of Pediatrics and Laboratory Medicine, University of Alberta, Edmonton, AB, Canada,Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada,Anatomy Pathology Division, Children's Hospital of Eastern Ontario, University of Ottawa, ON, Canada.
| |
Collapse
|
24
|
Ben-Haim Y, Behr ER. Genetics of sudden cardiac death. Curr Opin Cardiol 2022; 37:212-218. [DOI: 10.1097/hco.0000000000000946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Lin H, Koren SA, Cvetojevic G, Girardi P, Johnson GV. The role of BAG3 in health and disease: A "Magic BAG of Tricks". J Cell Biochem 2022; 123:4-21. [PMID: 33987872 PMCID: PMC8590707 DOI: 10.1002/jcb.29952] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/25/2021] [Indexed: 01/03/2023]
Abstract
The multi-domain structure of Bcl-2-associated athanogene 3 (BAG3) facilitates its interaction with many different proteins that participate in regulating a variety of biological pathways. After revisiting the BAG3 literature published over the past ten years with Citespace software, we classified the BAG3 research into several clusters, including cancer, cardiomyopathy, neurodegeneration, and viral propagation. We then highlighted recent key findings in each cluster. To gain greater insight into the roles of BAG3, we analyzed five different published mass spectrometry data sets of proteins that co-immunoprecipitate with BAG3. These data gave us insight into universal, as well as cell-type-specific BAG3 interactors in cancer cells, cardiomyocytes, and neurons. Finally, we mapped variable BAG3 SNPs and also mutation data from previous publications to further explore the link between the domains and function of BAG3. We believe this review will provide a better understanding of BAG3 and direct future studies towards understanding BAG3 function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Heng Lin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Shon A. Koren
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Gregor Cvetojevic
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Peter Girardi
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Gail V.W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| |
Collapse
|
26
|
Salzer‐Sheelo L, Fellner A, Orenstein N, Bazak L, Lev‐El Halabi N, Daue M, Smirin‐Yosef P, Van Hout CV, Fellig Y, Ruhrman‐Shahar N, Staples J, Magal N, Shuldiner AR, Mitchell BD, Nevo Y, Pollin TI, Gonzaga‐Jauregui C, Basel‐Salmon L. Biallelic Truncating Variants in the Muscular A‐Type Lamin‐Interacting Protein (
MLIP
) Gene Cause Myopathy with Hyper‐CKemia. Eur J Neurol 2021; 29:1174-1180. [DOI: 10.1111/ene.15218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Liat Salzer‐Sheelo
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Avi Fellner
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
- Department of Neurology Rabin Medical Center Beilinson Hospital Petah Tikva Israel
| | - Naama Orenstein
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
- Pediatric Genetics Clinic Schneider Children’s Medical Center of Israel Petah Tikva Israel
| | - Lily Bazak
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
| | - Noa Lev‐El Halabi
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
| | - Melanie Daue
- Division of Endocrinology Diabetes & Nutrition Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Pola Smirin‐Yosef
- Genomic Bioinformatics Laboratory Department of Molecular Biology Ariel University Israel
- Felsenstein Medical Research Center Rabin Medical Center Petah Tikva Israel
| | | | - Yakov Fellig
- Department of Pathology Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem Israel
| | - Noa Ruhrman‐Shahar
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
| | | | - Nurit Magal
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
| | | | - Braxton D. Mitchell
- Division of Endocrinology Diabetes & Nutrition Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Yoram Nevo
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
- Pediatric Neurology Unit Schneider Children’s Medical Center of Israel Petah Tikva Israel
| | - Toni I. Pollin
- Division of Endocrinology Diabetes & Nutrition Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Claudia Gonzaga‐Jauregui
- Regeneron Genetics Center Tarrytown NY USA
- International Laboratory for Human Genome Research Laboratorio Internacional de Investigación sobre el Genoma Humano Universidad Nacional Autónoma de México Juriquilla Querétaro Mexico
| | - Lina Basel‐Salmon
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Hospital Petah Tikva Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
- Felsenstein Medical Research Center Rabin Medical Center Petah Tikva Israel
| |
Collapse
|
27
|
Yang Y, Bartz TM, Brown MR, Guo X, Zilhao NR, Trompet S, Weiss S, Yao J, Brody JA, Defilippi CR, Hoogeveen RC, Lin HJ, Gudnason V, Ballantyne CM, Dorr M, Jukema JW, Petersmann A, Psaty BM, Rotter JI, Boerwinkle E, Fornage M, Jun G, Yu B. Identification of Functional Genetic Determinants of Cardiac Troponin T and I in a Multiethnic Population and Causal Associations With Atrial Fibrillation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003460. [PMID: 34732054 PMCID: PMC8692416 DOI: 10.1161/circgen.121.003460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Elevated cardiac troponin levels in blood are associated with increased risk of cardiovascular diseases and mortality. Cardiac troponin levels are heritable, but their genetic architecture remains elusive. METHODS We conducted a transethnic genome-wide association analysis on high-sensitivity cTnT (cardiac troponin T; hs-cTnT) and high-sensitivity cTnI (cardiac troponin I; hs-cTnI) levels in 24 617 and 14 336 participants free of coronary heart disease and heart failure from 6 population-based cohorts, followed by a series of bioinformatic analyses to decipher the genetic architecture of hs-cTnT and hs-cTnI. RESULTS We identified 4 genome-wide significant loci for hs-cTnT including a novel locus rs3737882 in PPFIA4 and 3 previously reported loci at NCOA2, TRAM1, and BCL2. One known locus at VCL was replicated for hs-cTnI. One copy of C allele for rs3737882 was associated with a 6% increase in hs-cTnT levels (minor allele frequency, 0.18; P=2.80×10-9). We observed pleiotropic loci located at BAG3 and ANO5. The proportions of variances explained by single-nucleotide polymorphisms were 10.15% and 7.74% for hs-cTnT and hs-cTnI, respectively. Single-nucleotide polymorphisms were colocalized with BCL2 expression in heart tissues and hs-cTnT and with ANO5 expression in artery, heart tissues, and whole blood and both troponins. Mendelian randomization analyses showed that genetically increased hs-cTnT and hs-cTnI levels were associated with higher odds of atrial fibrillation (odds ratio, 1.38 [95% CI, 1.25-1.54] for hs-cTnT and 1.21 [95% CI, 1.06-1.37] for hs-cTnI). CONCLUSIONS We identified a novel genetic locus associated with hs-cTnT in a multiethnic population and found that genetically regulated troponin levels were associated with atrial fibrillation.
Collapse
Affiliation(s)
- Yunju Yang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Michael R. Brown
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center and Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional Genomics; University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Ron C. Hoogeveen
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Henry J. Lin
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christie M. Ballantyne
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart Center, Houston, TX, USA
| | - Marcus Dorr
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B - Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, University Medicine Greifswald, Greifswald, Germany
| | - J. Wouter Jukema
- Department of Cardiology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden and the Netherlands Heart Institute, Utrecht, the Netherlands
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald and Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Oldenburg, Oldenburg, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, and Department of Health Services, University of Washington, Seattle, Washington, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Eric Boerwinkle
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Goo Jun
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
28
|
Parker LE, Landstrom AP. The clinical utility of pediatric cardiomyopathy genetic testing: From diagnosis to a precision medicine-based approach to care. PROGRESS IN PEDIATRIC CARDIOLOGY 2021; 62. [PMID: 34776723 DOI: 10.1016/j.ppedcard.2021.101413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Pediatric-onset cardiomyopathies are rare yet cause significant morbidity and mortality in affected children. Genetic testing has a major role in the clinical evaluation of pediatric-onset cardiomyopathies, and identification of a variant in an associated gene can be used to confirm the clinical diagnosis and exclude syndromic causes that may warrant different treatment strategies. Further, risk-predictive testing of first-degree relatives can assess who is at-risk of disease and requires continued clinical follow-up. Aim of Review In this review, we seek to describe the current role of genetic testing in the clinical diagnosis and management of patients and families with the five major cardiomyopathies. Further, we highlight the ongoing development of precision-based approaches to diagnosis, prognosis, and treatment. Key Scientific Concepts of Review Emerging application of genotype-phenotype correlations opens the door for genetics to guide a precision medicine-based approach to prognosis and potentially for therapies. Despite advances in our understanding of the genetic etiology of cardiomyopathy and increased accessibility of clinical genetic testing, not all pediatric cardiomyopathy patients have a clear genetic explanation for their disease. Expanded genomic studies are needed to understand the cause of disease in these patients, improve variant classification and genotype-driven prognostic predictions, and ultimately develop truly disease preventing treatment.
Collapse
Affiliation(s)
- Lauren E Parker
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
29
|
Lopes Abath Neto O, Medne L, Donkervoort S, Rodríguez-García ME, Bolduc V, Hu Y, Guadagnin E, Foley AR, Brandsema JF, Glanzman AM, Tennekoon GI, Santi M, Berger JH, Megeney LA, Komaki H, Inoue M, Cotrina-Vinagre FJ, Hernández-Lain A, Martin-Hernández E, Williams L, Borell S, Schorling D, Lin K, Kolokotronis K, Lichter-Konecki U, Kirschner J, Nishino I, Banwell B, Martínez-Azorín F, Burgon PG, Bönnemann CG. MLIP causes recessive myopathy with rhabdomyolysis, myalgia and baseline elevated serum creatine kinase. Brain 2021; 144:2722-2731. [PMID: 34581780 DOI: 10.1093/brain/awab275] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023] Open
Abstract
Striated muscle needs to maintain cellular homeostasis in adaptation to increases in physiological and metabolic demands. Failure to do so can result in rhabdomyolysis. The identification of novel genetic conditions associated with rhabdomyolysis helps to shed light on hitherto unrecognized homeostatic mechanisms. Here we report seven individuals in six families from different ethnic backgrounds with biallelic variants in MLIP, which encodes the muscular lamin A/C-interacting protein, MLIP. Patients presented with a consistent phenotype characterized by mild muscle weakness, exercise-induced muscle pain, variable susceptibility to episodes of rhabdomyolysis, and persistent basal elevated serum creatine kinase levels. The biallelic truncating variants were predicted to result in disruption of the nuclear localizing signal of MLIP. Additionally, reduced overall RNA expression levels of the predominant MLIP isoform were observed in patients' skeletal muscle. Collectively, our data increase the understanding of the genetic landscape of rhabdomyolysis to now include MLIP as a novel disease gene in humans and solidifies MLIP's role in normal and diseased skeletal muscle homeostasis.
Collapse
Affiliation(s)
- Osorio Lopes Abath Neto
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Pathology, Division of Neuropathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Livija Medne
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Maria Elena Rodríguez-García
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN), Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eleonora Guadagnin
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - John F Brandsema
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allan M Glanzman
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gihan I Tennekoon
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mariarita Santi
- Department of Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justin H Berger
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Michio Inoue
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Francisco Javier Cotrina-Vinagre
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN), Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | | | - Elena Martin-Hernández
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Pediátrica de Enfermedades Raras, Enfermedades Mitocondriales y Metabólicas Hereditarias, Hospital 12 de Octubre, Madrid, Spain
| | - Linford Williams
- Division of Medical Genetics, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Sabine Borell
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - David Schorling
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Kimberly Lin
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Uta Lichter-Konecki
- Division of Medical Genetics, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.,Department of Neuropediatrics, University Hospital Bonn, Faculty of Medicine, Bonn, Germany
| | - Ichizo Nishino
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Brenda Banwell
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Francisco Martínez-Azorín
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares (ERMN), Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Patrick G Burgon
- Department of Chemistry and Earth Science, College of Arts and Sciences, Qatar University, Qatar
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Moksnes MR, Røsjø H, Richmond A, Lyngbakken MN, Graham SE, Hansen AF, Wolford BN, Gagliano Taliun SA, LeFaive J, Rasheed H, Thomas LF, Zhou W, Aung N, Surakka I, Douville NJ, Campbell A, Porteous DJ, Petersen SE, Munroe PB, Welsh P, Sattar N, Smith GD, Fritsche LG, Nielsen JB, Åsvold BO, Hveem K, Hayward C, Willer CJ, Brumpton BM, Omland T. Genome-wide association study of cardiac troponin I in the general population. Hum Mol Genet 2021; 30:2027-2039. [PMID: 33961016 PMCID: PMC8522636 DOI: 10.1093/hmg/ddab124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
Circulating cardiac troponin proteins are associated with structural heart disease and predict incident cardiovascular disease in the general population. However, the genetic contribution to cardiac troponin I (cTnI) concentrations and its causal effect on cardiovascular phenotypes are unclear. We combine data from two large population-based studies, the Trøndelag Health Study and the Generation Scotland Scottish Family Health Study, and perform a genome-wide association study of high-sensitivity cTnI concentrations with 48 115 individuals. We further use two-sample Mendelian randomization to investigate the causal effects of circulating cTnI on acute myocardial infarction (AMI) and heart failure (HF). We identified 12 genetic loci (8 novel) associated with cTnI concentrations. Associated protein-altering variants highlighted putative functional genes: CAND2, HABP2, ANO5, APOH, FHOD3, TNFAIP2, KLKB1 and LMAN1. Phenome-wide association tests in 1688 phecodes and 83 continuous traits in UK Biobank showed associations between a genetic risk score for cTnI and cardiac arrhythmias, metabolic and anthropometric measures. Using two-sample Mendelian randomization, we confirmed the non-causal role of cTnI in AMI (5948 cases, 355 246 controls). We found indications for a causal role of cTnI in HF (47 309 cases and 930 014 controls), but this was not supported by secondary analyses using left ventricular mass as outcome (18 257 individuals). Our findings clarify the biology underlying the heritable contribution to circulating cTnI and support cTnI as a non-causal biomarker for AMI in the general population. Using genetically informed methods for causal inference helps inform the role and value of measuring cTnI in the general population.
Collapse
Affiliation(s)
- Marta R Moksnes
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Helge Røsjø
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Magnus N Lyngbakken
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Sarah E Graham
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ailin Falkmo Hansen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Brooke N Wolford
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah A Gagliano Taliun
- Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Montréal Heart Institute, Montréal, QC H1T 1C8, Canada
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonathon LeFaive
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Humaira Rasheed
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Laurent F Thomas
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, 7491 Trondheim. Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7491 Trondheim, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nay Aung
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Ida Surakka
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas J Douville
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archie Campbell
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David J Porteous
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Steffen E Petersen
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Patricia B Munroe
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonas B Nielsen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology Research, Statens Serum Institute, 2300 Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Bjørn Olav Åsvold
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Cristen J Willer
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ben M Brumpton
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Clinic of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Torbjørn Omland
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| |
Collapse
|
31
|
Abstract
Almost 25 years have passed since a mutation of a formin gene, DIAPH1, was identified as being responsible for a human inherited disorder: a form of sensorineural hearing loss. Since then, our knowledge of the links between formins and disease has deepened considerably. Mutations of DIAPH1 and six other formin genes (DAAM2, DIAPH2, DIAPH3, FMN2, INF2 and FHOD3) have been identified as the genetic cause of a variety of inherited human disorders, including intellectual disability, renal disease, peripheral neuropathy, thrombocytopenia, primary ovarian insufficiency, hearing loss and cardiomyopathy. In addition, alterations in formin genes have been associated with a variety of pathological conditions, including developmental defects affecting the heart, nervous system and kidney, aging-related diseases, and cancer. This review summarizes the most recent discoveries about the involvement of formin alterations in monogenic disorders and other human pathological conditions, especially cancer, with which they have been associated. In vitro results and experiments in modified animal models are discussed. Finally, we outline the directions for future research in this field.
Collapse
Affiliation(s)
| | - Miguel A. Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
32
|
Abstract
Cardiomyopathy affects approximately 1 in 500 adults and is the leading cause of death. Familial cases are common, and mutations in many genes are involved in cardiomyopathy, especially those in genes encoding cytoskeletal, sarcomere, and nuclear envelope proteins. Filamin C is an actin-binding protein encoded by filamin C (FLNC) gene and participates in sarcomere stability maintenance. FLNC was first demonstrated to be a causal gene of myofibrillar myopathy; recently, it has been found that FLNC mutation plays a critical role in the pathogenesis of cardiomyopathy. In this review, we summarized the physiological roles of filamin C in cardiomyocytes and the genetic evidence for links between FLNC mutations and cardiomyopathies. Truncated FLNC is enriched in dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy. Non-truncated FLNC is enriched in hypertrophic cardiomyopathy and restrictive cardiomyopathy. Two major pathomechanisms in FLNC-related cardiomyopathy have been described: protein aggregation resulting from non-truncating mutations and haploinsufficiency triggered by filamin C truncation. Therefore, it is important to understand the cellular biology and molecular regulation of FLNC to design new therapies to treat patients with FLNC-related cardiomyopathy.
Collapse
|
33
|
Lopes LR, Garcia-Hernández S, Lorenzini M, Futema M, Chumakova O, Zateyshchikov D, Isidoro-Garcia M, Villacorta E, Escobar-Lopez L, Garcia-Pavia P, Bilbao R, Dobarro D, Sandin-Fuentes M, Catalli C, Gener Querol B, Mezcua A, Garcia Pinilla J, Bloch Rasmussen T, Ferreira-Aguar A, Revilla-Martí P, Basurte Elorz MT, Bautista Paves A, Ramon Gimeno J, Figueroa AV, Franco-Gutierrez R, Fuentes-Cañamero ME, Martinez Moreno M, Ortiz-Genga M, Piqueras-Flores J, Analia Ramos K, Rudzitis A, Ruiz-Guerrero L, Stein R, Triguero-Bocharán M, de la Higuera L, Ochoa JP, Abu-Bonsrah D, Kwok CYT, Smith JB, Porrello ER, Akhtar MM, Jager J, Ashworth M, Syrris P, Elliott DA, Monserrat L, Elliott PM. Alpha-protein kinase 3 (ALPK3) truncating variants are a cause of autosomal dominant hypertrophic cardiomyopathy. Eur Heart J 2021; 42:3063-3073. [PMID: 34263907 PMCID: PMC8380059 DOI: 10.1093/eurheartj/ehab424] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/17/2021] [Accepted: 06/18/2021] [Indexed: 01/07/2023] Open
Abstract
AIMS The aim of this study was to determine the frequency of heterozygous truncating ALPK3 variants (ALPK3tv) in patients with hypertrophic cardiomyopathy (HCM) and confirm their pathogenicity using burden testing in independent cohorts and family co-segregation studies. METHODS AND RESULTS In a discovery cohort of 770 index patients with HCM, 12 (1.56%) were heterozygous for ALPK3tv [odds ratio(OR) 16.11, 95% confidence interval (CI) 7.94-30.02, P = 8.05e-11] compared to the Genome Aggregation Database (gnomAD) population. In a validation cohort of 2047 HCM probands, 32 (1.56%) carried heterozygous ALPK3tv (OR 16.17, 95% CI 10.31-24.87, P < 2.2e-16, compared to gnomAD). Combined logarithm of odds score in seven families with ALPK3tv was 2.99. In comparison with a cohort of genotyped patients with HCM (n = 1679) with and without pathogenic sarcomere gene variants (SP+ and SP-), ALPK3tv carriers had a higher prevalence of apical/concentric patterns of hypertrophy (60%, P < 0.001) and of a short PR interval (10%, P = 0.009). Age at diagnosis and maximum left ventricular wall thickness were similar to SP- and left ventricular systolic impairment (6%) and non-sustained ventricular tachycardia (31%) at baseline similar to SP+. After 5.3 ± 5.7 years, 4 (9%) patients with ALPK3tv died of heart failure or had cardiac transplantation (log-rank P = 0.012 vs. SP- and P = 0.425 vs. SP+). Imaging and histopathology showed extensive myocardial fibrosis and myocyte vacuolation. CONCLUSIONS Heterozygous ALPK3tv are pathogenic and segregate with a characteristic HCM phenotype.
Collapse
Affiliation(s)
- Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK.,Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| | - Soledad Garcia-Hernández
- Health in Code S.L., Cardiology and Scientific Department, As Xubias, s/n Edificio O Fortín, 15006 A Coruña, Spain
| | - Massimiliano Lorenzini
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK.,Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| | - Marta Futema
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK
| | - Olga Chumakova
- Federal Scientific Clinical Centre of Federal Medical and Biological Agency, 30, Volokolamskoe Shosse, Moscow, Russia.,Department of Cardiology, City Clinical Hospital, #17, Volynska st., 7, Moscow, Russia
| | - Dmitry Zateyshchikov
- Federal Scientific Clinical Centre of Federal Medical and Biological Agency, Genetic Laboratory, Moscow, Russia
| | - Maria Isidoro-Garcia
- Inherited Cardiac Disease Unit (CSUR), Biochemistry Department, Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Medicine Department, Facultad de Medicina, Universidad de Salamanca, Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Paseo de San Vicente, 58-182, 37007 Salamanca, Madrid, Spain
| | - Eduardo Villacorta
- Inherited Cardiac Disease Unit (CSUR), Cardiology Department, Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Medicine Department, Facultad de Medicina, Universidad de Salamanca, Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Paseo de San Vicente, 58-182, 37007 Salamanca and Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029, Madrid, Spain
| | - Luis Escobar-Lopez
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, CIBERCV, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 and Calle Joaquín Rodrigo, 1, 28222 Majadahonda, Madrid, Spain.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-GUARDHEART)
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, CIBERCV, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 and Calle Joaquín Rodrigo, 1, 28222 Majadahonda, Madrid, Spain.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-GUARDHEART).,Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, Carretera Pozuelo a Majadahonda, Km 1.800, 28223 Madrid, Spain
| | - Raquel Bilbao
- Heart Failure and Pulmonary Hypertension Unit, Hospital Alvaro Cunqueiro, Complexo Hospitalario Universitario de Vigo, Estrada de Clara Campoamor, 341, 36213 Vigo, Pontevedra, Spain
| | - David Dobarro
- Heart Failure and Pulmonary Hypertension Unit, Hospital Alvaro Cunqueiro, Complexo Hospitalario Universitario de Vigo, Estrada de Clara Campoamor, 341, 36213 Vigo, Pontevedra, Spain
| | - Maria Sandin-Fuentes
- Hospital Clínico Universitario de Valladolid, Cardiology, Av. Ramón y Cajal, 3, 47003 Valladolid, Spain
| | - Claudio Catalli
- Osakidetza Basque Health Service, Cruces University Hospital, Department of Genetics, Biocruces Bizkaia Health Research Institute, Cruces Plaza, 48903 Barakaldo, Bizkaia, Spain
| | - Blanca Gener Querol
- Osakidetza Basque Health Service, Cruces University Hospital, Department of Genetics, Biocruces Bizkaia Health Research Institute, Cruces Plaza, 48903 Barakaldo, Bizkaia, Spain
| | - Ainhoa Mezcua
- Heart Failure and Familial Heart Diseases Unit, Cardiology Service, Hospital Universitario Virgen de la Victoria, IBIMA, Campus de Teatinos, S/N, 29010 Málaga, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 Madrid, Spain
| | - Jose Garcia Pinilla
- Heart Failure and Familial Heart Diseases Unit, Cardiology Service, Hospital Universitario Virgen de la Victoria, IBIMA, Campus de Teatinos, S/N, 29010 Málaga, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 Madrid, Spain
| | - Torsten Bloch Rasmussen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99 DK-8200 Aarhus, Denmark
| | - Ana Ferreira-Aguar
- Inherited Cardiac Diseases Unit, Cardiology Department, Hospital Clínico Universitario Lozano Blesa, Avda, Calle de San Juan Bosco, 15, 50009 Zaragoza, Spain
| | - Pablo Revilla-Martí
- Inherited Cardiac Diseases Unit, Cardiology Department, Hospital Clínico Universitario Lozano Blesa, Avda, Calle de San Juan Bosco, 15, 50009 Zaragoza, Spain
| | | | - Alicia Bautista Paves
- Hospital Universitario San Cecilio Granada, Av. del Conocimiento, s/n, 18016 Granada, Cardiology
| | - Juan Ramon Gimeno
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-GUARDHEART).,Hospital Clínico Universitario Virgen de la Arrixaca, Inherited Cardiac Diseases Unit, Department of Cardiology, Ctra. Madrid-Cartagena, s/n, 30120 El Palmar, Murcia, Spain
| | | | - Raul Franco-Gutierrez
- Cardiology Department, Hospital Universitario Lucus Augusti, Lugo Biodiscovery HULA-USC Research Group, Institute for Health Research of Santiago de Compostela IDIS, s/n A, Travesía da Choupana, 15706 Santiago de Compostela, A Coruña
| | | | | | - Martin Ortiz-Genga
- Health in Code S.L., Scientific Department, As Xubias, s/n Edificio O Fortín, 15006 A Coruña, Spain
| | - Jesus Piqueras-Flores
- Cardiology Department, Inherited Cardiovascular Diseases Unit, Hospital General Universitario de Ciudad Real, Calle Obispo Rafael Torija, s/n, 13005 Ciudad Real, Spain
| | | | - Ainars Rudzitis
- Pauls Stradins Clinical University Hospital, Pilsoņu iela 13, Zemgales priekšpilsēta, Rīga, LV-1002, Latvia
| | - Luis Ruiz-Guerrero
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Av. de Valdecilla, 25, 39008 Santander, Spain
| | - Ricardo Stein
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Av. Paulo Gama, 110 Secretaria de Comunicação Social - 8º andar - Reitoria - Farroupilha, Porto Alegre - RS 90040-060, Brazil
| | - Mayte Triguero-Bocharán
- Cardiology Department, Inherited Cardiovascular Diseases Unit, Hospital General Universitario de Ciudad Real, Calle Obispo Rafael Torija, s/n, 13005 Ciudad Real, Spain
| | - Luis de la Higuera
- Health in Code S.L., Scientific Department, As Xubias, s/n Edificio O Fortín, 15006 A Coruña, Spain
| | - Juan Pablo Ochoa
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, CIBERCV, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 and Calle Joaquín Rodrigo, 1, 28222 Majadahonda, Madrid, Spain.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-GUARDHEART)
| | - Dad Abu-Bonsrah
- Murdoch Research Childrens Research Institute, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Cecilia Y T Kwok
- Murdoch Research Childrens Research Institute, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Jacob B Smith
- Murdoch Research Childrens Research Institute, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Enzo R Porrello
- Murdoch Research Childrens Research Institute, Royal Melbourne Hospital, Parkville, VIC 3052, Australia.,Dept. of Physiology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Mohammed M Akhtar
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK.,Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| | - Joanna Jager
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK
| | - Michael Ashworth
- Department of Histopathology, Great Ormond St Hospital for Children, London WC1N 3NN, UK
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK
| | - David A Elliott
- Murdoch Research Childrens Research Institute, Royal Melbourne Hospital, Parkville, VIC 3052, Australia.,Dept. of Physiology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Lorenzo Monserrat
- Health in Code S.L., Scientific Department, As Xubias, s/n Edificio O Fortín, 15006 A Coruña, Spain
| | - Perry M Elliott
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 62 Huntley St, London WC1E 6DD, UK.,Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| |
Collapse
|
34
|
Kirk JA, Cheung JY, Feldman AM. Therapeutic targeting of BAG3: considering its complexity in cancer and heart disease. J Clin Invest 2021; 131:e149415. [PMID: 34396980 DOI: 10.1172/jci149415] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bcl2-associated athanogene-3 (BAG3) is expressed ubiquitously in humans, but its levels are highest in the heart, the skeletal muscle, and the central nervous system; it is also elevated in many cancers. BAG3's diverse functions are supported by its multiple protein-protein binding domains, which couple with small and large heat shock proteins, members of the Bcl2 family, other antiapoptotic proteins, and various sarcomere proteins. In the heart, BAG3 inhibits apoptosis, promotes autophagy, couples the β-adrenergic receptor with the L-type Ca2+ channel, and maintains the structure of the sarcomere. In cancer cells, BAG3 binds to and supports an identical array of prosurvival proteins, and it may represent a therapeutic target. However, the development of strategies to block BAG3 function in cancer cells may be challenging, as they are likely to interfere with the essential roles of BAG3 in the heart. In this Review, we present the current knowledge regarding the biology of this complex protein in the heart and in cancer and suggest several therapeutic options.
Collapse
Affiliation(s)
- Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, USA
| | - Joseph Y Cheung
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Arthur M Feldman
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Affiliation(s)
- Anjali Tiku Owens
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sharlene M Day
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
36
|
Fullenkamp DE, Puckelwartz MJ, McNally EM. Genome-wide association for heart failure: from discovery to clinical use. Eur Heart J 2021; 42:2012-2014. [PMID: 33851998 DOI: 10.1093/eurheartj/ehab172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Dominic E Fullenkamp
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Megan J Puckelwartz
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Garnier S, Harakalova M, Weiss S, Mokry M, Regitz-Zagrosek V, Hengstenberg C, Cappola TP, Isnard R, Arbustini E, Cook SA, van Setten J, Calis JJA, Hakonarson H, Morley MP, Stark K, Prasad SK, Li J, O'Regan DP, Grasso M, Müller-Nurasyid M, Meitinger T, Empana JP, Strauch K, Waldenberger M, Marguiles KB, Seidman CE, Kararigas G, Meder B, Haas J, Boutouyrie P, Lacolley P, Jouven X, Erdmann J, Blankenberg S, Wichter T, Ruppert V, Tavazzi L, Dubourg O, Roizes G, Dorent R, de Groote P, Fauchier L, Trochu JN, Aupetit JF, Bilinska ZT, Germain M, Völker U, Hemerich D, Raji I, Bacq-Daian D, Proust C, Remior P, Gomez-Bueno M, Lehnert K, Maas R, Olaso R, Saripella GV, Felix SB, McGinn S, Duboscq-Bidot L, van Mil A, Besse C, Fontaine V, Blanché H, Ader F, Keating B, Curjol A, Boland A, Komajda M, Cambien F, Deleuze JF, Dörr M, Asselbergs FW, Villard E, Trégouët DA, Charron P. Genome-wide association analysis in dilated cardiomyopathy reveals two new players in systolic heart failure on chromosomes 3p25.1 and 22q11.23. Eur Heart J 2021; 42:2000-2011. [PMID: 33677556 PMCID: PMC8139853 DOI: 10.1093/eurheartj/ehab030] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/13/2020] [Accepted: 01/14/2021] [Indexed: 12/31/2022] Open
Abstract
AIMS Our objective was to better understand the genetic bases of dilated cardiomyopathy (DCM), a leading cause of systolic heart failure. METHODS AND RESULTS We conducted the largest genome-wide association study performed so far in DCM, with 2719 cases and 4440 controls in the discovery population. We identified and replicated two new DCM-associated loci on chromosome 3p25.1 [lead single-nucleotide polymorphism (SNP) rs62232870, P = 8.7 × 10-11 and 7.7 × 10-4 in the discovery and replication steps, respectively] and chromosome 22q11.23 (lead SNP rs7284877, P = 3.3 × 10-8 and 1.4 × 10-3 in the discovery and replication steps, respectively), while confirming two previously identified DCM loci on chromosomes 10 and 1, BAG3 and HSPB7. A genetic risk score constructed from the number of risk alleles at these four DCM loci revealed a 3-fold increased risk of DCM for individuals with 8 risk alleles compared to individuals with 5 risk alleles (median of the referral population). In silico annotation and functional 4C-sequencing analyses on iPSC-derived cardiomyocytes identify SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine transporter whose involvement in myocardial dysfunction and DCM is supported by numerous observations in humans and animals. At the 22q11.23 locus, in silico and data mining annotations, and to a lesser extent functional analysis, strongly suggest SMARCB1 as the candidate culprit gene. CONCLUSION This study provides a better understanding of the genetic architecture of DCM and sheds light on novel biological pathways underlying heart failure.
Collapse
Affiliation(s)
- Sophie Garnier
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
| | - Magdalena Harakalova
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Michal Mokry
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, the Netherlands
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charite University Hospital, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Christian Hengstenberg
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Austria
- Department of Internal Medicine, Medical University of Regensburg, Germany
| | - Thomas P Cappola
- Penn Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Isnard
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
- Cardiology Department, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Stuart A Cook
- National Heart and Lung Institute, Imperial College London, London, UK
- National Heart Centre Singapore, Singapore
- Duke-NUS, Singapore
| | - Jessica van Setten
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jorg J A Calis
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael P Morley
- Penn Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Klaus Stark
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Sanjay K Prasad
- National Heart Centre Singapore, Singapore
- Royal Brompton Hospital, London, UK
| | - Jin Li
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Declan P O'Regan
- Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Maurizia Grasso
- Centre for Inherited Cardiovascular Diseases—IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Thomas Meitinger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jean-Philippe Empana
- Université de Paris, INSERM, UMR-S970, Integrative Epidemiology of cardiovascular disease, Paris, France
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz 55101, Germany
| | - Melanie Waldenberger
- Research unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Kenneth B Marguiles
- Penn Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christine E Seidman
- Department of Medicine and Genetics Harvard Medical School, Boston, MA, USA
- Brigham & Women's Cardiovascular Genetics Center, Boston, MA, USA
| | - Georgios Kararigas
- Department of Physiology, Faculty of Medicine, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavík, Iceland
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg, Heidelberg University, Germany
- Stanford Genome Technology Center, Department of Genetics, Stanford Medical School, CA, USA
| | - Jan Haas
- Institute for Cardiomyopathies Heidelberg, Heidelberg University, Germany
| | - Pierre Boutouyrie
- Université de Paris, INSERM, UMR-S970, Integrative Epidemiology of cardiovascular disease, Paris, France
- Cardiology Department, APHP, Georges Pompidou European Hospital, Paris, France
| | | | - Xavier Jouven
- Université de Paris, INSERM, UMR-S970, Integrative Epidemiology of cardiovascular disease, Paris, France
- Cardiology Department, APHP, Georges Pompidou European Hospital, Paris, France
| | - Jeanette Erdmann
- Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | | | - Thomas Wichter
- Dept. of Cardiology and Angiology, Niels-Stensen-Kliniken Marienhospital Osnabrück, Heart Centre Osnabrück/Bad Rothenfelde, Osnabrück 49074, Germany
| | - Volker Ruppert
- Klinik für Innere Medizin-Kardiologie UKGM GmbH Standort Marburg Baldingerstrasse, Marburg, Germany
| | - Luigi Tavazzi
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Olivier Dubourg
- Université de Versailles-Saint Quentin, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Gérard Roizes
- Institut de Génétique Humaine, UPR 1142, CNRS, Montpellier, France
| | | | | | - Laurent Fauchier
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau, Tours, France
| | - Jean-Noël Trochu
- Université de Nantes, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes 44000, France
| | - Jean-François Aupetit
- Département de pathologie cardiovasculaire, Hôpital Saint-Joseph-Saint-Luc, Lyon, France
| | - Zofia T Bilinska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, National Institute of Cardiology, Warsaw, Poland
| | - Marine Germain
- Univ. Bordeaux, INSERM, BPH, U1219, Bordeaux 33000, France
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Daiane Hemerich
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ibticem Raji
- AP-HP, Département de Génétique, Centre de Référence Maladies Cardiaques Héréditaires, Hôpital Pitié-Salpêtrière, Paris, France
| | - Delphine Bacq-Daian
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Carole Proust
- Univ. Bordeaux, INSERM, BPH, U1219, Bordeaux 33000, France
| | - Paloma Remior
- Department of Cardiology, Hospital Universitario Puerta de Hierro, CIBERCV, Madrid, Spain
| | - Manuel Gomez-Bueno
- Department of Cardiology, Hospital Universitario Puerta de Hierro, CIBERCV, Madrid, Spain
| | - Kristin Lehnert
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Renee Maas
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robert Olaso
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Ganapathi Varma Saripella
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- SLU Bioinformatics Infrastructure (SLUBI), PlantLink, Department of Plant Breeding, Swedish University of Agricultural Sciences, Almas Allé 8, 750 07 Uppsala, Sweden
| | - Stephan B Felix
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Steven McGinn
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Laëtitia Duboscq-Bidot
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
| | - Alain van Mil
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Céline Besse
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Vincent Fontaine
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
| | - Hélène Blanché
- Laboratory of Excellence GENMED (Medical Genomics)
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Flavie Ader
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- APHP, UF Cardiogénétique et Myogénétique, service de Biochimie métabolique, Hôpital universitaire Pitié-Salpêtrière Paris, France
- Faculté de Pharmacie Paris Descartes, Département 3, Paris 75006, France
| | - Brendan Keating
- Division of Transplantation, Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Angélique Curjol
- AP-HP, Département de Génétique, Centre de Référence Maladies Cardiaques Héréditaires, Hôpital Pitié-Salpêtrière, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Michel Komajda
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
- Cardiology Department, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | | | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry 91057, France
- Laboratory of Excellence GENMED (Medical Genomics)
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Eric Villard
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
| | - David-Alexandre Trégouët
- Univ. Bordeaux, INSERM, BPH, U1219, Bordeaux 33000, France
- Laboratory of Excellence GENMED (Medical Genomics)
| | - Philippe Charron
- Sorbonne Université, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris 75013, France
- Cardiology Department, APHP, Pitié-Salpêtrière Hospital, Paris, France
- AP-HP, Département de Génétique, Centre de Référence Maladies Cardiaques Héréditaires, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
38
|
Walsh R, Tadros R, Bezzina CR. When genetic burden reaches threshold. Eur Heart J 2021; 41:3849-3855. [PMID: 32350504 PMCID: PMC7599032 DOI: 10.1093/eurheartj/ehaa269] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/07/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
Rare cardiac genetic diseases have generally been considered to be broadly Mendelian in nature, with clinical genetic testing for these conditions predicated on the detection of a primary causative rare pathogenic variant that will enable cascade genetic screening in families. However, substantial variability in penetrance and disease severity among carriers of pathogenic variants, as well as the inability to detect rare Mendelian variants in considerable proportions of patients, indicates that more complex aetiologies are likely to underlie these diseases. Recent findings have suggested genetic variants across a range of population frequencies and effect sizes may combine, along with non-genetic factors, to determine whether the threshold for expression of disease is reached and the severity of the phenotype. The availability of increasingly large genetically characterized cohorts of patients with rare cardiac diseases is enabling the discovery of common genetic variation that may underlie both variable penetrance in Mendelian diseases and the genetic aetiology of apparently non-Mendelian rare cardiac conditions. It is likely that the genetic architecture of rare cardiac diseases will vary considerably between different conditions as well as between patients with similar phenotypes, ranging from near-Mendelian disease to models more akin to common, complex disease. Uncovering the broad range of genetic factors that predispose patients to rare cardiac diseases offers the promise of improved risk prediction and more focused clinical management in patients and their families.
Collapse
Affiliation(s)
- Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Rafik Tadros
- Department of Medicine, Cardiovascular Genetics Center, Montreal Heart Institute and Faculty of Medicine, Université de Montréal, 5000 Belanger, Montreal, QC H1T 1C8, Canada
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| |
Collapse
|
39
|
Hershberger RE, Cowan J, Jordan E, Kinnamon DD. The Complex and Diverse Genetic Architecture of Dilated Cardiomyopathy. Circ Res 2021; 128:1514-1532. [PMID: 33983834 DOI: 10.1161/circresaha.121.318157] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our insight into the diverse and complex nature of dilated cardiomyopathy (DCM) genetic architecture continues to evolve rapidly. The foundations of DCM genetics rest on marked locus and allelic heterogeneity. While DCM exhibits a Mendelian, monogenic architecture in some families, preliminary data from our studies and others suggests that at least 20% to 30% of DCM may have an oligogenic basis, meaning that multiple rare variants from different, unlinked loci, determine the DCM phenotype. It is also likely that low-frequency and common genetic variation contribute to DCM complexity, but neither has been examined within a rare variant context. Other types of genetic variation are also likely relevant for DCM, along with gene-by-environment interaction, now established for alcohol- and chemotherapy-related DCM. Collectively, this suggests that the genetic architecture of DCM is broader in scope and more complex than previously understood. All of this elevates the impact of DCM genetics research, as greater insight into the causes of DCM can lead to interventions to mitigate or even prevent it and thus avoid the morbid and mortal scourge of human heart failure.
Collapse
Affiliation(s)
- Ray E Hershberger
- Divisions of Cardiovascular Medicine (R.E.H.), The Ohio State University Wexner Medical Center, Columbus.,Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Jason Cowan
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Elizabeth Jordan
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Daniel D Kinnamon
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
40
|
Fatkin D, Calkins H, Elliott P, James CA, Peters S, Kovacic JC. Contemporary and Future Approaches to Precision Medicine in Inherited Cardiomyopathies: JACC Focus Seminar 3/5. J Am Coll Cardiol 2021; 77:2551-2572. [PMID: 34016267 DOI: 10.1016/j.jacc.2020.12.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023]
Abstract
Inherited cardiomyopathies are commonly occurring myocardial disorders that are associated with substantial morbidity and mortality. Clinical management strategies have focused on treatment of heart failure and arrhythmic complications in symptomatic patients according to standardized guidelines. Clinicians are now being urged to implement precision medicine, but what does this involve? Advances in understanding of the genetic underpinnings of inherited cardiomyopathies have brought new possibilities for interventions that are tailored to genes, specific variants, or downstream mechanisms. However, the phenotypic variability that can occur with any given pathogenic variant suggests that factors other than single driver gene mutations are often involved. This is propelling a new imperative to elucidate the nuanced ways in which individual combinations of genetic variation, comorbidities, and lifestyle may influence cardiomyopathy phenotypes. Here, Part 3 of a 5-part precision medicine Focus Seminar series reviews the current status and future opportunities for precision medicine in the inherited cardiomyopathies.
Collapse
Affiliation(s)
- Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, New South Wales, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia.
| | - Hugh Calkins
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Perry Elliott
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom; Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Cynthia A James
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Stacey Peters
- Departments of Cardiology and Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason C Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, New South Wales, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
41
|
Kadhi A, Mohammed F, Nemer G. The Genetic Pathways Underlying Immunotherapy in Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:613295. [PMID: 33937353 PMCID: PMC8079649 DOI: 10.3389/fcvm.2021.613295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a global public health threat affecting 26 million individuals worldwide with an estimated prevalence increase of 46% by 2030. One of the main causes of HF and sudden death in children and adult is Dilated Cardiomyopathy (DCM). DCM is characterized by dilation and systolic dysfunction of one or both ventricles. It has an underlying genetic basis or can develop subsequent to various etiologies that cause myocardium inflammation (secondary causes). The morbidity and mortality rates of DCM remains high despite recent advancement to manage the disease. New insights have been dedicated to better understand the pathogenesis of DCM in respect to genetic and inflammatory basis by linking the two entities together. This cognizance in the field of cardiology might have an innovative approach to manage DCM through targeted treatment directed to the causative etiology. The following review summarizes the genetical and inflammatory causes underlying DCM and the pathways of the novel precision-medicine-based immunomodulatory strategies to salvage and prevent the associated heart failure linked to the disease.
Collapse
Affiliation(s)
- Ayat Kadhi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Fathima Mohammed
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Georges Nemer
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
42
|
Muranova LK, Shatov VM, Bukach OV, Gusev NB. Cardio-Vascular Heat Shock Protein (cvHsp, HspB7), an Unusual Representative of Small Heat Shock Protein Family. BIOCHEMISTRY (MOSCOW) 2021; 86:S1-S11. [PMID: 33827396 DOI: 10.1134/s0006297921140017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
HspB7 is one of ten human small heat shock proteins. This protein is expressed only in insulin-dependent tissues (heart, skeletal muscle, and fat tissue), and expression of HspB7 is regulated by many different factors. Single nucleotide polymorphism is characteristic for the HspB7 gene and this polymorphism correlates with cardio-vascular diseases and obesity. HspB7 has an unusual N-terminal sequence, a conservative α-crystallin domain, and very short C-terminal domain lacking conservative IPV tripeptide involved in a small heat shock proteins oligomer formation. Nevertheless, in the isolated state HspB7 forms both small oligomers (probably dimers) and very large oligomers (aggregates). HspB7 is ineffective in suppression of amorphous aggregation of model proteins induced by heating or reduction of disulfide bonds, however it is very effective in prevention of aggregation of huntingtin fragments enriched with Gln residues. HspB7 can be an effective sensor of electrophilic agents. This protein interacts with the contractile and cytoskeleton proteins (filamin C, titin, and actin) and participates in protection of the contractile apparatus and cytoskeleton from different adverse conditions. HspB7 possesses tumor suppressive activity. Further investigations are required to understand molecular mechanisms of HspB7 participation in numerous biological processes.
Collapse
Affiliation(s)
- Lydia K Muranova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Vladislav M Shatov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Olesya V Bukach
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nikolai B Gusev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
43
|
Sammani A, Baas AF, Asselbergs FW, te Riele ASJM. Diagnosis and Risk Prediction of Dilated Cardiomyopathy in the Era of Big Data and Genomics. J Clin Med 2021; 10:921. [PMID: 33652931 PMCID: PMC7956169 DOI: 10.3390/jcm10050921] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/19/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure and life-threatening ventricular arrhythmias (LTVA). Work-up and risk stratification of DCM is clinically challenging, as there is great heterogeneity in phenotype and genotype. Throughout the last decade, improved genetic testing of patients has identified genotype-phenotype associations and enhanced evaluation of at-risk relatives leading to better patient prognosis. The field is now ripe to explore opportunities to improve personalised risk assessments. Multivariable risk models presented as "risk calculators" can incorporate a multitude of clinical variables and predict outcome (such as heart failure hospitalisations or LTVA). In addition, genetic risk scores derived from genome/exome-wide association studies can estimate an individual's lifetime genetic risk of developing DCM. The use of clinically granular investigations, such as late gadolinium enhancement on cardiac magnetic resonance imaging, is warranted in order to increase predictive performance. To this end, constructing big data infrastructures improves accessibility of data by using electronic health records, existing research databases, and disease registries. By applying methods such as machine and deep learning, we can model complex interactions, identify new phenotype clusters, and perform prognostic modelling. This review aims to provide an overview of the evolution of DCM definitions as well as its clinical work-up and considerations in the era of genomics. In addition, we present exciting examples in the field of big data infrastructures, personalised prognostic assessment, and artificial intelligence.
Collapse
Affiliation(s)
- Arjan Sammani
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
| | - Annette F. Baas
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, University of Utrecht, 3582 CX Utrecht, The Netherlands;
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London WC1E 6BT, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London WC1E 6BT, UK
| | - Anneline S. J. M. te Riele
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
| |
Collapse
|
44
|
Tadros R, Francis C, Xu X, Vermeer AMC, Harper AR, Huurman R, Kelu Bisabu K, Walsh R, Hoorntje ET, Te Rijdt WP, Buchan RJ, van Velzen HG, van Slegtenhorst MA, Vermeulen JM, Offerhaus JA, Bai W, de Marvao A, Lahrouchi N, Beekman L, Karper JC, Veldink JH, Kayvanpour E, Pantazis A, Baksi AJ, Whiffin N, Mazzarotto F, Sloane G, Suzuki H, Schneider-Luftman D, Elliott P, Richard P, Ader F, Villard E, Lichtner P, Meitinger T, Tanck MWT, van Tintelen JP, Thain A, McCarty D, Hegele RA, Roberts JD, Amyot J, Dubé MP, Cadrin-Tourigny J, Giraldeau G, L'Allier PL, Garceau P, Tardif JC, Boekholdt SM, Lumbers RT, Asselbergs FW, Barton PJR, Cook SA, Prasad SK, O'Regan DP, van der Velden J, Verweij KJH, Talajic M, Lettre G, Pinto YM, Meder B, Charron P, de Boer RA, Christiaans I, Michels M, Wilde AAM, Watkins H, Matthews PM, Ware JS, Bezzina CR. Shared genetic pathways contribute to risk of hypertrophic and dilated cardiomyopathies with opposite directions of effect. Nat Genet 2021; 53:128-134. [PMID: 33495596 PMCID: PMC7611259 DOI: 10.1038/s41588-020-00762-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 12/10/2020] [Indexed: 01/29/2023]
Abstract
The heart muscle diseases hypertrophic (HCM) and dilated (DCM) cardiomyopathies are leading causes of sudden death and heart failure in young, otherwise healthy, individuals. We conducted genome-wide association studies and multi-trait analyses in HCM (1,733 cases), DCM (5,521 cases) and nine left ventricular (LV) traits (19,260 UK Biobank participants with structurally normal hearts). We identified 16 loci associated with HCM, 13 with DCM and 23 with LV traits. We show strong genetic correlations between LV traits and cardiomyopathies, with opposing effects in HCM and DCM. Two-sample Mendelian randomization supports a causal association linking increased LV contractility with HCM risk. A polygenic risk score explains a significant portion of phenotypic variability in carriers of HCM-causing rare variants. Our findings thus provide evidence that polygenic risk score may account for variability in Mendelian diseases. More broadly, we provide insights into how genetic pathways may lead to distinct disorders through opposing genetic effects.
Collapse
Affiliation(s)
- Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada.
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Catherine Francis
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Xiao Xu
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexa M C Vermeer
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Genetics, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART)
| | - Andrew R Harper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Oxford, UK
| | - Roy Huurman
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ken Kelu Bisabu
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Edgar T Hoorntje
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Wouter P Te Rijdt
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Rachel J Buchan
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hannah G van Velzen
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jentien M Vermeulen
- Department of Psychiatry, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Joost Allard Offerhaus
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wenjia Bai
- Data Science Institute, Imperial College London, London, UK
- Department of Brain Sciences and UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, Imperial College London, London, UK
| | - Antonio de Marvao
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Najim Lahrouchi
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Leander Beekman
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jacco C Karper
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Elham Kayvanpour
- Institute for Cardiomyopathies, Heidelberg Heart Center, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Antonis Pantazis
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
| | - A John Baksi
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicola Whiffin
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Francesco Mazzarotto
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Geraldine Sloane
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hideaki Suzuki
- Department of Brain Sciences and UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, Imperial College London, London, UK
- Department of Cardiovascular Medicine, Tohoku University Hospital, Seiryo, Aoba, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Seiryo, Aoba, Sendai, Japan
| | - Deborah Schneider-Luftman
- The Francis Crick Institute, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Pascale Richard
- Service de biochimie métabolique, UF de cardiogénétique et myogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Faculté de Médecine, Sorbonne Université, Paris, France
| | - Flavie Ader
- Service de biochimie métabolique, UF de cardiogénétique et myogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Faculté de Médecine, Sorbonne Université, Paris, France
- Faculté de Pharmacie, Université de Paris, Paris, France
| | - Eric Villard
- INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Faculté de Médecine, Sorbonne Université, Paris, France
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum Muenchen, Neuherberg, Germany
- Klinikum rechts der Isar der TU Muenchen School of Medicine, Institute of Human Genetics, Munich, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael W T Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam Public Health (APH), Amsterdam UMC, Amsterdam, the Netherlands
| | - J Peter van Tintelen
- Department of Clinical Genetics, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Andrew Thain
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - David McCarty
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jason D Roberts
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Julie Amyot
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Marie-Pierre Dubé
- Montreal Heart Institute Research Center, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Geneviève Giraldeau
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Philippe L L'Allier
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Patrick Garceau
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute Research Center, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - S Matthijs Boekholdt
- Department of Cardiology, University of Amsterdam, Heartcenter, Amsterdam UMC, Amsterdam, the Netherlands
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, Gibbs Building, London, UK
- Barts Heart Centre, Saint Bartholomew's Hospital, London, UK
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Institute of Cardiovascular Science and Institute of Health Informatics, Faculty of Population Health Sciences, University College London, London, UK
| | - Paul J R Barton
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Stuart A Cook
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Sanjay K Prasad
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Declan P O'Regan
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Karin J H Verweij
- Department of Psychiatry, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Mario Talajic
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute Research Center, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Yigal M Pinto
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART)
| | - Benjamin Meder
- Institute for Cardiomyopathies, Heidelberg Heart Center, University of Heidelberg, Heidelberg, Germany
| | - Philippe Charron
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART)
- INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Faculté de Médecine, Sorbonne Université, Paris, France
- Département de Génétique, Centre de référence des maladies cardiaques héréditaires ou rares, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Imke Christiaans
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Michels
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART)
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Oxford, UK
| | - Paul M Matthews
- Department of Brain Sciences and UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, Imperial College London, London, UK
| | - James S Ware
- Cardiovascular Research Centre, Royal Brompton and Harefield National Health Service Foundation Trust, London, UK.
- National Heart and Lung Institute, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, Imperial College London, London, UK.
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), .
| |
Collapse
|
45
|
Harper AR, Goel A, Grace C, Thomson KL, Petersen SE, Xu X, Waring A, Ormondroyd E, Kramer CM, Ho CY, Neubauer S, Tadros R, Ware JS, Bezzina CR, Farrall M, Watkins H. Common genetic variants and modifiable risk factors underpin hypertrophic cardiomyopathy susceptibility and expressivity. Nat Genet 2021; 53:135-142. [PMID: 33495597 DOI: 10.1038/s41588-020-00764-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common, serious, genetic heart disorder. Rare pathogenic variants in sarcomere genes cause HCM, but with unexplained phenotypic heterogeneity. Moreover, most patients do not carry such variants. We report a genome-wide association study of 2,780 cases and 47,486 controls that identified 12 genome-wide-significant susceptibility loci for HCM. Single-nucleotide polymorphism heritability indicated a strong polygenic influence, especially for sarcomere-negative HCM (64% of cases; h2g = 0.34 ± 0.02). A genetic risk score showed substantial influence on the odds of HCM in a validation study, halving the odds in the lowest quintile and doubling them in the highest quintile, and also influenced phenotypic severity in sarcomere variant carriers. Mendelian randomization identified diastolic blood pressure (DBP) as a key modifiable risk factor for sarcomere-negative HCM, with a one standard deviation increase in DBP increasing the HCM risk fourfold. Common variants and modifiable risk factors have important roles in HCM that we suggest will be clinically actionable.
Collapse
Affiliation(s)
- Andrew R Harper
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Anuj Goel
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christopher Grace
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kate L Thomson
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.,Oxford Medical Genetics Laboratories, Churchill Hospital, Oxford, UK
| | - Steffen E Petersen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Xiao Xu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam Waring
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elizabeth Ormondroyd
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefan Neubauer
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | | | - Rafik Tadros
- Cardiovascular Genetics Centre, Montréal Heart Institute, Montréal, Québec, Canada
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Martin Farrall
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK. .,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK. .,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
46
|
Spracklen TF, Chakafana G, Schwartz PJ, Kotta MC, Shaboodien G, Ntusi NAB, Sliwa K. Genetics of Peripartum Cardiomyopathy: Current Knowledge, Future Directions and Clinical Implications. Genes (Basel) 2021; 12:genes12010103. [PMID: 33467574 PMCID: PMC7830587 DOI: 10.3390/genes12010103] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a condition in which heart failure and systolic dysfunction occur late in pregnancy or within months following delivery. Over the last decade, genetic advances in heritable cardiomyopathy have provided new insights into the role of genetics in PPCM. In this review, we summarise current knowledge of the genetics of PPCM and potential avenues for further research, including the role of molecular chaperone mutations in PPCM. Evidence supporting a genetic basis for PPCM has emanated from observations of familial disease, overlap with familial dilated cardiomyopathy, and sequencing studies of PPCM cohorts. Approximately 20% of PPCM patients screened for cardiomyopathy genes have an identified pathogenic mutation, with TTN truncations most commonly implicated. As a stress-associated condition, PPCM may be modulated by molecular chaperones such as heat shock proteins (Hsps). Recent studies have led to the identification of Hsp mutations in a PPCM model, suggesting that variation in these stress-response genes may contribute to PPCM pathogenesis. Although some Hsp genes have been implicated in dilated cardiomyopathy, their roles in PPCM remain to be determined. Additional areas of future investigation may include the delineation of genotype-phenotype correlations and the screening of newly-identified cardiomyopathy genes for their roles in PPCM. Nevertheless, these findings suggest that the construction of a family history may be advised in the management of PPCM and that genetic testing should be considered. A better understanding of the genetics of PPCM holds the potential to improve treatment, prognosis, and family management.
Collapse
Affiliation(s)
- Timothy F. Spracklen
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Division of Cardiology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Graham Chakafana
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Division of Cardiology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Peter J. Schwartz
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano, IRCCS, 20135 Milan, Italy;
| | - Maria-Christina Kotta
- Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano, IRCCS, 20135 Milan, Italy;
| | - Gasnat Shaboodien
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Division of Cardiology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Ntobeko A. B. Ntusi
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Division of Cardiology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa & CHI, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa; (T.F.S.); (G.C.); (P.J.S.); (G.S.); (N.A.B.N.)
- Division of Cardiology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Correspondence:
| |
Collapse
|
47
|
Canut MI, Villa O, Kudsieh B, Mattlin H, Banchs I, González JR, Armengol L, Casaroli-Marano RP. MLIP genotype as a predictor of pharmacological response in primary open-angle glaucoma and ocular hypertension. Sci Rep 2021; 11:1583. [PMID: 33452295 PMCID: PMC7810753 DOI: 10.1038/s41598-020-80954-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/24/2020] [Indexed: 11/20/2022] Open
Abstract
Predicting the therapeutic response to ocular hypotensive drugs is crucial for the clinical treatment and management of glaucoma. Our aim was to identify a possible genetic contribution to the response to current pharmacological treatments of choice in a white Mediterranean population with primary open-angle glaucoma (POAG) or ocular hypertension (OH). We conducted a prospective, controlled, randomized, partial crossover study that included 151 patients of both genders, aged 18 years and older, diagnosed with and requiring pharmacological treatment for POAG or OH in one or both eyes. We sought to identify copy number variants (CNVs) associated with differences in pharmacological response, using a DNA pooling strategy of carefully phenotyped treatment responders and non-responders, treated for a minimum of 6 weeks with a beta-blocker (timolol maleate) and/or prostaglandin analog (latanoprost). Diurnal intraocular pressure reduction and comparative genome wide CNVs were analyzed. Our finding that copy number alleles of an intronic portion of the MLIP gene is a predictor of pharmacological response to beta blockers and prostaglandin analogs could be used as a biomarker to guide first-tier POAG and OH treatment. Our finding improves understanding of the genetic factors modulating pharmacological response in POAG and OH, and represents an important contribution to the establishment of a personalized approach to the treatment of glaucoma.
Collapse
Affiliation(s)
- María I Canut
- Centro de Oftalmología Barraquer, Instituto Universitario Barraquer (UAB), Barcelona, Spain
| | - Olaya Villa
- Quantitative Genomic Medicine Laboratories (qGenomics), Esplugues del Llobregat, Spain
| | | | - Heidi Mattlin
- Quantitative Genomic Medicine Laboratories (qGenomics), Esplugues del Llobregat, Spain
| | - Isabel Banchs
- Quantitative Genomic Medicine Laboratories (qGenomics), Esplugues del Llobregat, Spain
| | - Juan R González
- Barcelona Institute for Global Health (ISGlobal) and Centro de Investigación Biomédica en Red en Epidemiologia Y Salud Pública (CIBERESP), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Lluís Armengol
- Quantitative Genomic Medicine Laboratories (qGenomics), Esplugues del Llobregat, Spain.
| | - Ricardo P Casaroli-Marano
- Department of Surgery, School of Medicine and Health Sciences and Hospital Clinic de Barcelona (IDIBAPS), University of Barcelona, Calle Sabino de Arana 1 (2nd floor, Ophthalmology), 08028, Barcelona, Spain. .,Institute of Biomedical Research Sant Pau (IIB-Sant Pau, SGR1113) and Barcelona Tissue Bank (BST), Barcelona, Spain.
| |
Collapse
|
48
|
Yingling CV, Pruyne D. FHOD formin and SRF promote post-embryonic striated muscle growth through separate pathways in C. elegans. Exp Cell Res 2021; 398:112388. [PMID: 33221314 PMCID: PMC7750259 DOI: 10.1016/j.yexcr.2020.112388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 11/28/2022]
Abstract
Previous work with cultured cells has shown transcription of muscle genes by serum response factor (SRF) can be stimulated by actin polymerization driven by proteins of the formin family. However, it is not clear if endogenous formins similarly promote SRF-dependent transcription during muscle development in vivo. We tested whether formin activity promotes SRF-dependent transcription in striated muscle in the simple animal model, Caenorhabditis elegans. Our lab has shown FHOD-1 is the only formin that directly promotes sarcomere formation in the worm's striated muscle. We show here FHOD-1 and SRF homolog UNC-120 both support muscle growth and also muscle myosin II heavy chain A expression. However, while a hypomorphic unc-120 allele blunts expression of a set of striated muscle genes, these genes are largely upregulated or unchanged by absence of FHOD-1. Instead, pharmacological inhibition of the proteasome restores myosin protein levels in worms lacking FHOD-1, suggesting elevated proteolysis accounts for their myosin deficit. Interestingly, proteasome inhibition does not restore normal muscle growth to fhod-1(Δ) mutants, suggesting formin contributes to muscle growth by some alternative mechanism. Overall, we find SRF does not depend on formin to promote muscle gene transcription in a simple in vivo system.
Collapse
Affiliation(s)
- Curtis V Yingling
- Department of Cell and Developmental Biology, 107 Weiskotten Hall, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA.
| | - David Pruyne
- Department of Cell and Developmental Biology, 107 Weiskotten Hall, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA.
| |
Collapse
|
49
|
de Denus S, Mottet F, Korol S, Feroz Zada Y, Provost S, Mongrain I, Asselin G, Oussaïd E, Busseuil D, Lettre G, Rioux J, Racine N, O'Meara E, White M, Rouleau J, Tardif JC, Dubé MP. A genetic association study of heart failure: more evidence for the role of BAG3 in idiopathic dilated cardiomyopathy. ESC Heart Fail 2020; 7:4384-4389. [PMID: 32869539 PMCID: PMC7754954 DOI: 10.1002/ehf2.12934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/22/2023] Open
Abstract
Aims Few investigations have been conducted to identify genetic determinants of common, polygenetic forms of heart failure (HF), and only a limited number of these genetic associations have been validated by multiple groups. Methods and results We performed a case–control study to further investigate the potential impact of 14 previously reported candidate genes on the risk of HF and specific HF sub‐types. We also performed an exploratory genome‐wide study. We included 799 patients with HF and 1529 controls. After adjusting for age, sex, and genetic ancestry, we found that the C allele of rs2234962 in BAG3 was associated with a decreased risk of idiopathic dilated cardiomyopathy (odds ratio 0.42, 95% confidence interval 0.25–0.68, P = 0.0005), consistent with a previous report. No association for the other primary variants or exploratory genome‐wide study was found. Conclusions Our findings provide independent replication for the association between a common coding variant (rs2234962) in BAG3 and the risk of idiopathic dilated cardiomyopathy.
Collapse
Affiliation(s)
- Simon de Denus
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - Fannie Mottet
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Sandra Korol
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Yassamin Feroz Zada
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - Sylvie Provost
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - Ian Mongrain
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - Géraldine Asselin
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - Essaïd Oussaïd
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada
| | - David Busseuil
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - John Rioux
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Normand Racine
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Eileen O'Meara
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jean Rouleau
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jean Claude Tardif
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Marie-Pierre Dubé
- Montreal Heart Institute, 5000 Bélanger, Montreal, QC, H1T 1C8, Canada.,Université de Montreal Beaulieu-Saucier Pharmacogenomics Center, Montreal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
50
|
Liu J, Huang ZP, Nie M, Wang G, Silva WJ, Yang Q, Freire PP, Hu X, Chen H, Deng Z, Pu WT, Wang DZ. Regulation of myonuclear positioning and muscle function by the skeletal muscle-specific CIP protein. Proc Natl Acad Sci U S A 2020; 117:19254-19265. [PMID: 32719146 PMCID: PMC7430979 DOI: 10.1073/pnas.1922911117] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The appropriate arrangement of myonuclei within skeletal muscle myofibers is of critical importance for normal muscle function, and improper myonuclear localization has been linked to a variety of skeletal muscle diseases, such as centronuclear myopathy and muscular dystrophies. However, the molecules that govern myonuclear positioning remain elusive. Here, we report that skeletal muscle-specific CIP (sk-CIP) is a regulator of nuclear positioning. Genetic deletion of sk-CIP in mice results in misalignment of myonuclei along the myofibers and at specialized structures such as neuromuscular junctions (NMJs) and myotendinous junctions (MTJs) in vivo, impairing myonuclear positioning after muscle regeneration, leading to severe muscle dystrophy in mdx mice, a mouse model of Duchenne muscular dystrophy. sk-CIP is localized to the centrosome in myoblasts and relocates to the outer nuclear envelope in myotubes upon differentiation. Mechanistically, we found that sk-CIP interacts with the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex and the centriole Microtubule Organizing Center (MTOC) proteins to coordinately modulate myonuclear positioning and alignment. These findings indicate that sk-CIP may function as a muscle-specific anchoring protein to regulate nuclear position in multinucleated muscle cells.
Collapse
MESH Headings
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Co-Repressor Proteins
- Humans
- Mice
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Myoblasts/metabolism
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/physiopathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organ Specificity
Collapse
Affiliation(s)
- Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Zhan-Peng Huang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Center for Translational Medicine, National Health Commission (NHC) Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Gang Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - William J Silva
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Laboratório de Biologia Celular e Molecular do Músculo Estriado, University of São Paulo, CEP 05508-000 Cidade Universitária, São Paulo, Brazil
| | - Qiumei Yang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Animal Sciences, Sichuan Agriculture University, Chengdu, 611130, China
| | - Paula P Freire
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Morphology, Institute of Biosciences, São Paulo State University, CEP 18618-000, Botucatu, São Paulo, Brazil
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Biology, Nanjing Normal University, Nanjing, 225300, China
| | - Zhongliang Deng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|