51
|
Veith M, El-Battrawy I, Roterberg G, Raschwitz L, Lang S, Wolpert C, Schimpf R, Zhou X, Akin I, Borggrefe M. Long-Term Follow-Up of Patients with Catecholaminergic Polymorphic Ventricular Arrhythmia. J Clin Med 2020; 9:jcm9040903. [PMID: 32218223 PMCID: PMC7230751 DOI: 10.3390/jcm9040903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 11/24/2022] Open
Abstract
Background: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a rare inherited disorder causing life-threatening arrhythmias. Long-term outcome studies of the channelopathy are limited. Objective: The aim of the present study was to summarize our knowledge on CPVT patients, including the clinical profile treatment approach and long-term outcome. Methods: In this single center study, we retrospectively and prospectively collected data from nine CPVT patients and analyzed them. Results: We reviewed nine patients with CPVT in seven families (22% male), with a median follow-up time of 8.6 years. Mean age at diagnosis was 26.4 ± 12 years. Symptoms at admission were syncope (four patients) and aborted cardiac arrest (four patients). Family history of sudden cardiac death was screened in five patients. In genetic analyses, we found five patients with ryanodine type 2 receptor (RYR2) mutations. Seven patients were treated with beta-blockers, and if symptoms persisted flecainide was added (four patients). Despite beta-blocker treatment, three patients suffered from seven adverse cardiac events. An implantable cardioverter defibrillator was implanted in seven patients (one primary, six secondary prevention). Over the follow-up period, three patients suffered from ventricular tachycardia (ten times) and five patients from ventricular fibrillation (nine times). No one died during follow-up. Conclusion: Our CPVT cohort showed a high risk of cardiac events. Family screening, optimal medical therapy and individualized treatment are necessary in affected patients in referral centers.
Collapse
Affiliation(s)
- Michael Veith
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- Correspondence: (M.V.); (I.E.-B.)
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167 Mannheim, Germany
- Correspondence: (M.V.); (I.E.-B.)
| | - Gretje Roterberg
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
| | - Laura Raschwitz
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167 Mannheim, Germany
| | - Christian Wolpert
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
| | - Rainer Schimpf
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167 Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167 Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany; (G.R.); (L.R.); (S.L.); (C.W.); (R.S.); (X.Z.); (I.A.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167 Mannheim, Germany
| |
Collapse
|
52
|
Clemens DJ, Gray B, Bagnall RD, Tester DJ, Dotzler SM, Giudicessi JR, Matthews E, Semsarian C, Behr ER, Ackerman MJ. Triadin Knockout Syndrome Is Absent in a Multi-Center Molecular Autopsy Cohort of Sudden Infant Death Syndrome and Sudden Unexplained Death in the Young and Is Extremely Rare in the General Population. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e002731. [PMID: 32167373 DOI: 10.1161/circgen.119.002731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Triadin knockout syndrome (TKOS) is a potentially lethal arrhythmia disorder caused by recessively inherited null variants in TRDN-encoded cardiac triadin. Despite its malignant phenotype, the prevalence of TKOS in sudden infant death syndrome and sudden unexplained death in the young is unknown. METHODS Exome sequencing was performed on 599 sudden infant death syndrome and 258 sudden unexplained death in the young cases. Allele frequencies of all TRDN null variants identified in the cardiac-specific isoform of TRDN in the Genome Aggregation Database were used to determine the estimated prevalence and ethnic distribution of TKOS. RESULTS No triadin null individuals were identified in 599 sudden infant death syndrome and 258 sudden unexplained death in the young exomes. Using the Genome Aggregation Database, we estimate the overall prevalence of TKOS to be ≈1:22.7 million individuals. However, TKOS prevalence is 5.5-fold higher in those of African descent (≈1:4.1 million). CONCLUSIONS TKOS is an exceedingly rare clinical entity that does not contribute meaningfully to either sudden infant death syndrome or sudden unexplained death in the young. However, despite its rarity and absence in large sudden death cohorts, TKOS remains a malignant and potentially lethal disorder which requires further research to better care for these patients.
Collapse
Affiliation(s)
- Daniel J Clemens
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics (D.J.C., D.J.T., S.M.D., M.J.A.), Mayo Clinic, Rochester, MN
| | - Belinda Gray
- Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom (B.G., E.R.B.).,Cardiology Clinical Academic Group, St George's University Hospitals' National Health Service (NHS) Foundation Trust, London, United Kingdom (B.G., E.R.B.).,Agnes Ginges Centre for Molecular Cardiology at Centenary Institute (B.G., R.D.B., C.S.), The University of Sydney, Australia.,Sydney Medical School, Faculty of Medicine and Health (B.G., R.D.B., C.S.), The University of Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Australia (B.G., C.S.)
| | - Richard D Bagnall
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute (B.G., R.D.B., C.S.), The University of Sydney, Australia.,Sydney Medical School, Faculty of Medicine and Health (B.G., R.D.B., C.S.), The University of Sydney, Australia
| | - David J Tester
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics (D.J.C., D.J.T., S.M.D., M.J.A.), Mayo Clinic, Rochester, MN.,Division of Heart Rhythm Services, Department of Cardiovascular Medicine (D.J.T., J.R.G., M.J.A.), Mayo Clinic, Rochester, MN
| | - Steven M Dotzler
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics (D.J.C., D.J.T., S.M.D., M.J.A.), Mayo Clinic, Rochester, MN
| | - John R Giudicessi
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine (D.J.T., J.R.G., M.J.A.), Mayo Clinic, Rochester, MN
| | - Emma Matthews
- Medical Research Council Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, University College London Institute of Neurology, Queen Square, United Kingdom (E.M.)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute (B.G., R.D.B., C.S.), The University of Sydney, Australia.,Sydney Medical School, Faculty of Medicine and Health (B.G., R.D.B., C.S.), The University of Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Australia (B.G., C.S.)
| | - Elijah R Behr
- Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom (B.G., E.R.B.).,Cardiology Clinical Academic Group, St George's University Hospitals' National Health Service (NHS) Foundation Trust, London, United Kingdom (B.G., E.R.B.)
| | - Michael J Ackerman
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics (D.J.C., D.J.T., S.M.D., M.J.A.), Mayo Clinic, Rochester, MN.,Division of Heart Rhythm Services, Department of Cardiovascular Medicine (D.J.T., J.R.G., M.J.A.), Mayo Clinic, Rochester, MN.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine (M.J.A.), Mayo Clinic, Rochester, MN
| |
Collapse
|
53
|
Olubando D, Hopton C, Eden J, Caswell R, Lowri Thomas N, Roberts SA, Morris-Rosendahl D, Venetucci L, Newman WG. Classification and correlation of RYR2 missense variants in individuals with catecholaminergic polymorphic ventricular tachycardia reveals phenotypic relationships. J Hum Genet 2020; 65:531-539. [PMID: 32152366 DOI: 10.1038/s10038-020-0738-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 11/09/2022]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is predominantly caused by heterozygous missense variants in the cardiac ryanodine receptor, RYR2. However, many RYR2 missense variants are classified as variants of uncertain significance (VUS). We systematically re-evaluated all RYR2 variants in healthy individuals and those with CPVT or arrhythmia using the 2015 American College of Medical Genomics guidelines. RYR2 variants were identified by the NW Genomic Laboratory Hub, from the published literature and databases of sequence variants. Each variant was assessed based on minor allele frequencies, in silico prediction tools and appraisal of functional studies and classified according to the ACMG-AMP guidelines. Phenotype data was collated where available. Of the 326 identified RYR2 missense variants, 55 (16.9%), previously disease-associated variants were reclassified as benign. Application of the gnomAD database of >140,000 controls allowed reclassification of 11 variants more than the ExAC database. CPVT-associated RYR2 variants clustered predominantly between amino acid positions 3949-4332 and 4867-4967 as well as the RyR and IP3R homology-associated and ion transport domains (p < 0.005). CPVT-associated RYR2 variants occurred at more conserved amino acid positions compared with controls, and variants associated with sudden death had higher conservation scores (p < 0.005). There were five potentially pathogenic RYR2 variants associated with sudden death during sleep which were located almost exclusively in the C-terminus of the protein. In conclusion, control sequence databases facilitate reclassification of RYR2 variants but the majority remain as VUS. Notably, pathogenic variants in RYR2 are associated with death in sleep.
Collapse
Affiliation(s)
- Damilola Olubando
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, M13 9WL, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Claire Hopton
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, M13 9WL, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - James Eden
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, M13 9WL, UK
| | - Richard Caswell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - N Lowri Thomas
- School of Pharmacy and Pharmaceutical Sciences, Redwood Building, University of Cardiff, Cardiff, CF10 3NB, UK
| | - Stephen A Roberts
- Division of Population Health, Health Services Research and Primary Care, University of Manchester, Manchester, UK
| | - Deborah Morris-Rosendahl
- Clinical Genetics and Genomics, Royal Brompton and Harefield NHS Foundation Trust, London, UK.,National Heart and Lung Institute (NHLI), Imperial College London, London, UK
| | - Luigi Venetucci
- Manchester Heart Centre, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.,Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - William G Newman
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, M13 9WL, UK. .,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK. .,Peking University Health Sciences Center, Beijing, PR China.
| |
Collapse
|
54
|
Wang Y, Li C, Shi L, Chen X, Cui C, Huang J, Chen B, Hall DD, Pan Z, Lu M, Hong J, Song LS, Zhao S. Integrin β1D Deficiency-Mediated RyR2 Dysfunction Contributes to Catecholamine-Sensitive Ventricular Tachycardia in Arrhythmogenic Right Ventricular Cardiomyopathy. Circulation 2020; 141:1477-1493. [PMID: 32122157 DOI: 10.1161/circulationaha.119.043504] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a hereditary heart disease characterized by fatty infiltration, life-threatening arrhythmias, and increased risk of sudden cardiac death. The guideline for management of ARVC in patients is to improve quality of life by reducing arrhythmic symptoms and to prevent sudden cardiac death. However, the mechanism underlying ARVC-associated cardiac arrhythmias remains poorly understood. METHODS Using protein mass spectrometry analyses, we identified that integrin β1 is downregulated in ARVC hearts without changes to Ca2+-handling proteins. As adult cardiomyocytes express only the β1D isoform, we generated a cardiac specific β1D knockout mouse model and performed functional imaging and biochemical analyses to determine the consequences of integrin β1D loss on function in the heart in vivo and in vitro. RESULTS Integrin β1D deficiency and RyR2 Ser-2030 hyperphosphorylation were detected by Western blotting in left ventricular tissues from patients with ARVC but not in patients with ischemic or hypertrophic cardiomyopathy. Using lipid bilayer patch clamp single channel recordings, we found that purified integrin β1D protein could stabilize RyR2 function by decreasing RyR2 open probability, mean open time, and increasing mean close time. Also, β1D knockout mice exhibited normal cardiac function and morphology but presented with catecholamine-sensitive polymorphic ventricular tachycardia, consistent with increased RyR2 Ser-2030 phosphorylation and aberrant Ca2+ handling in β1D knockout cardiomyocytes. Mechanistically, we revealed that loss of DSP (desmoplakin) induces integrin β1D deficiency in ARVC mediated through an ERK1/2 (extracellular signal-regulated kinase 1 and 2)-fibronectin-ubiquitin/lysosome pathway. CONCLUSIONS Our data suggest that integrin β1D deficiency represents a novel mechanism underlying the increased risk of ventricular arrhythmias in patients with ARVC.
Collapse
Affiliation(s)
- Yihui Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| | - Chunyan Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| | - Ling Shi
- Department of Pharmacology, College of Pharmacy, and State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Heilongjiang, China (L.S., Z.P.)
| | - Xiuyu Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| | - Chen Cui
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| | | | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City (B.C., D.D.H., L.-S.S.)
| | - Duane D Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City (B.C., D.D.H., L.-S.S.)
| | - Zhenwei Pan
- Department of Pharmacology, College of Pharmacy, and State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Heilongjiang, China (L.S., Z.P.)
| | - Minjie Lu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| | - Jiang Hong
- Department of Cardiology, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, China (J.H.)
- Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, China (J.H.)
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City (B.C., D.D.H., L.-S.S.)
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City (L.-S.S.)
- Department of Veterans Affairs Medical Center, Iowa City, IA (L.-S.S.)
| | - Shihua Zhao
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.W., C.L., X.C., C.C., M.L., S.Z.)
| |
Collapse
|
55
|
Schartner V, Laporte J, Böhm J. Abnormal Excitation-Contraction Coupling and Calcium Homeostasis in Myopathies and Cardiomyopathies. J Neuromuscul Dis 2020; 6:289-305. [PMID: 31356215 DOI: 10.3233/jnd-180314] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Muscle contraction requires specialized membrane structures with precise geometry and relies on the concerted interplay of electrical stimulation and Ca2+ release, known as excitation-contraction coupling (ECC). The membrane structure hosting ECC is called triad in skeletal muscle and dyad in cardiac muscle, and structural or functional defects of triads and dyads have been observed in a variety of myopathies and cardiomyopathies. Based on their function, the proteins localized at the triad/dyad can be classified into three molecular pathways: the Ca2+ release complex (CRC), store-operated Ca2+ entry (SOCE), and membrane remodeling. All three are mechanistically linked, and consequently, aberrations in any of these pathways cause similar disease entities. This review provides an overview of the clinical and genetic spectrum of triad and dyad defects with a main focus of attention on the underlying pathomechanisms.
Collapse
Affiliation(s)
- Vanessa Schartner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| |
Collapse
|
56
|
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a rare congenital arrhythmogenic disorder induced by physical or emotional stress. It mainly affects children and younger adults and is characterized by rapid polymorphic and bidirectional ventricular tachycardia. Symptoms can include dizziness, palpitations, and presyncope, which may progress to syncope, hypotonia, convulsive movements, and sudden cardiac death. CPVT is the result of perturbations in Ca ion handling in the sarcoplasmic reticulum of cardiac myocytes. Mutations in the cardiac ryanodine receptor gene and the calsequestrin isoform 2 gene are most commonly seen in familial CPVT patients. Under catecholaminergic stimulation, either mutation can result in an excess Ca load during diastole resulting in delayed after depolarization and subsequent arrhythmogenesis. The current first-line treatment for CPVT is β-blocker therapy. Other therapeutic interventions that can be used in conjunction with β-blockers include moderate exercise training, flecainide, left cardiac sympathetic denervation, and implantable cardioverter-defibrillators. Several potential therapeutic interventions, including verapamil, dantrolene, JTV519, and gene therapy, are also discussed.
Collapse
|
57
|
Cortez AG, Ortiz NA, Argüellez EA, Molina TV, Serra AT, Torres PI, Márquez MF. Catecholaminergic polymorphic ventricular tachycardia due to de novo RyR2 mutation: recreational cycling as a trigger of lethal arrhythmias. Arch Med Sci 2020; 16:466-470. [PMID: 32190159 PMCID: PMC7069429 DOI: 10.5114/aoms.2019.89691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/31/2018] [Indexed: 12/04/2022] Open
Affiliation(s)
- Antonio Gallegos Cortez
- Hospital Regional de Alta Especialidad del Bajío, San Carlos la Roncha, León Guanajuato, Mexico
| | | | | | | | | | | | | |
Collapse
|
58
|
Sarquella-Brugada G, Fernandez-Falgueras A, Cesar S, Arbelo E, Jordà P, García-Álvarez A, Cruzalegui JC, Merchan EF, Fiol V, Brugada J, Brugada R, Campuzano O. Pediatric Malignant Arrhythmias Caused by Rare Homozygous Genetic Variants in TRDN: A Comprehensive Interpretation. Front Pediatr 2020; 8:601708. [PMID: 33692971 PMCID: PMC7938306 DOI: 10.3389/fped.2020.601708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
Aim: To perform a comprehensive phenotype-genotype correlation of all rare variants in Triadin leading to malignant arrhythmias in pediatrics. Methods: Triadin knockout syndrome is a rare entity reported in pediatric population. This syndrome is caused by rare variants in the TRDN gene. Malignant ventricular arrhythmias and sudden cardiac death can be a primary manifestation of disease. Although pharmacological measures are effective, some patients require an implantable defibrillator due to high risk of arrhythmogenic episodes. Main Results: Fourteen rare genetic alterations in TRDN have been reported to date. All of these potentially pathogenic alterations are located in a specific area of TRDN, highlighting this hot spot as an arrhythmogenic gene region. Conclusions: Early recognition and comprehensive interpretation of alterations in Triadin are crucial to adopt preventive measures and avoid malignant arrhythmogenic episodes in pediatric population.
Collapse
Affiliation(s)
- Georgia Sarquella-Brugada
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Medical Science Department, School of Medicine, University of Girona, Girona, Spain
| | | | - Sergi Cesar
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Elena Arbelo
- Arrhythmias Unit, Hospital Clinic, University of Barcelona-IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Paloma Jordà
- Arrhythmias Unit, Hospital Clinic, University of Barcelona-IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ana García-Álvarez
- Arrhythmias Unit, Hospital Clinic, University of Barcelona-IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | - Victoria Fiol
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Josep Brugada
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Arrhythmias Unit, Hospital Clinic, University of Barcelona-IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ramon Brugada
- Medical Science Department, School of Medicine, University of Girona, Girona, Spain.,Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Cardiology Service, Hospital Josep Trueta, University of Girona, Girona, Spain
| | - Oscar Campuzano
- Medical Science Department, School of Medicine, University of Girona, Girona, Spain.,Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
59
|
Cacheux M, Fauconnier J, Thireau J, Osseni A, Brocard J, Roux-Buisson N, Brocard J, Fauré J, Lacampagne A, Marty I. Interplay between Triadin and Calsequestrin in the Pathogenesis of CPVT in the Mouse. Mol Ther 2019; 28:171-179. [PMID: 31607542 DOI: 10.1016/j.ymthe.2019.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 01/07/2023] Open
Abstract
Recessive forms of catecholaminergic polymorphic ventricular tachycardia (CPVT) are induced by mutations in genes encoding triadin or calsequestrin, two proteins that belong to the Ca2+ release complex, responsible for intracellular Ca2+ release triggering cardiac contractions. To better understand the mechanisms of triadin-induced CPVT and to assay multiple therapeutic interventions, we used a triadin knockout mouse model presenting a CPVT-like phenotype associated with a decrease in calsequestrin protein level. We assessed different approaches to rescue protein expression and to correct intracellular Ca2+ release and cardiac function: pharmacological treatment with kifunensine or a viral gene transfer-based approach, using adeno-associated virus serotype 2/9 (AAV2/9) encoding the triadin or calsequestrin. We observed that the levels of triadin and calsequestrin are intimately linked, and that reduction of both proteins contributes to the CPVT phenotype. Different combinations of triadin and calsequestrin expression level were obtained using these therapeutic approaches. A full expression of each is not necessary to correct the phenotype; a fine-tuning of the relative re-expression of both triadin and calsequestrin is required to correct the CPVT phenotype and rescue the cardiac function. AAV-mediated gene delivery of calsequestrin or triadin and treatment with kifunensine are potential treatments for recessive forms of CPVT due to triadin mutations.
Collapse
Affiliation(s)
- Marine Cacheux
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Jérémy Fauconnier
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France
| | - Jérôme Thireau
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France
| | - Alexis Osseni
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Jacques Brocard
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Nathalie Roux-Buisson
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Julie Brocard
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Julien Fauré
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Alain Lacampagne
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France.
| | - Isabelle Marty
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France.
| |
Collapse
|
60
|
Rossi D, Gigli L, Gamberucci A, Bordoni R, Pietrelli A, Lorenzini S, Pierantozzi E, Peretto G, De Bellis G, Della Bella P, Ferrari M, Sorrentino V, Benedetti S, Sala S, Di Resta C. A novel homozygous mutation in the TRDN gene causes a severe form of pediatric malignant ventricular arrhythmia. Heart Rhythm 2019; 17:296-304. [PMID: 31437535 DOI: 10.1016/j.hrthm.2019.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Triadin is a protein expressed in cardiac and skeletal muscle that has an essential role in the structure and functional regulation of calcium release units and excitation-contraction coupling. Mutations in the triadin gene (TRDN) have been described in different forms of human arrhythmia syndromes with early onset and severe arrhythmogenic phenotype, including triadin knockout syndrome. OBJECTIVE The purpose of this study was to characterize the pathogenetic mechanism underlying a case of severe pediatric malignant arrhythmia associated with a defect in the TRDN gene. METHODS We used a trio whole exome sequencing approach to identify the genetic defect in a 2-year-old boy who had been resuscitated from sudden cardiac arrest and had frequent episodes of ventricular fibrillation and a family history positive for sudden death. We then performed in vitro functional analysis to investigate possible pathogenic mechanisms underlying this severe phenotype. RESULTS We identified a novel homozygous missense variant (p.L56P) in the TRDN gene in the proband that was inherited from the heterozygous unaffected parents. Expression of a green fluorescent protein (GFP)-tagged mutant human cardiac triadin isoform (TRISK32-L56P-GFP) in heterologous systems revealed that the mutation alters protein dynamics. Furthermore, when co-expressed with the type 2 ryanodine receptor, caffeine-induced calcium release from TRISK32-L56P-GFP was relatively lower compared to that observed with the wild-type construct. CONCLUSION The results of this study allowed us to hypothesize a pathogenic mechanism underlying this rare arrhythmogenic recessive form, suggesting that the mutant protein potentially can trigger arrhythmias by altering calcium homeostasis.
Collapse
Affiliation(s)
- Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lorenzo Gigli
- Department of Arrhythmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Roberta Bordoni
- Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| | - Alessandro Pietrelli
- Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| | - Stefania Lorenzini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giovanni Peretto
- Department of Arrhythmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| | - Paolo Della Bella
- Department of Arrhythmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Maurizio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Laboratory of Clinical Molecular Biology and Cytogenetics, IRCCS San Raffaele Hospital, Milan, Italy; Genomic Unit for the Diagnosis of Human Pathologies, Division of Genetics and Cellular Biology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Sara Benedetti
- Laboratory of Clinical Molecular Biology and Cytogenetics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Simone Sala
- Department of Arrhythmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Di Resta
- Vita-Salute San Raffaele University, Milan, Italy; Genomic Unit for the Diagnosis of Human Pathologies, Division of Genetics and Cellular Biology, IRCCS San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
61
|
Rodriguez AP, Badiye A, Lambrakos LK, Ghodsizad A, Myerburg RJ, Goldberger JJ. Refractory ventricular tachycardia storm associated with severe hypokalemia in Fanconi syndrome. HeartRhythm Case Rep 2019; 5:374-378. [PMID: 31341780 PMCID: PMC6630180 DOI: 10.1016/j.hrcr.2019.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Alex P Rodriguez
- Cardiovascular Division, Miller School of Medicine, University of Miami, Miami, Florida
| | - Amit Badiye
- Cardiovascular Division, Miller School of Medicine, University of Miami, Miami, Florida
| | - Litsa K Lambrakos
- Cardiovascular Division, Miller School of Medicine, University of Miami, Miami, Florida
| | - Ali Ghodsizad
- Cardiothoracic Surgery Division, Miller School of Medicine, University of Miami, Miami, Florida
| | - Robert J Myerburg
- Cardiovascular Division, Miller School of Medicine, University of Miami, Miami, Florida
| | - Jeffrey J Goldberger
- Cardiovascular Division, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
62
|
Da'as SI, Thanassoulas A, Calver BL, Beck K, Salem R, Saleh A, Kontogianni I, Al-Maraghi A, Nasrallah GK, Safieh-Garabedian B, Toft E, Nounesis G, Lai FA, Nomikos M. Arrhythmogenic calmodulin E105A mutation alters cardiac RyR2 regulation leading to cardiac dysfunction in zebrafish. Ann N Y Acad Sci 2019; 1448:19-29. [PMID: 30937913 DOI: 10.1111/nyas.14033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 01/03/2023]
Abstract
Calmodulin (CaM) is a universal calcium (Ca2+ )-binding messenger that regulates many vital cellular events. In cardiac muscle, CaM associates with ryanodine receptor 2 (RyR2) and regulates excitation-contraction coupling. Mutations in human genes CALM1, CALM2, and CALM3 have been associated with life-threatening heart disorders, such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia. A novel de novo LQTS-associated missense CaM mutation (E105A) was recently identified in a 6-year-old boy, who experienced an aborted first episode of cardiac arrest. Herein, we report the first molecular characterization of the CaM E105A mutation. Expression of the CaM E105A mutant in zebrafish embryos resulted in cardiac arrhythmia and increased heart rate, suggestive of ventricular tachycardia. In vitro biophysical and biochemical analysis revealed that E105A confers a deleterious effect on protein stability and a reduced Ca2+ -binding affinity due to loss of cooperativity. Finally, the CaM E105A mutation resulted in reduced CaM-RyR2 interaction and defective modulation of ryanodine binding. Our findings suggest that the CaM E105A mutation dysregulates normal cardiac function by a complex mechanism involving alterations in both CaM-Ca2+ and CaM-RyR2 interactions.
Collapse
Affiliation(s)
- Sahar I Da'as
- Translational Medicine, Sidra Medicine, Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Brian L Calver
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Konrad Beck
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Rola Salem
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Alaaeldin Saleh
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Iris Kontogianni
- National Center for Scientific Research "Demokritos,", Aghia Paraskevi, Greece
| | - Ali Al-Maraghi
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Biomedical Research Center, Qatar University, Doha, Qatar.,Department of Biomedical Sciences, College of Health Science, Qatar University, Doha, Qatar
| | | | - Egon Toft
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - George Nounesis
- National Center for Scientific Research "Demokritos,", Aghia Paraskevi, Greece
| | - F Anthony Lai
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK.,College of Medicine, Member of QU Health, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Michail Nomikos
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
63
|
Xie L, Hou C, Jiang X, Zhao J, Li Y, Xiao T. A compound heterozygosity of Tecrl gene confirmed in a catecholaminergic polymorphic ventricular tachycardia family. Eur J Med Genet 2019; 62:103631. [PMID: 30790670 DOI: 10.1016/j.ejmg.2019.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 11/27/2022]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is one of the most common causes of sudden cardiac death (SCD) during childhood and in adolescence. Trans-2, 3-enoyl-CoA reductase-like (Tecrl) gene mutations (Arg196Gln and c.331+1G > A splice site mutation) were first reported in CPVT. Tecrl homozygous c.331+1G > A splice site mutation in iPSCs revealed a definite correlation between Tecrl and Ca2+ transport in cardiomyocytes. However, no other researchers have confirmed Tecrl mutations in CPVT with literature review. In this study, a case of compound heterozygosity in the Tecrl gene (Arg196Gln and c.918+3T > G splice site mutation) was first identified in a 13-year-old boy with CPVT by whole-exome sequencing (WES) and confirmed by Sanger sequence. Support vector machine and neural network analysis predicted that Arg196Gln mutation could decrease the stability of Tecrl structure, the confidence scores were -0.8929 and -0.9930. A STRUM server also confirmed that Arg196Gln mutation may decrease the binding capacity of the substrate and cause an amino acid substitution immediately upstream of the 3-oxo-5-alpha steroid 4-dehydrogenase domain. According to the "human splicing finder" indication and Alamut Visual Splicing Prediction, the c.918 + 3T > G mutation could influence Tecrl variable splicing. Thus, we confirmed that Tecrl as a new gene which is associated with CPVT.
Collapse
Affiliation(s)
- Lijian Xie
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China
| | - Cuilan Hou
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China
| | - Xunwei Jiang
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China
| | - Jian Zhao
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China
| | - Yun Li
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China
| | - Tingting Xiao
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, No. 355 Luding Road, Shanghai, 200062, China.
| |
Collapse
|
64
|
Crotti L, Ghidoni A, Dagradi F. Genetics of Adult and Fetal Forms of Long QT Syndrome. GENETIC CAUSES OF CARDIAC DISEASE 2019. [DOI: 10.1007/978-3-030-27371-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
65
|
|
66
|
Skinner JR, Winbo A, Abrams D, Vohra J, Wilde AA. Channelopathies That Lead to Sudden Cardiac Death: Clinical and Genetic Aspects. Heart Lung Circ 2019; 28:22-30. [DOI: 10.1016/j.hlc.2018.09.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 09/20/2018] [Accepted: 09/23/2018] [Indexed: 12/19/2022]
|
67
|
London B. Catecholaminergic Polymorphic (Right) Ventricular Tachycardia? JACC Clin Electrophysiol 2019; 5:128-130. [DOI: 10.1016/j.jacep.2018.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 10/27/2022]
|
68
|
Huang L, Chen Y, Lin Y, Tam POS, Cheng Y, Shi Y, Gong B, Lu F, Yang J, Wang H, Yin Y, Cao Y, Jiang D, Zhong L, Xue B, Wang J, Hao F, Lee DY, Pang CP, Sun X, Yang Z. Genome-wide analysis identified 17 new loci influencing intraocular pressure in Chinese population. SCIENCE CHINA-LIFE SCIENCES 2018; 62:153-164. [PMID: 30591961 DOI: 10.1007/s11427-018-9430-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022]
Abstract
Intraocular pressure (IOP) is a major risk factor for glaucoma. Genetic determinants of intraocular pressure can provide critical insights into the genetic architecture of glaucoma and, as a result, open new avenues for therapeutic intervention. We performed a genome-wide association study and replication analysis of 8,552 Chinese participants. In the genome-wide association study, we identified 51 loci that surpassed the significance of P<9×10-7, and we formally replicated these loci. A combined discovery and replication meta-analysis identified 21 genome-wide loci that surpassed the genome-wide significance of P<5×10-8, including 4 previously reported loci: rs145063132 (7p21.2, ETV1/DGKB), rs548030386 (7q31.2, ST7 near CAV1/CAV2), rs7047871 (9p24.2, GLIS3), and rs2472494 (9q31.1, ABCA1/SLC44A1). Of the 17 newly identified loci, five were reported to have ocular related phenotypes: PTCH2 (rs7525308 in 1p34.1), LRIF1/DRAM2 (rs1282146 in 1p13.3), COLEC11 (rs201143466 in 2p25.3), SPTBN1 (rs4514918 in 2p16.2), and CRK (rs11078446 in 17p13.3). The genetic loci identified in this study not only increase our understanding of the genes involved in intraocular pressure but also provide important genetic markers to improve future genetic screening and drug discovery for intraocular pressure disorders.
Collapse
Affiliation(s)
- Lulin Huang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610072, China.
| | - Yuhong Chen
- Department of Ophthalmology Visual Science, Eye and ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, 200031, China
| | - Ying Lin
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610072, China
| | - Pancy O S Tam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yilian Cheng
- Department of Ophthalmology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yi Shi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Bo Gong
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Fang Lu
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jialiang Yang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Haixin Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Yin
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yong Cao
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Dan Jiang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ling Zhong
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Bai Xue
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jing Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Fang Hao
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Dean-Yao Lee
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Chi-Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xinghuai Sun
- Department of Ophthalmology Visual Science, Eye and ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, 200031, China
| | - Zhenglin Yang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610072, China.
| |
Collapse
|
69
|
Pancaroglu R, Van Petegem F. Calcium Channelopathies: Structural Insights into Disorders of the Muscle Excitation–Contraction Complex. Annu Rev Genet 2018; 52:373-396. [DOI: 10.1146/annurev-genet-120417-031311] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ion channels are membrane proteins responsible for the passage of ions down their electrochemical gradients and across biological membranes. In this, they generate and shape action potentials and provide secondary messengers for various signaling pathways. They are often part of larger complexes containing auxiliary subunits and regulatory proteins. Channelopathies arise from mutations in the genes encoding ion channels or their associated proteins. Recent advances in cryo-electron microscopy have resulted in an explosion of ion channel structures in multiple states, generating a wealth of new information on channelopathies. Disease-associated mutations fall into different categories, interfering with ion permeation, protein folding, voltage sensing, ligand and protein binding, and allosteric modulation of channel gating. Prime examples of these are Ca2+-selective channels expressed in myocytes, for which multiple structures in distinct conformational states have recently been uncovered. We discuss the latest insights into these calcium channelopathies from a structural viewpoint.
Collapse
Affiliation(s)
- Raika Pancaroglu
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
70
|
Roston TM, Haji-Ghassemi O, LaPage MJ, Batra AS, Bar-Cohen Y, Anderson C, Lau YR, Maginot K, Gebauer RA, Etheridge SP, Potts JE, Van Petegem F, Sanatani S. Catecholaminergic polymorphic ventricular tachycardia patients with multiple genetic variants in the PACES CPVT Registry. PLoS One 2018; 13:e0205925. [PMID: 30403697 PMCID: PMC6221297 DOI: 10.1371/journal.pone.0205925] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/03/2018] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Catecholaminergic polymorphic ventricular tachycardia (CPVT) is often a life-threatening arrhythmia disorder with variable penetrance and expressivity. Little is known about the incidence or outcomes of CPVT patients with ≥2 variants. METHODS The phenotypes, genotypes and outcomes of patients in the Pediatric and Congenital Electrophysiology Society CPVT Registry with ≥2 variants in genes linked to CPVT were ascertained. The American College of Medical Genetics & Genomics (ACMG) criteria and structural mapping were used to predict the pathogenicity of variants (3D model of pig RyR2 in open-state). RESULTS Among 237 CPVT subjects, 193 (81%) had genetic testing. Fifteen patients (8%) with a median age of 9 years (IQR 5-12) had ≥2 variants. Sudden cardiac arrest occurred in 11 children (73%), although none died during a median follow-up of 4.3 years (IQR 2.5-6.1). Thirteen patients (80%) had at least two RYR2 variants, while the remaining two patients had RYR2 variants plus variants in other CPVT-linked genes. Among all variants identified, re-classification of the commercial laboratory interpretation using ACMG criteria led to the upgrade from variant of unknown significance (VUS) to pathogenic/likely pathogenic (P/LP) for 5 variants, and downgrade from P/LP to VUS for 6 variants. For RYR2 variants, 3D mapping using the RyR2 model suggested that 2 VUS by ACMG criteria were P/LP, while 2 variants were downgraded to likely benign. CONCLUSIONS This severely affected cohort demonstrates that a minority of CPVT cases are related to ≥2 variants, which may have implications on family-based genetic counselling. While multi-variant CPVT patients were at high-risk for sudden cardiac arrest, there are insufficient data to conclude that this genetic phenomenon has prognostic implications at present. Further research is needed to determine the significance and generalizability of this observation. This study also shows that a rigorous approach to variant re-classification using the ACMG criteria and 3D mapping is important in reaching an accurate diagnosis, especially in the multi-variant population.
Collapse
Affiliation(s)
- Thomas M. Roston
- Departments of Medicine, Pediatrics, and Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Omid Haji-Ghassemi
- Departments of Medicine, Pediatrics, and Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Martin J. LaPage
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States of America
| | - Anjan S. Batra
- Department of Pediatrics, University of California at Irvine Medical Center, Irvine, CA, United States of America
| | - Yaniv Bar-Cohen
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States of America
| | - Chris Anderson
- Providence Sacred Heart Children’s Hospital, Spokane, WA, United States of America
| | - Yung R. Lau
- Division of Pediatric Cardiology, University of Alabama at Birmingham, Birmingham, AB, United States of America
| | - Kathleen Maginot
- Department of Pediatrics, University of Wisconsin School of Medicine & Public Health, Madison, WI, United States of America
| | - Roman A. Gebauer
- Department of Pediatric Cardiology, University of Leipzig, Leipzig, Germany
| | - Susan P. Etheridge
- Department of Pediatrics, University of Utah, and Primary Children’s Hospital, Salt Lake City, UT, United States of America
| | - James E. Potts
- Departments of Medicine, Pediatrics, and Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Departments of Medicine, Pediatrics, and Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Shubhayan Sanatani
- Departments of Medicine, Pediatrics, and Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
71
|
Plakophilin-2 Truncation Variants in Patients Clinically Diagnosed With Catecholaminergic Polymorphic Ventricular Tachycardia and Decedents With Exercise-Associated Autopsy Negative Sudden Unexplained Death in the Young. JACC Clin Electrophysiol 2018; 5:120-127. [PMID: 30678776 DOI: 10.1016/j.jacep.2018.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/25/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES This study determined if radical plakophilin-2 (PKP2) variants might underlie some cases of clinically diagnosed catecholaminergic polymorphic ventricular tachycardia (CPVT) and exercise-associated, autopsy-negative sudden unexplained death in the young (SUDY). BACKGROUND Pathogenic variants in PKP2 cause arrhythmogenic right ventricular cardiomyopathy (ARVC). Recently, a cardiomyocyte-specific PKP2 knockout mouse model revealed that loss of PKP2 markedly reduced expression of genes critical in intracellular calcium handling. The mice with structurally normal hearts exhibited isoproterenol-triggered polymorphic ventricular arrhythmias that mimicked CPVT. METHODS A PKP2 gene mutational analysis was performed on DNA from 18 unrelated patients (9 males; average age at diagnosis: 19.6 ± 12.8 years) clinically diagnosed with CPVT but who were RYR2-, CASQ2-, KCNJ2-, and TRDN-negative, and 19 decedents with SUDY during exercise (13 males; average age at death: 14 ± 3 years). Only radical (i.e., frame-shift, canonical splice site, or nonsense) variants with a minor allele frequency of ≤0.00005 in the genome aggregation database (gnomAD) were considered pathogenic. RESULTS Radical PKP2 variants were identified in 5 of 18 (27.7%) CPVT patients and 1 of 19 (5.3%) exercise-related SUDY cases compared with 96 of 138,632 (0.069%) individuals in gnomAD (p = 3.1 × 10-13). Cardiac imaging or autopsy demonstrated a structurally normal heart in all patients at the time of their CPVT diagnosis or sudden death. CONCLUSIONS Our data suggested that the progression of the PKP2-dependent electropathy can be independent of structural perturbations and can precipitate exercise-associated sudden cardiac arrest or sudden cardiac death before the presence of overt cardiomyopathy, which clinically mimics CPVT, similar to the PKP2 knockout mouse model. Thus, CPVT and SUDY genetic test panels should now include PKP2.
Collapse
|
72
|
Hylind RJ, Chandler SF, Skinner JR, Abrams DJ. Genetic Testing for Inherited Cardiac Arrhythmias: Current State-of-the-Art and Future Avenues. J Innov Card Rhythm Manag 2018; 9:3406-3416. [PMID: 32494476 PMCID: PMC7252877 DOI: 10.19102/icrm.2018.091102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/14/2018] [Indexed: 12/24/2022] Open
Abstract
The seminal discovery that sequence variation in genes encoding cardiac ion channels was behind the inherited cardiac arrhythmic syndromes has led to major advances in understanding the functional biological mechanisms of cardiomyocyte depolarization and repolarization. The cost and speed with which these genes can now be sequenced have allowed for genetic testing to become a major component of clinical care and have led to important ramifications, yet interpretation of specific variants needs to be performed within the context of the clinical findings in the proband and extended family. As technology continues to advance, the promise of therapeutic manipulation of certain genetic pathways grows ever more real.
Collapse
Affiliation(s)
- Robyn J. Hylind
- Inherited Cardiac Arrhythmia Program, Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie F. Chandler
- Inherited Cardiac Arrhythmia Program, Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jonathan R. Skinner
- Green Lane Paediatric and Congenital Cardiac Services, Starship Children’s Hospital, Auckland, New Zealand
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
| | - Dominic J. Abrams
- Inherited Cardiac Arrhythmia Program, Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
73
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018. [PMID: 30425651 DOI: 10.3389/fphys.2018.01517, 10.3389/fpls.2018.01517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
74
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
75
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517,+10.3389/fpls.2018.01517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States,*Correspondence: Dmitry Terentyev,
| |
Collapse
|
76
|
Gray B, Behr ER. New Insights Into the Genetic Basis of Inherited Arrhythmia Syndromes. ACTA ACUST UNITED AC 2018; 9:569-577. [PMID: 27998945 DOI: 10.1161/circgenetics.116.001571] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Belinda Gray
- From the Department of Cardiology, Royal Prince Alfred Hospital, New South Wales, Australia (B.G.); Sydney Medical School, University of Sydney, Australia (B.G.), Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, New South Wales, Australia (B.G.); Cardiology Clinical Academic Group, St George's University of London, United Kingdom (E.R.B.); and St George's University Hospitals NHS Foundation Trust, London, United Kingdom (E.R.B.)
| | - Elijah R Behr
- From the Department of Cardiology, Royal Prince Alfred Hospital, New South Wales, Australia (B.G.); Sydney Medical School, University of Sydney, Australia (B.G.), Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, New South Wales, Australia (B.G.); Cardiology Clinical Academic Group, St George's University of London, United Kingdom (E.R.B.); and St George's University Hospitals NHS Foundation Trust, London, United Kingdom (E.R.B.).
| |
Collapse
|
77
|
Ozawa J, Ohno S, Fujii Y, Makiyama T, Suzuki H, Saitoh A, Horie M. Differential Diagnosis Between Catecholaminergic Polymorphic Ventricular Tachycardia and Long QT Syndrome Type 1 ― Modified Schwartz Score ―. Circ J 2018; 82:2269-2276. [DOI: 10.1253/circj.cj-17-1032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Junichi Ozawa
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences
| | - Seiko Ohno
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science
- Center for Epidemiologic Research in Asia, Shiga University of Medical Science
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center
| | - Yusuke Fujii
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science
| | - Takeru Makiyama
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | - Hiroshi Suzuki
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences
| | - Akihiko Saitoh
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science
| |
Collapse
|
78
|
O’Callaghan BM, Hancox JC, Stuart AG, Armstrong C, Williams MM, Hills A, Pearce H, Dent CL, Gable M, Walsh MA. A unique triadin exon deletion causing a null phenotype. HeartRhythm Case Rep 2018; 4:514-518. [PMID: 30479949 PMCID: PMC6241331 DOI: 10.1016/j.hrcr.2018.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Jules C. Hancox
- School of Physiology and Pharmacology, Cardiovascular Research Laboratories, University of Bristol, Bristol, United Kingdom
| | - Alan G. Stuart
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| | | | - Maggie M. Williams
- Genomic Diagnostics Laboratory, University Hospital Bristol, Bristol, United Kingdom
| | - Alison Hills
- Genomic Diagnostics Laboratory, University Hospital Bristol, Bristol, United Kingdom
| | - Hazel Pearce
- Genomic Diagnostics Laboratory, University Hospital Bristol, Bristol, United Kingdom
| | - Carolyn L. Dent
- Genomic Diagnostics Laboratory, University Hospital Bristol, Bristol, United Kingdom
| | - Mary Gable
- Genomic Diagnostics Laboratory, University Hospital Bristol, Bristol, United Kingdom
| | - Mark A. Walsh
- Bristol Royal Hospital for Children, Bristol, United Kingdom
- Address reprint requests and correspondence: Dr Mark A. Walsh, Our Lady's Children's Hospital, Crumlin, Cooley Road, Drimnagh, Dublin, Ireland.
| |
Collapse
|
79
|
Bruyneel AAN, McKeithan WL, Feyen DAM, Mercola M. Using iPSC Models to Probe Regulation of Cardiac Ion Channel Function. Curr Cardiol Rep 2018; 20:57. [DOI: 10.1007/s11886-018-1000-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
80
|
Schiaffino S. Knockout of human muscle genes revealed by large scale whole-exome studies. Mol Genet Metab 2018; 123:411-415. [PMID: 29452748 DOI: 10.1016/j.ymgme.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
Large scale whole-exome sequence studies have revealed that a number of individuals from different populations have predicted loss-of-function of different genes due to nonsense, frameshift, or canonical splice-site mutations. Surprisingly, many of these mutations do not apparently show the deleterious phenotypic consequences expected from gene knockout. These homozygous null mutations, when confirmed, can provide insight into human gene function and suggest novel approaches to correct gene dysfunction, as the lack of the expected disease phenotype may reflect the existence of modifier genes that reveal potential therapeutic targets. Human knockouts complement the information derived from mouse knockouts, which are not always good models of human disease. We have examined human knockout datasets searching for genes expressed exclusively or predominantly in striated muscle. A number of well-known muscle genes was found in one or more datasets, including genes coding for sarcomeric myosins, components of the sarcomeric cytoskeleton, sarcoplasmic reticulum and plasma membrane, and enzymes involved in muscle metabolism. The surprising absence of phenotype in some of these human knockouts is critically discussed, focusing on the comparison with the corresponding mouse knockouts.
Collapse
|
81
|
Abstract
During the complex series of events leading to muscle contraction, the initial electric signal coming from motor neurons is transformed into an increase in calcium concentration that triggers sliding of myofibrils. This process, referred to as excitation-contraction coupling, is reliant upon the calcium-release complex, which is restricted spatially to a sub-compartment of muscle cells ("the triad") and regulated precisely. Any dysfunction in the calcium-release complex leads to muscle impairment and myopathy. Various causes can lead to alterations in excitation-contraction coupling and to muscle diseases. The latter are reviewed and classified into four categories: (i) mutation in a protein of the calcium-release complex; (ii) alteration in triad structure; (iii) modification of regulation of channels; (iv) modification in calcium stores within the muscle. Current knowledge of the pathophysiologic mechanisms in each category is described and discussed.
Collapse
Affiliation(s)
- Isabelle Marty
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000 Grenoble, France.,INSERM, U1216, F-38000 Grenoble, France
| | - Julien Fauré
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000 Grenoble, France.,INSERM, U1216, F-38000 Grenoble, France.,CHU de Grenoble, F-38000 Grenoble, France
| |
Collapse
|
82
|
Pathogenic mechanism of a catecholaminergic polymorphic ventricular tachycardia causing-mutation in cardiac calcium release channel RyR2. J Mol Cell Cardiol 2018; 117:26-35. [PMID: 29477366 DOI: 10.1016/j.yjmcc.2018.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 12/27/2022]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a condition that is characterized by an abnormal heart rhythm in response to physical or emotional stress. The majority CPVT patients carry mutations in the RYR2 gene that encodes the calcium release channel/ryanodine receptor (RyR2) in cardiomyocytes. The pathogenic mechanisms that account for the clinical phenotypes of CPVT are still elusive. We have identified a de novo mutation, A165D, from a CPVT patient. We found that CPVT phenotypes are recapitulated in A165D knock-in mice. The mutant RyR2 channels enhanced sarcoplasmic reticulum Ca2+ release, triggered delayed afterdepolarization in cardiomyocytes. Structural analysis revealed that the A165D mutation is located in a loop that is involved in inter-subunit interactions in the RyR2 tetrameric structure, it disrupted conformational stability of the RyR2, which favored a closed-to-open state transition, resulting in a leaky channel. The loop also harbors several other CPVT mutations, which suggests a common pathogenic molecular mechanism of CPVT-causing mutations. Our data illustrated disease-relevant functional defects and provide a deeper mechanistic understanding of a life-threatening cardiac arrhythmia.
Collapse
|
83
|
Ion Channel Disorders and Sudden Cardiac Death. Int J Mol Sci 2018; 19:ijms19030692. [PMID: 29495624 PMCID: PMC5877553 DOI: 10.3390/ijms19030692] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/19/2022] Open
Abstract
Long QT syndrome, short QT syndrome, Brugada syndrome and catecholaminergic polymorphic ventricular tachycardia are inherited primary electrical disorders that predispose to sudden cardiac death in the absence of structural heart disease. Also known as cardiac channelopathies, primary electrical disorders respond to mutations in genes encoding cardiac ion channels and/or their regulatory proteins, which result in modifications in the cardiac action potential or in the intracellular calcium handling that lead to electrical instability and life-threatening ventricular arrhythmias. These disorders may have low penetrance and expressivity, making clinical diagnosis often challenging. However, because sudden cardiac death might be the first presenting symptom of the disease, early diagnosis becomes essential. Genetic testing might be helpful in this regard, providing a definite diagnosis in some patients. Yet important limitations still exist, with a significant proportion of patients remaining with no causative mutation identifiable after genetic testing. This review aims to provide the latest knowledge on the genetic basis of cardiac channelopathies and discuss the role of the affected proteins in the pathophysiology of each one of these diseases.
Collapse
|
84
|
Abstract
PURPOSE OF REVIEW Cardiomyopathies due to genetic mutations are a heterogeneous group of disorders that comprise diseases of contractility, myocardial relaxation, and arrhythmias. Our goal here is to discuss a limited list of genetically inherited cardiomyopathies and the specific therapeutic strategies used to treat them. RECENT FINDINGS Research into the molecular pathophysiology of the development of these cardiomyopathies is leading to the development of novel treatment approaches. Therapies targeting these specific mutations with gene therapy vectors are on the horizon, while other therapies which indirectly affect the physiologic derangements of the mutations are currently being studied and used clinically. Many of these therapies are older medications being given new roles such as mexiletine for Brugada syndrome and diflunisal for transthyretin amyloid cardiomyopathy. A newer targeted therapy, the inhibitor of myosin ATPase MYK-461, has been shown to suppress the development of ventricular hypertrophy, fibrosis, and myocyte disarray and is being studied as a potential therapy in patients with hypertrophic cardiomyopathy. While this field is too large to be completely contained in a single review, we present a large cross section of recent developments in the field of therapeutics for inherited cardiomyopathies. New therapies are on the horizon, and their development will likely result in improved outcomes for patients inflicted by these conditions.
Collapse
Affiliation(s)
- Kenneth Varian
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA. .,Center for Clinical Genomics, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
85
|
Coll M, Pérez-Serra A, Mates J, Del Olmo B, Puigmulé M, Fernandez-Falgueras A, Iglesias A, Picó F, Lopez L, Brugada R, Campuzano O. Incomplete Penetrance and Variable Expressivity: Hallmarks in Channelopathies Associated with Sudden Cardiac Death. BIOLOGY 2017; 7:biology7010003. [PMID: 29278359 PMCID: PMC5872029 DOI: 10.3390/biology7010003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
Abstract
Sudden cardiac death is defined as an unexpected decease of cardiac origin. In individuals under 35 years old, most of these deaths are due to familial arrhythmogenic syndromes of genetic origin, also known as channelopathies. These familial cardiac syndromes commonly follow an autosomal dominant pattern of inheritance. Diagnosis, however, can be difficult, mainly due to incomplete penetrance and variable expressivity, which are hallmarks in these syndromes. The clinical manifestation of these diseases can range from asymptomatic to syncope but sudden death can sometimes be the first symptom of disease. Early identification of at-risk individuals is crucial to prevent a lethal episode. In this review, we will focus on the genetic basis of channelopathies and the effect of genetic and non-genetic modifiers on their phenotypes.
Collapse
Affiliation(s)
- Monica Coll
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Alexandra Pérez-Serra
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Jesus Mates
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Bernat Del Olmo
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Marta Puigmulé
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain.
| | | | - Anna Iglesias
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Ferran Picó
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Laura Lopez
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
| | - Ramon Brugada
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain.
- Cardiology Service, Hospital Josep Trueta, 17003 Girona, Spain.
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, 17190 Salt, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain.
| |
Collapse
|
86
|
Hancox JC, James AF, Walsh MA, Stuart AG. Triadin mutations - a cause of ventricular arrhythmias in children and young adults. JOURNAL OF CONGENITAL CARDIOLOGY 2017. [DOI: 10.1186/s40949-017-0011-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
87
|
Liu Y, Wei J, Wong King Yuen SM, Sun B, Tang Y, Wang R, Van Petegem F, Chen SRW. CPVT-associated cardiac ryanodine receptor mutation G357S with reduced penetrance impairs Ca2+ release termination and diminishes protein expression. PLoS One 2017; 12:e0184177. [PMID: 28961276 PMCID: PMC5621672 DOI: 10.1371/journal.pone.0184177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/18/2017] [Indexed: 11/18/2022] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is one of the most lethal inherited cardiac arrhythmias mostly linked to cardiac ryanodine receptor (RyR2) mutations with high disease penetrance. Interestingly, a novel RyR2 mutation G357S discovered in a large family of more than 1400 individuals has reduced penetrance. The molecular basis for the incomplete disease penetrance in this family is unknown. To gain insights into the variable disease expression in this family, we determined the impact of the G357S mutation on RyR2 function and expression. We assessed spontaneous Ca2+ release in HEK293 cells expressing RyR2 wildtype and the G357S mutant during store Ca2+ overload, also known as store overload induced Ca2+ release (SOICR). We found that the G357S mutation reduced the percentage of RyR2-expressing cells that showed SOICR. However, in cells that displayed SOICR, G357S reduced the thresholds for the activation and termination of SOICR. Furthermore, G357S decreased the thermal stability of the N-terminal domain of RyR2, and markedly reduced the protein expression of the full-length RyR2. On the other hand, the G357S mutation did not alter the Ca2+ activation of [3H]ryanodine binding or the Ca2+ induced release of Ca2+ from the intracellular stores in HEK293 cells. These data indicate that the CPVT-associated G357S mutation enhances the arrhythmogenic SOICR and reduces RyR2 protein expression, which may be attributable to the incomplete penetrance of CPVT in this family.
Collapse
Affiliation(s)
- Yingjie Liu
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jinhong Wei
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Siobhan M Wong King Yuen
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, Canada
| | - Bo Sun
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Yijun Tang
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
88
|
Gene Expression Networks in the Murine Pulmonary Myocardium Provide Insight into the Pathobiology of Atrial Fibrillation. G3-GENES GENOMES GENETICS 2017; 7:2999-3017. [PMID: 28720711 PMCID: PMC5592927 DOI: 10.1534/g3.117.044651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pulmonary myocardium is a muscular coat surrounding the pulmonary and caval veins. Although its definitive physiological function is unknown, it may have a pathological role as the source of ectopic beats initiating atrial fibrillation. How the pulmonary myocardium gains pacemaker function is not clearly defined, although recent evidence indicates that changed transcriptional gene expression networks are at fault. The gene expression profile of this distinct cell type in situ was examined to investigate underlying molecular events that might contribute to atrial fibrillation. Via systems genetics, a whole-lung transcriptome data set from the BXD recombinant inbred mouse resource was analyzed, uncovering a pulmonary cardiomyocyte gene network of 24 transcripts, coordinately regulated by chromosome 1 and 2 loci. Promoter enrichment analysis and interrogation of publicly available ChIP-seq data suggested that transcription of this gene network may be regulated by the concerted activity of NKX2-5, serum response factor, myocyte enhancer factor 2, and also, at a post-transcriptional level, by RNA binding protein motif 20. Gene ontology terms indicate that this gene network overlaps with molecular markers of the stressed heart. Therefore, we propose that perturbed regulation of this gene network might lead to altered calcium handling, myocyte growth, and contractile force contributing to the aberrant electrophysiological properties observed in atrial fibrillation. We reveal novel molecular interactions and pathways representing possible therapeutic targets for atrial fibrillation. In addition, we highlight the utility of recombinant inbred mouse resources in detecting and characterizing gene expression networks of relatively small populations of cells that have a pathological significance.
Collapse
|
89
|
Györke S, Belevych AE, Liu B, Kubasov IV, Carnes CA, Radwański PB. The role of luminal Ca regulation in Ca signaling refractoriness and cardiac arrhythmogenesis. J Gen Physiol 2017; 149:877-888. [PMID: 28798279 PMCID: PMC5583712 DOI: 10.1085/jgp.201711808] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/19/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023] Open
Abstract
Györke et al. discuss the role of sarcoplasmic reticulum Ca2+ in cardiac refractoriness and pathological implications.
Collapse
Affiliation(s)
- Sándor Györke
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH .,Davis Heart and Lung Research Institute, Columbus, OH
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Bin Liu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Igor V Kubasov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Cynthia A Carnes
- College of Pharmacy, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Przemysław B Radwański
- College of Pharmacy, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| |
Collapse
|
90
|
Abstract
There has been a significant progress in our understanding of the molecular mechanisms by which calcium (Ca2+) ions mediate various types of cardiac arrhythmias. A growing list of inherited gene defects can cause potentially lethal cardiac arrhythmia syndromes, including catecholaminergic polymorphic ventricular tachycardia, congenital long QT syndrome, and hypertrophic cardiomyopathy. In addition, acquired deficits of multiple Ca2+-handling proteins can contribute to the pathogenesis of arrhythmias in patients with various types of heart disease. In this review article, we will first review the key role of Ca2+ in normal cardiac function-in particular, excitation-contraction coupling and normal electric rhythms. The functional involvement of Ca2+ in distinct arrhythmia mechanisms will be discussed, followed by various inherited arrhythmia syndromes caused by mutations in Ca2+-handling proteins. Finally, we will discuss how changes in the expression of regulation of Ca2+ channels and transporters can cause acquired arrhythmias, and how these mechanisms might be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Andrew P Landstrom
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Dobromir Dobrev
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Xander H T Wehrens
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.).
| |
Collapse
|
91
|
Abstract
Catecholaminergic polymorphic ventricular tachycardia is a rare cause of exercise-induced arrhythmia and sudden cardiac death in the pediatric patient. This arrhythmia is difficult to diagnose in the emergency department, given the range of presentations; thus, a familiarity with and high index of suspicion for this pathology are crucial. Furthermore, recognition of the characteristic electrocardiogram findings and knowledge of the management of the symptomatic patient are necessary, given the risk of arrhythmia recurrence and cardiac arrest. In this review, we discuss the presentation, differential diagnosis, and management of catecholaminergic polymorphic ventricular tachycardia for the emergency care provider.
Collapse
|
92
|
Wang S, Li L, Tao R, Gao Y. Ion channelopathies associated genetic variants as the culprit for sudden unexplained death. Forensic Sci Int 2017; 275:128-137. [PMID: 28363160 DOI: 10.1016/j.forsciint.2017.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/23/2017] [Accepted: 03/13/2017] [Indexed: 11/29/2022]
Abstract
Forensic identification of sudden unexplained death (SUD) has always been a ticklish issue because it used to be defined as sudden death without a conclusive diagnosis after autopsy. However, benefiting from the developments in genome research, a growing body of evidence points to the importance of ion channelopathies associated genetic variants in the pathogenesis of SUD. Genetic diagnosis of the deceased is also a new trend in epidemiological studies, for it enables the undertaking for preventive approach in individuals with high risks. In this review, we briefly discuss the molecular structure of ion channels and the role of genetic variants in regulating their functions as well as the diverse mechanisms underlying the ion channelopathies at gene level.
Collapse
Affiliation(s)
- Shouyu Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Lijuan Li
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Ruiyang Tao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Yuzhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
93
|
Engel AG, Redhage KR, Tester DJ, Ackerman MJ, Selcen D. Congenital myopathy associated with the triadin knockout syndrome. Neurology 2017; 88:1153-1156. [PMID: 28202702 DOI: 10.1212/wnl.0000000000003745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/27/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Triadin is a component of the calcium release complex of cardiac and skeletal muscle. Our objective was to analyze the skeletal muscle phenotype of the triadin knockout syndrome. METHODS We performed clinical evaluation, analyzed morphologic features by light and electron microscopy, and immunolocalized triadin in skeletal muscle. RESULTS A 6-year-old boy with lifelong muscle weakness had a triadin knockout syndrome caused by compound heterozygous null mutations in triadin. Light microscopy of a deltoid muscle specimen shows multiple small abnormal spaces in all muscle fibers. Triadin immunoreactivity is absent from type 1 fibers and barely detectable in type 2 fibers. Electron microscopy reveals focally distributed dilation and degeneration of the lateral cisterns of the sarcoplasmic reticulum and loss of the triadin anchors from the preserved lateral cisterns. CONCLUSIONS Absence of triadin in humans can result in a congenital myopathy associated with profound pathologic alterations in components of the sarcoplasmic reticulum. Why only some triadin-deficient patients develop a skeletal muscle phenotype remains an unsolved question.
Collapse
Affiliation(s)
- Andrew G Engel
- From the Departments of Neurology and Muscle Research Laboratory (A.G.E., D.S.), Cardiovascular Diseases/Division of Heart Rhythm Services (A.G.E., D.S.), Pediatric and Adolescent Medicine/Division of Pediatric Cardiology (K.R.R., D.J.T., M.J.A.), and Molecular Pharmacology & Experimental Therapeutics/Windland Smith Rice Sudden Death Genomics Laboratory (K.R.R., D.J.T., M.J.A.), Mayo College of Medicine, Rochester, MN.
| | - Keeley R Redhage
- From the Departments of Neurology and Muscle Research Laboratory (A.G.E., D.S.), Cardiovascular Diseases/Division of Heart Rhythm Services (A.G.E., D.S.), Pediatric and Adolescent Medicine/Division of Pediatric Cardiology (K.R.R., D.J.T., M.J.A.), and Molecular Pharmacology & Experimental Therapeutics/Windland Smith Rice Sudden Death Genomics Laboratory (K.R.R., D.J.T., M.J.A.), Mayo College of Medicine, Rochester, MN
| | - David J Tester
- From the Departments of Neurology and Muscle Research Laboratory (A.G.E., D.S.), Cardiovascular Diseases/Division of Heart Rhythm Services (A.G.E., D.S.), Pediatric and Adolescent Medicine/Division of Pediatric Cardiology (K.R.R., D.J.T., M.J.A.), and Molecular Pharmacology & Experimental Therapeutics/Windland Smith Rice Sudden Death Genomics Laboratory (K.R.R., D.J.T., M.J.A.), Mayo College of Medicine, Rochester, MN
| | - Michael J Ackerman
- From the Departments of Neurology and Muscle Research Laboratory (A.G.E., D.S.), Cardiovascular Diseases/Division of Heart Rhythm Services (A.G.E., D.S.), Pediatric and Adolescent Medicine/Division of Pediatric Cardiology (K.R.R., D.J.T., M.J.A.), and Molecular Pharmacology & Experimental Therapeutics/Windland Smith Rice Sudden Death Genomics Laboratory (K.R.R., D.J.T., M.J.A.), Mayo College of Medicine, Rochester, MN
| | - Duygu Selcen
- From the Departments of Neurology and Muscle Research Laboratory (A.G.E., D.S.), Cardiovascular Diseases/Division of Heart Rhythm Services (A.G.E., D.S.), Pediatric and Adolescent Medicine/Division of Pediatric Cardiology (K.R.R., D.J.T., M.J.A.), and Molecular Pharmacology & Experimental Therapeutics/Windland Smith Rice Sudden Death Genomics Laboratory (K.R.R., D.J.T., M.J.A.), Mayo College of Medicine, Rochester, MN
| |
Collapse
|
94
|
Fernández-Falgueras A, Sarquella-Brugada G, Brugada J, Brugada R, Campuzano O. Cardiac Channelopathies and Sudden Death: Recent Clinical and Genetic Advances. BIOLOGY 2017; 6:biology6010007. [PMID: 28146053 PMCID: PMC5372000 DOI: 10.3390/biology6010007] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
Sudden cardiac death poses a unique challenge to clinicians because it may be the only symptom of an inherited heart condition. Indeed, inherited heart diseases can cause sudden cardiac death in older and younger individuals. Two groups of familial diseases are responsible for sudden cardiac death: cardiomyopathies (mainly hypertrophic cardiomyopathy, dilated cardiomyopathy, and arrhythmogenic cardiomyopathy) and channelopathies (mainly long QT syndrome, Brugada syndrome, short QT syndrome, and catecholaminergic polymorphic ventricular tachycardia). This review focuses on cardiac channelopathies, which are characterized by lethal arrhythmias in the structurally normal heart, incomplete penetrance, and variable expressivity. Arrhythmias in these diseases result from pathogenic variants in genes encoding cardiac ion channels or associated proteins. Due to a lack of gross structural changes in the heart, channelopathies are often considered as potential causes of death in otherwise unexplained forensic autopsies. The asymptomatic nature of channelopathies is cause for concern in family members who may be carrying genetic risk factors, making the identification of these genetic factors of significant clinical importance.
Collapse
Affiliation(s)
| | | | - Josep Brugada
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona 08950, Spain.
| | - Ramon Brugada
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
- Familial Cardiomyopathies Unit, Hospital Josep Trueta, Girona 17007, Spain.
| | - Oscar Campuzano
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
| |
Collapse
|
95
|
Walweel K, Molenaar P, Imtiaz MS, Denniss A, Dos Remedios C, van Helden DF, Dulhunty AF, Laver DR, Beard NA. Ryanodine receptor modification and regulation by intracellular Ca 2+ and Mg 2+ in healthy and failing human hearts. J Mol Cell Cardiol 2017; 104:53-62. [PMID: 28131631 DOI: 10.1016/j.yjmcc.2017.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/01/2017] [Accepted: 01/24/2017] [Indexed: 11/30/2022]
Abstract
RATIONALE Heart failure is a multimodal disorder, of which disrupted Ca2+ homeostasis is a hallmark. Central to Ca2+ homeostasis is the major cardiac Ca2+ release channel - the ryanodine receptor (RyR2) - whose activity is influenced by associated proteins, covalent modification and by Ca2+ and Mg2+. That RyR2 is remodelled and its function disturbed in heart failure is well recognized, but poorly understood. OBJECTIVE To assess Ca2+ and Mg2+ regulation of RyR2 from left ventricles of healthy, cystic fibrosis and failing hearts, and to correlate these functional changes with RyR2 modifications and remodelling. METHODS AND RESULTS The function of RyR2 from left ventricular samples was assessed using lipid bilayer single-channel measurements, whilst RyR2 modification and protein:protein interactions were determined using Western Blots and co-immunoprecipitation. In all failing hearts there was an increase in RyR2 activity at end-diastolic cytoplasmic Ca2+ (100nM), a decreased cytoplasmic [Ca2+] required for half maximal activation (Ka) and a decrease in inhibition by cytoplasmic Mg2+. This was accompanied by significant hyperphosphorylation of RyR2 S2808 and S2814, reduced free thiol content and a reduced interaction with FKBP12.0 and FKBP12.6. Either dephosphorylation of RyR2 using PP1 or thiol reduction using DTT eliminated any significant difference in the activity of RyR2 from healthy and failing hearts. We also report a subgroup of RyR2 in failing hearts that were not responsive to regulation by intracellular Ca2+ or Mg2+. CONCLUSION Despite different aetiologies, disrupted RyR2 Ca2+ sensitivity and biochemical modification of the channel are common constituents of failing heart RyR2 and may underlie the pathological disturbances in intracellular Ca2+ signalling.
Collapse
Affiliation(s)
- K Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia
| | - P Molenaar
- Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4000, Northside Clinical School, School of Clinical Medicine, University of Queensland and Critical Care Research Group, The Prince Charles Hospital, Chermside, QLD, 4032, Australia
| | - M S Imtiaz
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
| | - A Denniss
- Health Research Institute, Faculty of Education Science and Mathematics, University of Canberra, Bruce, ACT 2617, Australia
| | - C Dos Remedios
- Bosch Institute, Discipline of Anatomy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - D F van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia
| | - A F Dulhunty
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, 0200, Australia
| | - D R Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia
| | - N A Beard
- Health Research Institute, Faculty of Education Science and Mathematics, University of Canberra, Bruce, ACT 2617, Australia; John Curtin School of Medical Research, Australian National University, Canberra, ACT, 0200, Australia.
| |
Collapse
|
96
|
Abstract
Since the sentinel description of exercise-triggered ventricular arrhythmias in 21 children, our recognition and understanding of catecholaminergic polymorphic ventricular tachycardia has improved substantially. A variety of treatments are now available, but reaching a diagnosis before cardiac arrest remains a challenge. Most cases are related to variants in the gene encoding for ryanodine receptor-2 (RyR2), which mediates calcium-induced calcium release. Up to half of cases remain genetically elusive. The condition is presently incurable, but one basic intervention, the universal administration of β-blockers, has improved survival. In the past, implantable cardioverter-defibrillators (ICDs) were frequently implanted, especially in those with a history of cardiac arrest. Treatment limitations include under-dosing and poor compliance with β-blockers, and potentially lethal ICD-related electrical storm. Newer therapies include flecainide and sympathetic ganglionectomy. Limited data have suggested that genotype may predict phenotype in catecholaminergic polymorphic ventricular tachycardia, including a higher risk of life-threatening cardiac events in subjects with variants in the C-terminus of ryanodine receptor-2 (RyR2). At present, international efforts are underway to better understand this condition through large prospective registries. The recent publication of gene therapy in an animal model of the recessive form of the disease highlights the importance of improving our understanding of the genetic underpinnings of the disease.
Collapse
|
97
|
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a challenging and serious disease with a high incidence of sudden cardiac deaths. Patients with CPVT should not be exposed to physical or emotional exertion that might induce ventricular tachycardia. This article presents a case with CPVT and discusses the clinical features of the disease, its genetic background, and the management of CPVT.
Collapse
|
98
|
Omar A, Zhou M, Berman A, Sorrentino RA, Yar N, Weintraub NL, Kim IM, Lei W, Tang Y. Genomic-based diagnosis of arrhythmia disease in a personalized medicine era. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016; 1:497-504. [PMID: 28944294 PMCID: PMC5606339 DOI: 10.1080/23808993.2016.1264258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Although thousands of potentially disease-causing mutations have been identified in a handful of genes, the genetic heterogeneity has led to diagnostic confusions, stemming directly from the limitations in our arsenal of genetic tools. AREAS COVERED We discuss the genetic basis of cardiac ion channelopathies, the gaps in our knowledge and how Next-generation sequencing technology (NGS) and can be used to bridge them, and how induced pluripotent stem cell (iPSC) derived-cardiomyocytes can be used for drug discovery. EXPERT COMMENTARY Univariate, arrhythmogenic arrhythmias can explain some congenital arrhythmias, however, it is far from a comprehensive understanding of the complexity of many arrhythmias. Mutational screening is a critical step in personalized medicine and is critical to the management of patients with arrhythmias. The success of personalized medicine requires a more efficient way to identify a high number of genetic variants potentially implicated in cardiac arrhythmogenic diseases than traditional sequencing methods (eg, Sanger sequencing). Next-generation sequencing technology provides us with unprecedented opportunities to achieve high-throughput, rapid, and cost-effective detection of congenital arrhythmias in patients. Moreover, in personalized medicine era, IPSC derived-cardiomyocytes can be used as 'cardiac arrhythmia in a dish' model for drug discovery, and help us improve management of arrhythmias in patients by developing patient-specific drug therapies with target specificity.
Collapse
Affiliation(s)
- Abdullah Omar
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mi Zhou
- Cardiac Surgery department, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Adam Berman
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Robert A. Sorrentino
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Neela Yar
- Purdue University, West Lafayette, IN, USA
| | - Neal L. Weintraub
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-man Kim
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wei Lei
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yaoliang Tang
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
99
|
Devalla HD, Gélinas R, Aburawi EH, Beqqali A, Goyette P, Freund C, Chaix MA, Tadros R, Jiang H, Le Béchec A, Monshouwer-Kloots JJ, Zwetsloot T, Kosmidis G, Latour F, Alikashani A, Hoekstra M, Schlaepfer J, Mummery CL, Stevenson B, Kutalik Z, de Vries AA, Rivard L, Wilde AA, Talajic M, Verkerk AO, Al-Gazali L, Rioux JD, Bhuiyan ZA, Passier R. TECRL, a new life-threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT. EMBO Mol Med 2016; 8:1390-1408. [PMID: 27861123 PMCID: PMC5167130 DOI: 10.15252/emmm.201505719] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genetic causes of many familial arrhythmia syndromes remain elusive. In this study, whole‐exome sequencing (WES) was carried out on patients from three different families that presented with life‐threatening arrhythmias and high risk of sudden cardiac death (SCD). Two French Canadian probands carried identical homozygous rare variant in TECRL gene (p.Arg196Gln), which encodes the trans‐2,3‐enoyl‐CoA reductase‐like protein. Both patients had cardiac arrest, stress‐induced atrial and ventricular tachycardia, and QT prolongation on adrenergic stimulation. A third patient from a consanguineous Sudanese family diagnosed with catecholaminergic polymorphic ventricular tachycardia (CPVT) had a homozygous splice site mutation (c.331+1G>A) in TECRL. Analysis of intracellular calcium ([Ca2+]i) dynamics in human induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CMs) generated from this individual (TECRLHom‐hiPSCs), his heterozygous but clinically asymptomatic father (TECRLHet‐hiPSCs), and a healthy individual (CTRL‐hiPSCs) from the same Sudanese family, revealed smaller [Ca2+]i transient amplitudes as well as elevated diastolic [Ca2+]i in TECRLHom‐hiPSC‐CMs compared with CTRL‐hiPSC‐CMs. The [Ca2+]i transient also rose markedly slower and contained lower sarcoplasmic reticulum (SR) calcium stores, evidenced by the decreased magnitude of caffeine‐induced [Ca2+]i transients. In addition, the decay phase of the [Ca2+]i transient was slower in TECRLHom‐hiPSC‐CMs due to decreased SERCA and NCX activities. Furthermore, TECRLHom‐hiPSC‐CMs showed prolonged action potentials (APs) compared with CTRL‐hiPSC‐CMs. TECRL knockdown in control human embryonic stem cell‐derived CMs (hESC‐CMs) also resulted in significantly longer APs. Moreover, stimulation by noradrenaline (NA) significantly increased the propensity for triggered activity based on delayed afterdepolarizations (DADs) in TECRLHom‐hiPSC‐CMs and treatment with flecainide, a class Ic antiarrhythmic drug, significantly reduced the triggered activity in these cells. In summary, we report that mutations in TECRL are associated with inherited arrhythmias characterized by clinical features of both LQTS and CPVT. Patient‐specific hiPSC‐CMs recapitulated salient features of the clinical phenotype and provide a platform for drug screening evidenced by initial identification of flecainide as a potential therapeutic. These findings have implications for diagnosis and treatment of inherited cardiac arrhythmias.
Collapse
Affiliation(s)
- Harsha D Devalla
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Roselle Gélinas
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Elhadi H Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Abdelaziz Beqqali
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Christian Freund
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center hiPSC Core Facility, Leiden, The Netherlands
| | - Marie-A Chaix
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Rafik Tadros
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hui Jiang
- Beijing Genomics Institute, Shenzhen, China.,Shenzhen Key Laboratory of Genomics, Shenzhen, China.,The Guangdong Enterprise Key Laboratory of Human Disease Genomics, Shenzhen, China
| | - Antony Le Béchec
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Tom Zwetsloot
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Georgios Kosmidis
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Maaike Hoekstra
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jurg Schlaepfer
- Service de Cardiologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Christine L Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brian Stevenson
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Zoltan Kutalik
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Institute of Social and Preventive Medicine, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Antoine Af de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Léna Rivard
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Arthur Am Wilde
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, Jeddah, Saudi Arabia
| | - Mario Talajic
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Arie O Verkerk
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lihadh Al-Gazali
- Department of Pediatrics, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - John D Rioux
- Montreal Heart Institute, Montreal, QC, Canada .,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Zahurul A Bhuiyan
- Laboratoire Génétiqué Moléculaire, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Robert Passier
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands .,Department of Applied Stem Cell Technologies, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| |
Collapse
|
100
|
Sasaki K, Makiyama T, Yoshida Y, Wuriyanghai Y, Kamakura T, Nishiuchi S, Hayano M, Harita T, Yamamoto Y, Kohjitani H, Hirose S, Chen J, Kawamura M, Ohno S, Itoh H, Takeuchi A, Matsuoka S, Miura M, Sumitomo N, Horie M, Yamanaka S, Kimura T. Patient-Specific Human Induced Pluripotent Stem Cell Model Assessed with Electrical Pacing Validates S107 as a Potential Therapeutic Agent for Catecholaminergic Polymorphic Ventricular Tachycardia. PLoS One 2016; 11:e0164795. [PMID: 27764147 PMCID: PMC5072719 DOI: 10.1371/journal.pone.0164795] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022] Open
Abstract
Introduction Human induced pluripotent stem cells (hiPSCs) offer a unique opportunity for disease modeling. However, it is not invariably successful to recapitulate the disease phenotype because of the immaturity of hiPSC-derived cardiomyocytes (hiPSC-CMs). The purpose of this study was to establish and analyze iPSC-based model of catecholaminergic polymorphic ventricular tachycardia (CPVT), which is characterized by adrenergically mediated lethal arrhythmias, more precisely using electrical pacing that could promote the development of new pharmacotherapies. Method and Results We generated hiPSCs from a 37-year-old CPVT patient and differentiated them into cardiomyocytes. Under spontaneous beating conditions, no significant difference was found in the timing irregularity of spontaneous Ca2+ transients between control- and CPVT-hiPSC-CMs. Using Ca2+ imaging at 1 Hz electrical field stimulation, isoproterenol induced an abnormal diastolic Ca2+ increase more frequently in CPVT- than in control-hiPSC-CMs (control 12% vs. CPVT 43%, p<0.05). Action potential recordings of spontaneous beating hiPSC-CMs revealed no significant difference in the frequency of delayed afterdepolarizations (DADs) between control and CPVT cells. After isoproterenol application with pacing at 1 Hz, 87.5% of CPVT-hiPSC-CMs developed DADs, compared to 30% of control-hiPSC-CMs (p<0.05). Pre-incubation with 10 μM S107, which stabilizes the closed state of the ryanodine receptor 2, significantly decreased the percentage of CPVT-hiPSC-CMs presenting DADs to 25% (p<0.05). Conclusions We recapitulated the electrophysiological features of CPVT-derived hiPSC-CMs using electrical pacing. The development of DADs in the presence of isoproterenol was significantly suppressed by S107. Our model provides a promising platform to study disease mechanisms and screen drugs.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adult
- Animals
- Anti-Asthmatic Agents/chemistry
- Anti-Asthmatic Agents/pharmacology
- Anti-Asthmatic Agents/therapeutic use
- Calcium/metabolism
- Calreticulin/genetics
- Calreticulin/metabolism
- Calsequestrin/genetics
- Calsequestrin/metabolism
- Cell Differentiation/drug effects
- Cells, Cultured
- Electric Stimulation
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Isoproterenol/pharmacology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Models, Biological
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/transplantation
- Ryanodine/pharmacology
- Ryanodine Receptor Calcium Release Channel/chemistry
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Tachycardia, Ventricular/drug therapy
- Tachycardia, Ventricular/pathology
- Tachycardia, Ventricular/therapy
- Thiazepines/chemistry
- Thiazepines/pharmacology
- Thiazepines/therapeutic use
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Kenichi Sasaki
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeru Makiyama
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- * E-mail: (TM); (Y. Yoshida)
| | - Yoshinori Yoshida
- Kyoto University iPS Cell Research and Application, Kyoto, Japan
- * E-mail: (TM); (Y. Yoshida)
| | - Yimin Wuriyanghai
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Tsukasa Kamakura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Suguru Nishiuchi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mamoru Hayano
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Harita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuta Yamamoto
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohiko Kohjitani
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sayako Hirose
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jiarong Chen
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mihoko Kawamura
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Seiko Ohno
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Hideki Itoh
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masaru Miura
- Division of Cardiology, Tokyo Metropolitan Children’s Medical Center, Tokyo, Japan
| | - Naokata Sumitomo
- Department of Pediatric Cardiology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Shinya Yamanaka
- Kyoto University iPS Cell Research and Application, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|