51
|
Hong L, Zhang M, Sridhar A, Darbar D. Pathogenic mutations perturb calmodulin regulation of Na v1.8 channel. Biochem Biophys Res Commun 2020; 533:168-174. [PMID: 32948286 PMCID: PMC11038804 DOI: 10.1016/j.bbrc.2020.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
The voltage-gated sodium channels play a key role in the generation and propagation of the cardiac action potential. Emerging data indicate that the Nav1.8 channel, encoded by the SCN10A gene, is a modulator of cardiac conduction and variation in the gene has been associated with arrhythmias such as atrial fibrillation (AF) and Brugada syndrome (BrS). The voltage gated sodium channels contain a calmodulin (CaM)-binding IQ domain involved in channel slow inactivation, we here investigated the role of CaM regulation of Nav1.8 channel function, and showed that CaM enhanced slow inactivation of the Nav1.8 channel and hyperpolarized steady-state inactivation curve of sodium currents. The effects of CaM on the channel gating were disrupted in the Nav1.8 channel truncated IQ domain. We studied Nav1.8 IQ domain mutations associated with AF and BrS, and found that a BrS-linked mutation (R1863Q) reduced the CaM-induced hyperpolarization shift, AF-linked mutations (R1869C and R1869G) disrupted CaM-induced enhanced inactivation, and effects of CaM on both development and recovery from slow inactivation were attenuated in all pathogenic mutations. Our findings indicate a role of CaM in the regulation of Nav1.8 channel function in cardiac arrhythmias.
Collapse
Affiliation(s)
- Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Meihong Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Arvind Sridhar
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dawood Darbar
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Administration, Chicago, IL, USA.
| |
Collapse
|
52
|
Blok M, Boukens BJ. Mechanisms of Arrhythmias in the Brugada Syndrome. Int J Mol Sci 2020; 21:ijms21197051. [PMID: 32992720 PMCID: PMC7582368 DOI: 10.3390/ijms21197051] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmias in Brugada syndrome patients originate in the right ventricular outflow tract (RVOT). Over the past few decades, the characterization of the unique anatomy and electrophysiology of the RVOT has revealed the arrhythmogenic nature of this region. However, the mechanisms that drive arrhythmias in Brugada syndrome patients remain debated as well as the exact site of their occurrence in the RVOT. Identifying the site of origin and mechanism of Brugada syndrome would greatly benefit the development of mechanism-driven treatment strategies.
Collapse
Affiliation(s)
- Michiel Blok
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-(0)20-566-4659
| |
Collapse
|
53
|
Tse G, Lee S, Liu T, Yuen HC, Wong ICK, Mak C, Mok NS, Wong WT. Identification of Novel SCN5A Single Nucleotide Variants in Brugada Syndrome: A Territory-Wide Study From Hong Kong. Front Physiol 2020; 11:574590. [PMID: 33071830 PMCID: PMC7531256 DOI: 10.3389/fphys.2020.574590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Background The aim of this study is to report on the genetic composition of Brugada syndrome (BrS) patients undergoing genetic testing in Hong Kong. Methods Patients with suspected BrS who presented to the Hospital Authority of Hong Kong between 1997 and 2019, and underwent genetic testing, were analyzed retrospectively. Results A total of 65 subjects were included (n = 65, 88% male, median presenting age 42 [30–54] years old, 58% type 1 pattern). Twenty-two subjects (34%) showed abnormal genetic test results, identifying the following six novel, pathogenic or likely pathogenic mutations in SCN5A: c.674G > A, c.2024-11T > A, c.2042A > C, c.4279G > T, c.5689C > T, c.429del. Twenty subjects (31%) in the cohort suffered from spontaneous ventricular tachycardia/ventricular fibrillation (VT/VF) and 18 (28%) had incident VT/VF over a median follow-up of 83 [Q1–Q3: 52–112] months. Univariate Cox regression demonstrated that syncope (hazard ratio [HR]: 4.27 [0.95–19.30]; P = 0.059), prior VT/VF (HR: 21.34 [5.74–79.31; P < 0.0001) and T-wave axis (HR: 0.970 [0.944–0.998]; P = 0.036) achieved P < 0.10 for predicting incident VT/VF. After multivariate adjustment, only prior VT/VF remained a significant predictor (HR: 12.39 [2.97–51.67], P = 0.001). Conclusion This study identified novel mutations in SCN5A in a Chinese cohort of BrS patients.
Collapse
Affiliation(s)
- Gary Tse
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China.,Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Sharen Lee
- Laboratory of Cardiovascular Physiology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ho Chuen Yuen
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Kowloon, China
| | - Ian Chi Kei Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, China.,School of Pharmacy, University College London, London, United Kingdom
| | - Chloe Mak
- Department of Pathology, Hong Kong Children's Hospital, Kowloon, China
| | - Ngai Shing Mok
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Kowloon, China
| | - Wing Tak Wong
- State Key Laboratory of Agrobiotechnology (CUHK), School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
54
|
Barajas-Martinez H, Smith M, Hu D, Goodrow RJ, Puleo C, Hasdemir C, Antzelevitch C, Pfeiffer R, Treat JA, Cordeiro JM. Susceptibility to Ventricular Arrhythmias Resulting from Mutations in FKBP1B, PXDNL, and SCN9A Evaluated in hiPSC Cardiomyocytes. Stem Cells Int 2020; 2020:8842398. [PMID: 32952569 PMCID: PMC7481990 DOI: 10.1155/2020/8842398] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We report an inherited cardiac arrhythmia syndrome consisting of Brugada and Early Repolarization Syndrome associated with variants in SCN9A, PXDNL, and FKBP1B. The proband inherited the 3 mutations and exhibited palpitations and arrhythmia-mediated syncope, whereas the parents and sister, who carried one or two of the mutations, were asymptomatic. METHODS AND RESULTS We assessed the functional impact of these mutations in induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) derived from the proband and an unaffected family member. Current and voltage clamp recordings, as well as confocal microscopy analysis of Ca2+ transients, were evaluated in hiPSC-CMs from the proband and compared these results with hiPSC-CMs from undiseased controls. Genetic analysis using next-generation DNA sequencing revealed heterozygous mutations in SCN9A, PXDNL, and FKBP1B in the proband. The proband displayed right bundle branch block and exhibited episodes of syncope. The father carried a mutation in FKBP1B, whereas the mother and sister carried the SCN9A mutation. None of the 3 family members screened developed cardiac events. Action potential recordings from control hiPSC-CM showed spontaneous activity and a low upstroke velocity. In contrast, the hiPSC-CM from the proband showed irregular spontaneous activity. Confocal microscopy of the hiPSC-CM of the proband revealed low fluorescence intensity Ca2+ transients that were episodic in nature. Patch-clamp measurements in hiPSC-CM showed no difference in I Na but reduced I Ca in the proband compared with control. Coexpression of PXDNL-R391Q with SCN5A-WT displayed lower I Na density compared to PXDNL-WT. In addition, coexpression of PXDNL-R391Q with KCND3-WT displayed significantly higher I to density compared to PXDNL-WT. CONCLUSION SCN9A, PXDNL, and FKBP1B variants appeared to alter spontaneous activity in hiPSC-CM. Only the proband carrying all 3 mutations displayed the ERS/BrS phenotype, whereas one nor two mutations alone did not produce the clinical phenotype. Our results suggest a polygenic cause of the BrS/ERS arrhythmic phenotype due to mutations in these three gene variants caused a very significant loss of function of I Na and I Ca and gain of function of I to.
Collapse
Affiliation(s)
- Hector Barajas-Martinez
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
| | - Maya Smith
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Dan Hu
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiology & Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, China
| | - Robert J. Goodrow
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Colleen Puleo
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Charles Antzelevitch
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
- Kimmel College of Medicine of Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryan Pfeiffer
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jacqueline A. Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jonathan M. Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| |
Collapse
|
55
|
A novel loss-of-function mutation of PBK associated with human kidney stone disease. Sci Rep 2020; 10:10282. [PMID: 32581305 PMCID: PMC7314804 DOI: 10.1038/s41598-020-66936-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/12/2020] [Indexed: 01/17/2023] Open
Abstract
Kidney stone disease (KSD) is a prevalent disorder that causes human morbidity worldwide. The etiology of KSD is heterogeneous, ranging from monogenic defect to complex interaction between genetic and environmental factors. Since mutations of genes responsible for KSD in a majority of families are still unknown, our group is identifying mutations of these genes by means of genomic and genetic analyses. In this study, we identified a novel loss-of-function mutation of PBK, encoding the PDZ binding kinase, that was found to be associated with KSD in an affected Thai family. Glycine (Gly) substituted by arginine (Arg) at position 43 (p.Gly43Arg) in PBK cosegregated with the disease in affected members of this family, but was absent in 180 normal control subjects from the same local population. Gly43 is highly evolutionarily conserved in vertebrates, and its substitution affects protein structure by alterations in H-bond forming patterns. This p.Gly43Arg substitution results in instability of the variant PBK protein as examined in HEK293T cells. The variant PBK protein (p.Gly43Arg) demonstrated decreased kinase activity to phosphorylate p38 MAPK as analyzed by immunoblotting and antibody microarray techniques. Taken together, these findings suggest a possible new mechanism of KSD associated with pathogenic PBK variation.
Collapse
|
56
|
Chang D, Saleh M, Garcia-Bengo Y, Choi E, Epstein L, Willner J. COVID-19 Infection Unmasking Brugada Syndrome. HeartRhythm Case Rep 2020; 6:237-240. [PMID: 32292696 PMCID: PMC7138191 DOI: 10.1016/j.hrcr.2020.03.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- David Chang
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Moussa Saleh
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Youssef Garcia-Bengo
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Evan Choi
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Laurence Epstein
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Jonathan Willner
- Department of Cardiology, Division of Electrophysiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| |
Collapse
|
57
|
Monasky MM, Micaglio E, Ciconte G, Pappone C. Brugada Syndrome: Oligogenic or Mendelian Disease? Int J Mol Sci 2020; 21:ijms21051687. [PMID: 32121523 PMCID: PMC7084676 DOI: 10.3390/ijms21051687] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Brugada syndrome (BrS) is diagnosed by a coved-type ST-segment elevation in the right precordial leads on the electrocardiogram (ECG), and it is associated with an increased risk of sudden cardiac death (SCD) compared to the general population. Although BrS is considered a genetic disease, its molecular mechanism remains elusive in about 70-85% of clinically-confirmed cases. Variants occurring in at least 26 different genes have been previously considered causative, although the causative effect of all but the SCN5A gene has been recently challenged, due to the lack of systematic, evidence-based evaluations, such as a variant's frequency among the general population, family segregation analyses, and functional studies. Also, variants within a particular gene can be associated with an array of different phenotypes, even within the same family, preventing a clear genotype-phenotype correlation. Moreover, an emerging concept is that a single mutation may not be enough to cause the BrS phenotype, due to the increasing number of common variants now thought to be clinically relevant. Thus, not only the complete list of genes causative of the BrS phenotype remains to be determined, but also the interplay between rare and common multiple variants. This is particularly true for some common polymorphisms whose roles have been recently re-evaluated by outstanding works, including considering for the first time ever a polygenic risk score derived from the heterozygous state for both common and rare variants. The more common a certain variant is, the less impact this variant might have on heart function. We are aware that further studies are warranted to validate a polygenic risk score, because there is no mutated gene that connects all, or even a majority, of BrS cases. For the same reason, it is currently impossible to create animal and cell line genetic models that represent all BrS cases, which would enable the expansion of studies of this syndrome. Thus, the best model at this point is the human patient population. Further studies should first aim to uncover genetic variants within individuals, as well as to collect family segregation data to identify potential genetic causes of BrS.
Collapse
Affiliation(s)
| | | | | | - Carlo Pappone
- Correspondence: ; Tel.: +39-0252-774260; Fax: +39-0252-774306
| |
Collapse
|
58
|
Affiliation(s)
- Katja E Odening
- Department of Cardiology and Angiology I and Institute of Experimental Cardiovascular Medicine, Heart Center University of Freiburg, Medical Faculty, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| |
Collapse
|
59
|
Heathfield LJ, Martin LJ, Ramesar R. Massively parallel sequencing in sudden unexpected death in infants: A case report in South Africa. FORENSIC SCIENCE INTERNATIONAL GENETICS SUPPLEMENT SERIES 2019. [DOI: 10.1016/j.fsigss.2019.10.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
60
|
Casini S, Marchal GA, Kawasaki M, Nariswari FA, Portero V, van den Berg NWE, Guan K, Driessen AHG, Veldkamp MW, Mengarelli I, de Groot JR, Verkerk AO, Remme CA. Absence of Functional Na v1.8 Channels in Non-diseased Atrial and Ventricular Cardiomyocytes. Cardiovasc Drugs Ther 2019; 33:649-660. [PMID: 31916131 PMCID: PMC6994555 DOI: 10.1007/s10557-019-06925-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Several studies have indicated a potential role for SCN10A/NaV1.8 in modulating cardiac electrophysiology and arrhythmia susceptibility. However, by which mechanism SCN10A/NaV1.8 impacts on cardiac electrical function is still a matter of debate. To address this, we here investigated the functional relevance of NaV1.8 in atrial and ventricular cardiomyocytes (CMs), focusing on the contribution of NaV1.8 to the peak and late sodium current (INa) under normal conditions in different species. METHODS The effects of the NaV1.8 blocker A-803467 were investigated through patch-clamp analysis in freshly isolated rabbit left ventricular CMs, human left atrial CMs and human-induced pluripotent stem cell-derived CMs (hiPSC-CMs). RESULTS A-803467 treatment caused a slight shortening of the action potential duration (APD) in rabbit CMs and hiPSC-CMs, while it had no effect on APD in human atrial cells. Resting membrane potential, action potential (AP) amplitude, and AP upstroke velocity were unaffected by A-803467 application. Similarly, INa density was unchanged after exposure to A-803467 and NaV1.8-based late INa was undetectable in all cell types analysed. Finally, low to absent expression levels of SCN10A were observed in human atrial tissue, rabbit ventricular tissue and hiPSC-CMs. CONCLUSION We here demonstrate the absence of functional NaV1.8 channels in non-diseased atrial and ventricular CMs. Hence, the association of SCN10A variants with cardiac electrophysiology observed in, e.g. genome wide association studies, is likely the result of indirect effects on SCN5A expression and/or NaV1.8 activity in cell types other than CMs.
Collapse
Affiliation(s)
- Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands.
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Makiri Kawasaki
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Fransisca A Nariswari
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | | | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Antoine H G Driessen
- Department of Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Marieke W Veldkamp
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Joris R de Groot
- Department of Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
- Department of Medical Biology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|
61
|
Coppola G, Corrado E, Curnis A, Maglia G, Oriente D, Mignano A, Brugada P. Update on Brugada Syndrome 2019. Curr Probl Cardiol 2019; 46:100454. [PMID: 31522883 DOI: 10.1016/j.cpcardiol.2019.100454] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022]
Abstract
Brugada syndrome (BrS) was first described in 1992 as an aberrant pattern of ST segment elevation in right precordial leads with a high incidence of sudden cardiac death (SCD) in patients with structurally normal heart. It represents 4% ∼ 12% of all SCD and 20% of SCD in patients with structurally normal heart. The extremely wide genetic heterogeneity of BrS and other inherited cardiac disorders makes this new area of genetic arrhytmology a fascinating one. This review shows the state of art in diagnosis, management, and treatment of BrS focusing all the aspects regarding genetics and Preimplant Genetic Diagnosis (PGD) of embryos, overlapping syndromes, risk stratification, familial screening, and future perspectives. Moreover the review analyzes key points like electrocardiogram (ECG) criteria, the role of electrophysiological study (the role of ventricular programmed stimulation and the need of universal accepted protocol) and the importance of a correct risk stratification to clarify when implantable cardioverter defibrillator or a close follow-up is needed. In recent years, cardiovascular studies have been focused on personalized risk assessment and to determine the most optimal therapy for an individual. The BrS syndrome has also benefited of these advances although there remain several key points to be elucidated. We will review the present knowledge, progress made, and future research directions on BrS.
Collapse
|
62
|
Zhang Y, Li A, Huang CLH, Wang G, Wang D. Generation of induced pluripotent stem cells (iPSCs) from an infant with catecholaminergic polymorphic ventricular tachycardia carrying the double heterozygous mutations A1855D in RyR2 and Q1362H in SCN10A. Stem Cell Res 2019; 39:101509. [PMID: 31382203 DOI: 10.1016/j.scr.2019.101509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 07/14/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) were generated from peripheral blood mononuclear cells (PBMCs) isolated from the peripheral blood of a 4 month-old boy with catecholaminergic polymorphic ventricular tachycardia carrying the double heterozygous mutations RyR2-A1855D and SCN10A-Q1362H. PBMCs were reprogrammed using non-integrative Sendai viral vectors containing reprogramming factors OCT4, SOX2, KLF4 and C-MYC. The iPSCs were shown to express pluripotent markers, have trilineage differentiation potential, carry RyR2-A1855D and SCN10A-Q1362H mutations and have a normal karyotype. They will be useful for studying the pathogenesis of CPVT patients with ≥2 variants.
Collapse
Affiliation(s)
- Yanmin Zhang
- Children's Research Institute of Shaanxi Province, China; Xi'an Key Laboratory of Children's Health and Diseases, China; Department of Cardiology, Affiliate Children's Hospital of Xi'an Jiaotong University, Xi'an 710003, China.
| | - Anmao Li
- Children's Research Institute of Shaanxi Province, China; Xi'an Key Laboratory of Children's Health and Diseases, China
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB3 0DF, United Kingdom
| | - Guoxia Wang
- Children's Research Institute of Shaanxi Province, China; Xi'an Key Laboratory of Children's Health and Diseases, China
| | | |
Collapse
|
63
|
Rodríguez -Constaín JS, López-Garzón NA, Navia-Amézquita CA, Mora-Obando DL, Dueñas-Cuellar RA. Síndrome de Brugada. Aspectos fisiopatológicos, clínicos y su asociación con enfermedades infecciosas. IATREIA 2019. [DOI: 10.17533/udea.iatreia.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
El síndrome de Brugada (SBr) es una enfermedad cardiaca no estructural que afecta los canales iónicos cardiacos, caracterizado por manifestaciones clínicas como arritmias, taquicardia, síncope y muerte súbita, entre otras. Su diagnóstico es netamente electrocardiográfico, con un patrón altamente sugestivo pero no patognomónico, por lo que existen diagnósticos diferenciales desde el punto de vista electrocardiográfico.Existen tres patrones electrocardiográficos en los pacientes con SBr, de los cuales el tipo I es el patrón más característico. Actualmente, múltiples genes se han relacionado con la presentación de este síndrome, entre los cuales se destaca el gen SCN5A, el más descrito en la literatura. Se conoce que este síndrome es más frecuente en el género masculino; sin embargo, no existen estudios epidemiológicos en Latinoamérica que lo confirmen. Pese a que la investigación alrededor de los mecanismos causales del síndrome ha avanzado, existen varias cuestiones sin resolver, como su desenmascaramiento por los signos que producen algunas enfermedades infecciosas causadas principalmente por virus. Por lo tanto, dada la relevancia clínica del tema para el médico general y para el especialista, el objetivo de esta revisión es describir no solo aspectos fisiopatológicos y clínicos de la enfermedad, sino también resaltar casos de pacientes con enfermedades infecciosas quienes posteriormente han sido diagnosticados con el síndrome de Brugada.
Collapse
|
64
|
El-Battrawy I, Borggrefe M, Lang S, Zhou X, Akin I. Genotype-phenotype association in patients with SCN4A mutation. Lancet 2019; 393:2301. [PMID: 31180026 DOI: 10.1016/s0140-6736(19)31298-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, 68167, Germany; German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Mannheim, Germany.
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, 68167, Germany; German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Siegried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, 68167, Germany; German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, 68167, Germany; German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Ibrahim Akin
- German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Mannheim, Germany
| |
Collapse
|
65
|
Matthews E, Männikkö R, Behr E, Hanna MG. Genotype-phenotype association in patients with SCN4A mutation - Authors' reply. Lancet 2019; 393:2301-2302. [PMID: 31180027 DOI: 10.1016/s0140-6736(19)30214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/14/2019] [Indexed: 11/24/2022]
Affiliation(s)
- Emma Matthews
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.
| | - Roope Männikkö
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Elijah Behr
- Cardiology Clinical Academic Group, St George's University of London, London, UK; St George's University Hospitals NHS Foundation Trust, London, UK
| | - Michael G Hanna
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
66
|
Gando I, Williams N, Fishman GI, Sampson BA, Tang Y, Coetzee WA. Functional characterization of SCN10A variants in several cases of sudden unexplained death. Forensic Sci Int 2019; 301:289-298. [PMID: 31195250 DOI: 10.1016/j.forsciint.2019.05.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/03/2019] [Accepted: 05/21/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Multiple genome-wide association studies (GWAS) and targeted gene sequencing have identified common variants in SCN10A in cases of PR and QRS duration abnormalities, atrial fibrillation and Brugada syndrome. The New York City Office of Chief Medical Examiner has now also identified five SCN10A variants of uncertain significance in six separate cases within a cohort of 330 sudden unexplained death events. The gene product of SCN10A is the Nav1.8 sodium channel. The purpose of this study was to characterize effects of these variants on Nav1.8 channel function to provide better information for the reclassification of these variants. METHODS AND RESULTS Patch clamp studies were performed to assess effects of the variants on whole-cell Nav1.8 currents. We also performed RNA-seq analysis and immunofluorescence confocal microcopy to determine Nav1.8 expression in heart. We show that four of the five rare 'variants of unknown significance' (L388M, L867F, P1102S and V1518I) are associated with altered functional phenotypes. The R756W variant behaved similar to wild-type under our experimental conditions. We failed to detect Nav1.8 protein expression in immunofluorescence microscopy in rat heart. Furthermore, RNA-seq analysis failed to detect full-length SCN10A mRNA transcripts in human ventricle or mouse specialized cardiac conduction system, suggesting that the effect of Nav1.8 on cardiac function is likely to be extra-cardiac in origin. CONCLUSIONS We have demonstrated that four of five SCN10A variants of uncertain significance, identified in unexplained death, have deleterious effects on channel function. These data extend the genetic testing of SUD cases, but significantly more clinical evidence is needed to satisfy the criteria needed to associate these variants with the onset of SUD.
Collapse
Affiliation(s)
| | - Nori Williams
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, United States
| | - Glenn I Fishman
- Neuroscience & Physiology, New York, NY, United States; Biochemistry and Molecular Pharmacology, New York, NY, United States; Medicine NYU School of Medicine, New York, NY, United States
| | - Barbara A Sampson
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, United States
| | - Yingying Tang
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, United States
| | - William A Coetzee
- Pediatrics, New York, NY, United States; Neuroscience & Physiology, New York, NY, United States; Biochemistry and Molecular Pharmacology, New York, NY, United States.
| |
Collapse
|
67
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
68
|
Romero J, Li DL, Avendano R, Diaz JC, Tung R, Di Biase L. Brugada Syndrome: Progress in Genetics, Risk Stratification and Management. Arrhythm Electrophysiol Rev 2019; 8:19-27. [PMID: 30918663 PMCID: PMC6434506 DOI: 10.15420/aer.2018.66.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brugada syndrome (BrS) is one of the most common causes of sudden cardiac death in normal structural heart individuals. First characterised in 1992, the global prevalence of BrS is unclear, with estimates placing it at around 0.05% and presenting most frequently in southeast Asian countries. This review aims to summarise the development in the understanding of BrS and, importantly, progress in its management, underpinned by knowledge regarding its genetics and molecular mechanisms. It also provides update on risk stratification and promising new therapies for BrS, including epicardial ablation. Future studies are required to increase understanding of the pathogenesis of this disease and to guide clinical practice.
Collapse
Affiliation(s)
- Jorge Romero
- Montefiore Medical Center, Albert Einstein College of Medicine Bronx, NY, US
| | - Dan L Li
- Montefiore Medical Center, Albert Einstein College of Medicine Bronx, NY, US.,Cardiovascular Division, Department of Medicine, Vanderbilt University Medical Center Nashville, TN, US
| | - Ricardo Avendano
- Montefiore Medical Center, Albert Einstein College of Medicine Bronx, NY, US.,Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine New Haven, CT, US
| | - Juan Carlos Diaz
- Montefiore Medical Center, Albert Einstein College of Medicine Bronx, NY, US
| | - Roderick Tung
- University of Chicago, School of Medicine Chicago, IL, US
| | - Luigi Di Biase
- Montefiore Medical Center, Albert Einstein College of Medicine Bronx, NY, US
| |
Collapse
|
69
|
Eijkenboom I, Sopacua M, Hoeijmakers JGJ, de Greef BTA, Lindsey P, Almomani R, Marchi M, Vanoevelen J, Smeets HJM, Waxman SG, Lauria G, Merkies ISJ, Faber CG, Gerrits MM. Yield of peripheral sodium channels gene screening in pure small fibre neuropathy. J Neurol Neurosurg Psychiatry 2019; 90:342-352. [PMID: 30554136 DOI: 10.1136/jnnp-2018-319042] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/01/2018] [Accepted: 11/18/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Neuropathic pain is common in peripheral neuropathy. Recent genetic studies have linked pathogenic voltage-gated sodium channel (VGSC) variants to human pain disorders. Our aims are to determine the frequency of SCN9A, SCN10A and SCN11A variants in patients with pure small fibre neuropathy (SFN), analyse their clinical features and provide a rationale for genetic screening. METHODS Between September 2009 and January 2017, 1139 patients diagnosed with pure SFN at our reference centre were screened for SCN9A, SCN10A and SCN11A variants. Pathogenicity of variants was classified according to established guidelines of the Association for Clinical Genetic Science and frequencies were determined. Patients with SFN were grouped according to the VGSC variants detected, and clinical features were compared. RESULTS Among 1139 patients with SFN, 132 (11.6%) patients harboured 73 different (potentially) pathogenic VGSC variants, of which 50 were novel and 22 were found in ≥ 1 patient. The frequency of (potentially) pathogenic variants was 5.1% (n=58/1139) for SCN9A, 3.7% (n=42/1139) for SCN10A and 2.9% (n=33/1139) for SCN11A. Only erythromelalgia-like symptoms and warmth-induced pain were significantly more common in patients harbouring VGSC variants. CONCLUSION (Potentially) pathogenic VGSC variants are present in 11.6% of patients with pure SFN. Therefore, genetic screening of SCN9A, SCN10A and SCN11A should be considered in patients with pure SFN, independently of clinical features or underlying conditions.
Collapse
Affiliation(s)
- Ivo Eijkenboom
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Maurice Sopacua
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bianca T A de Greef
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patrick Lindsey
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Jo Vanoevelen
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Hubertus J M Smeets
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Centre for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Ingemar S J Merkies
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Catharina G Faber
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
70
|
Chen CYJ, Lu TP, Lin LY, Liu YB, Ho LT, Huang HC, Lai LP, Hwang JJ, Yeh SFS, Wu CK, Juang JMJ, Antzelevitch C. Impact of Ancestral Differences and Reassessment of the Classification of Previously Reported Pathogenic Variants in Patients With Brugada Syndrome in the Genomic Era: A SADS-TW BrS Registry. Front Genet 2019; 9:680. [PMID: 30662450 PMCID: PMC6328444 DOI: 10.3389/fgene.2018.00680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) is a heritable disease that results in sudden cardiac death. In the exome/genomic era, certain reported pathogenic variants in some genetic diseases have been reclassified as benign owing to their high frequency in some ancestries. In the present study, we comprehensively reassessed all previously reported pathogenic variants of BrS. We collected all pathogenic variants of BrS reported in the Human Gene Mutation Database and ClinVar throughout April 2017. We compared the minor allele frequency (MAF) of each variant among different ancestries by searching public whole-genome and exome databases. After considering the maximum credible allele frequency, variants with a MAF ≥ 0.001 were considered to be of questionable pathogenicity. We also investigated the percentage of SCN5A variants with a MAF ≥ 0.001 in 124 BrS patients from the Han Chinese population. We collected a total of 440 BrS variants, of which 18 had a MAF ≥ 0.001. There was a greater percentage of non-SCN5A variants with a MAF ≥ 0.001 than of SCN5A variants (21.8 versus 1.6%, p < 0.0001). There were fewer frameshift and nonsense mutations than missense mutations (0.9 versus 5.6%, p = 0.032). Of the 18 variants, 14 (77.8%) were present only in the reference Asian population. In our cohort, we identified two SCN5A variants (p.A226V and p.V1340I) with MAFs ≥ 0.001 (0.45%). In conclusion, ancestral differences are important when considering the pathogenicity of BrS variants, especially in the case of missense variants and non-SCN5A variants, which may be pathogenic in some ancestries but only disease-predisposing in others.
Collapse
Affiliation(s)
- Ching-Yu Julius Chen
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzu-Pin Lu
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Lian-Yu Lin
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Bin Liu
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Ting Ho
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hui-Chun Huang
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ling-Ping Lai
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Juey-Jen Hwang
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Fan Sherri Yeh
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cho-Kai Wu
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jyh-Ming Jimmy Juang
- Department of Internal Medicine, Cardiovascular Center and Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood, PA, United States.,Lankenau Heart Institute, Wynnewood, PA, United States.,Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
71
|
Werner R, Niederseer D, Saguner AM, Brunckhorst CB. [CME ECG 62/Answer: An Atypical Right Bundle Branch Block?]. PRAXIS 2019; 108:157-160. [PMID: 30722744 DOI: 10.1024/1661-8157/a003152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Affiliation(s)
- Regine Werner
- 1 Klinik für Kardiologie, Universitäres Herzzentrum, Universitätsspital Zürich
| | - David Niederseer
- 1 Klinik für Kardiologie, Universitäres Herzzentrum, Universitätsspital Zürich
| | - Ardan M Saguner
- 1 Klinik für Kardiologie, Universitäres Herzzentrum, Universitätsspital Zürich
| | | |
Collapse
|
72
|
|
73
|
Abstract
In 2000, with the completion of the human genome project, nine related channels were found to comprise the complete voltage-gated sodium gene family and they were renamed NaV1.1–NaV1.9. This millennial event reflected the extraordinary impact of molecular genetics on our understanding of electrical signalling in the nervous system. In this review, studies of animal electricity from the time of Galvani to the present day are described. The seminal experiments and models of Hodgkin and Huxley coupled with the discovery of the structure of DNA, the genetic code and the application of molecular genetics have resulted in an appreciation of the extraordinary diversity of sodium channels and their surprisingly broad repertoire of functions. In the present era, unsuspected roles for sodium channels in a huge range of pathologies have become apparent.
Collapse
Affiliation(s)
- John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|
74
|
Casado Arroyo R, Sieira J, Kubala M, Latcu DG, Maeda S, Brugada P. Electrophysiological Basis for Early Repolarization Syndrome. Front Cardiovasc Med 2018; 5:161. [PMID: 30460246 PMCID: PMC6232947 DOI: 10.3389/fcvm.2018.00161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
During last centuries, Early Repolarization pattern has been interpreted as an ECG manifestation not linked to serious cardiovascular events. This view has been challenged on the basis of sporadic clinical observations that linked the J-wave with ventricular arrhythmias and sudden cardiac death. The particular role of this characteristic pattern in initiating ventricular fibrillation has been sustained by clinical descriptions of a marked and consistent J-wave elevation preceding the onset of the ventricular arrhythmia. Until now, Early Repolarization syndrome patients have been evaluated using ECG and theorizing different interpretations of the findings. Nonetheless, ECG analysis is not able to reveal all depolarization and repolarization properties and the explanation for this clinical events. Recent studies have characterized the epicardial substrate in these patients on the basis of high-resolution data, in an effort to provide insights into the substrate properties that support arrhythmogenicity in these patients. An overview for the current evidence supporting different theories explaining Early Repolarization Syndrome is provided in this review. Finally, future developments in the field directed toward individualized treatment strategies are examined.
Collapse
Affiliation(s)
- Rubén Casado Arroyo
- Department of Cardiology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Juan Sieira
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Maciej Kubala
- Department of Cardiology, Centre Hospitalier Universitaire, Amiens, France
| | | | - Shigo Maeda
- Advanced Arrhythmia Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Pedro Brugada
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
75
|
Thomas D, Christ T, Fabritz L, Goette A, Hammwöhner M, Heijman J, Kockskämper J, Linz D, Odening KE, Schweizer PA, Wakili R, Voigt N. German Cardiac Society Working Group on Cellular Electrophysiology state-of-the-art paper: impact of molecular mechanisms on clinical arrhythmia management. Clin Res Cardiol 2018; 108:577-599. [PMID: 30306295 DOI: 10.1007/s00392-018-1377-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/24/2018] [Indexed: 12/19/2022]
Abstract
Cardiac arrhythmias remain a common challenge and are associated with significant morbidity and mortality. Effective and safe rhythm control strategies are a primary, yet unmet need in everyday clinical practice. Despite significant pharmacological and technological advances, including catheter ablation and device-based therapies, the development of more effective alternatives is of significant interest to increase quality of life and to reduce symptom burden, hospitalizations and mortality. The mechanistic understanding of pathophysiological pathways underlying cardiac arrhythmias has advanced profoundly, opening up novel avenues for mechanism-based therapeutic approaches. Current management of arrhythmias, however, is primarily guided by clinical and demographic characteristics of patient groups as opposed to individual, patient-specific mechanisms and pheno-/genotyping. With this state-of-the-art paper, the Working Group on Cellular Electrophysiology of the German Cardiac Society aims to close the gap between advanced molecular understanding and clinical decision-making in cardiac electrophysiology. The significance of cellular electrophysiological findings for clinical arrhythmia management constitutes the main focus of this document. Clinically relevant knowledge of pathophysiological pathways of arrhythmias and cellular mechanisms of antiarrhythmic interventions are summarized. Furthermore, the specific molecular background for the initiation and perpetuation of atrial and ventricular arrhythmias and mechanism-based strategies for therapeutic interventions are highlighted. Current "hot topics" in atrial fibrillation are critically appraised. Finally, the establishment and support of cellular and translational electrophysiology programs in clinical rhythmology departments is called for to improve basic-science-guided patient management.
Collapse
Affiliation(s)
- Dierk Thomas
- Department of Cardiology, Medical University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany. .,HCR (Heidelberg Center for Heart Rhythm Disorders), Heidelberg, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.,Department of Cardiology, UHB NHS Trust, Birmingham, UK.,Department of Cardiovascular Medicine, Division of Rhythmology, University Hospital Münster, Münster, Germany
| | - Andreas Goette
- St. Vincenz-Hospital, Paderborn, Germany.,Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Magdeburg, Germany
| | - Matthias Hammwöhner
- St. Vincenz-Hospital, Paderborn, Germany.,Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Magdeburg, Germany
| | - Jordi Heijman
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jens Kockskämper
- Biochemical and Pharmacological Center (BPC) Marburg, Institute of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Dominik Linz
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, SA, Australia.,Experimental Electrophysiology, University Hospital of Saarland, Homburg, Saar, Germany
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute for Experimental Cardiovascular Medicine, Heart Center University of Freiburg, Freiburg, Germany
| | - Patrick A Schweizer
- Department of Cardiology, Medical University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.,Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany
| | - Reza Wakili
- Department of Cardiology and Vascular Medicine, Medical Faculty, West German Heart Center, University Hospital Essen, Essen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
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
|
Di Stolfo G, Palumbo P, Castellana S, Mastroianno S, Biagini T, Palumbo O, Leone MP, De Luca G, Potenza DR, Mazza T, Russo AA, Carella M. Sudden cardiac death in J wave syndrome with short QT associated to a novel mutation in Nav 1.8 coding gene SCN10A: First case report for a possible pharmacogenomic role. J Electrocardiol 2018; 51:809-813. [DOI: 10.1016/j.jelectrocard.2018.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/08/2018] [Indexed: 11/16/2022]
|
78
|
Loss-of-function of Nav1.8/D1639N linked to human pain can be rescued by lidocaine. Pflugers Arch 2018; 470:1787-1801. [PMID: 30099632 DOI: 10.1007/s00424-018-2189-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 01/31/2023]
Abstract
Mutations in voltage-gated sodium channels are associated with altered pain perception in humans. Most of these mutations studied to date present with a direct and intuitive link between the altered electrophysiological function of the channel and the phenotype of the patient. In this study, we characterize a variant of Nav1.8, D1639N, which has been previously identified in a patient suffering from the chronic pain syndrome "small fiber neuropathy". Using a heterologous expression system and patch-clamp analysis, we show that Nav1.8/D1639N reduces current density without altering biophysical gating properties of Nav1.8. Therefore, the D1639N variant causes a loss-of-function of the Nav1.8 sodium channel in a patient suffering from chronic pain. Using immunocytochemistry and biochemical approaches, we show that Nav1.8/D1639N impairs trafficking of the channel to the cell membrane. Neither co-expression of β1 or β3 subunit, nor overnight incubation at 27 °C rescued current density of the D1639N variant. On the other hand, overnight incubation with lidocaine fully restored current density of Nav1.8/D1639N most likely by overcoming the trafficking defect, whereas phenytoin failed to do so. Since lidocaine rescues the loss-of-function of Nav1.8/D1639N, it may offer a future therapeutic option for the patient carrying this variant. These results demonstrate that the D1639N variant, identified in a patient suffering from chronic pain, causes loss-of-function of the channel due to impaired cell surface trafficking and that this trafficking defect can be rescued by lidocaine.
Collapse
|
79
|
Sicouri S, Antzelevitch C. Mechanisms Underlying the Actions of Antidepressant and Antipsychotic Drugs That Cause Sudden Cardiac Arrest. Arrhythm Electrophysiol Rev 2018; 7:199-209. [PMID: 30416734 PMCID: PMC6141916 DOI: 10.15420/aer.2018.29.2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/19/2018] [Indexed: 12/18/2022] Open
Abstract
A number of antipsychotic and antidepressant drugs are known to increase the risk of ventricular arrhythmias and sudden cardiac death. Based largely on a concern over the development of life-threatening arrhythmias, a number of antipsychotic drugs have been temporarily or permanently withdrawn from the market or their use restricted. While many antidepressants and antipsychotics have been linked to QT prolongation and the development of torsade de pointes arrhythmias, some have been associated with a Brugada syndrome phenotype and the development of polymorphic ventricular arrhythmias. This article examines the arrhythmic liability of antipsychotic and antidepressant drugs capable of inducing long QT and/or Brugada syndrome phenotypes. The goal of this article is to provide an update on the ionic and cellular mechanisms thought to be involved in, and the genetic and environmental factors that predispose to, the development of cardiac arrhythmias and sudden cardiac death among patients taking antidepressant and antipsychotic drugs that are in clinical use.
Collapse
Affiliation(s)
- Serge Sicouri
- Lankenau Institute for Medical ResearchWynnewood, PA, USA
| | - Charles Antzelevitch
- Lankenau Institute for Medical ResearchWynnewood, PA, USA
- Lankenau Heart InstituteWynnewood, PA
- Sidney Kimmel Medical College of Thomas Jefferson UniversityPhiladelphia, PA, USA
| |
Collapse
|
80
|
Lin Y, Williams N, Wang D, Coetzee W, Zhou B, Eng LS, Um SY, Bao R, Devinsky O, McDonald TV, Sampson BA, Tang Y. Applying High-Resolution Variant Classification to Cardiac Arrhythmogenic Gene Testing in a Demographically Diverse Cohort of Sudden Unexplained Deaths. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.117.001839. [PMID: 29247119 DOI: 10.1161/circgenetics.117.001839] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/31/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Genetic variant interpretation contributes to testing yield differences reported for sudden unexplained death. Adapting a high-resolution variant interpretation framework, which considers disease prevalence, reduced penetrance, genetic heterogeneity, and allelic contribution to determine the maximum tolerated allele count in gnomAD, we report an evaluation of cardiac channelopathy and cardiomyopathy genes in a large, demographically diverse sudden unexplained death cohort that underwent thorough investigation in the United States' largest medical examiner's office. METHODS AND RESULTS The cohort has 296 decedents: 147 Blacks, 64 Hispanics, 49 Whites, 22 Asians, and 14 mixed ethnicities; 142 infants (1 to 11 months), 39 children (1 to 17 years), 74 young adults (18 to 34 years), and 41 adults (35 to 55 years). Eighty-nine cardiac disease genes were evaluated. Using a high-resolution variant interpretation workflow, we classified 17 variants as pathogenic or likely pathogenic (2 of which were incidental findings and excluded in testing yield analysis), 46 novel variants of uncertain significance, and 130 variants of uncertain significance. Nine pathogenic or likely pathogenic variants in ClinVar were reclassified to likely benign and excluded in testing yield analysis. The yields of positive cases by ethnicity and age were 21.4% in mixed ethnicities, 10.2% Whites, 4.5% Asians, 3.1% Hispanics, and 2% Blacks; 7.7% children, 7.3% in adults, 5.4% young adults, and 2.8% infants. The percentages of uncertain cases with variants of uncertain significance by ethnicity were 45.5% in Asians, 45.3% Hispanics, 44.20% Blacks, 36.7% Whites, and 14.3% in mixed ethnicities. CONCLUSIONS High-resolution variant interpretation provides diagnostic accuracy and healthcare efficiency. Under-represented populations warrant greater inclusion in future studies.
Collapse
Affiliation(s)
- Ying Lin
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Nori Williams
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Dawei Wang
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - William Coetzee
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Bo Zhou
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Lucy S Eng
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Sung Yon Um
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Ruijun Bao
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Orrin Devinsky
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Thomas V McDonald
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Barbara A Sampson
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.)
| | - Yingying Tang
- From the Laboratory Division, Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner (Y.L., N.W., D.W., B.Z., L.S.E., S.Y.U., R.B., B.A.S., Y.T.); Departments of Pediatrics, Biochemistry and Molecular Pharmacology, and Physiology and Neuroscience, NYU School of Medicine (W.C.); Department of Neurology, NYU Langone Medical Center (O.D.); and Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY (T.V.M.).
| |
Collapse
|
81
|
Bonilla JC, Parra-Medina R, Chaves JJ, Campuzano O, Sarquella-Brugada G, Brugada R, Brugada J. [Molecular autopsy in sudden cardiac death]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2018; 88:306-312. [PMID: 30030015 DOI: 10.1016/j.acmx.2018.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 06/06/2018] [Accepted: 06/09/2018] [Indexed: 10/28/2022] Open
Abstract
Currently, there are a significant percentage of autopsies left without a conclusive diagnosis of death, especially when this lethal event occurs suddenly. Genetic analysis has been recently incorporated into the field of forensic medicine, especially in patients with sudden death and where no conclusive cause of death is identified after a complete medical-legal autopsy. Inherited arrhythmogenic diseases are the main cause of death in these cases. To date, more than 40 genes have been associated with arrhythmogenic disease, and causing sudden cardiac death has been described. The main arrhythmogenic diseases are Long QT Syndrome, Catecholaminergic Polymorphic Ventricular Tachycardia, Brugada Syndrome, and Short QT Syndrome. These post-mortem genetic studies, not only allow a diagnosis of the cause of death, but also allow a clinical translation in relatives, focusing on the early identification of individuals at risk of syncope, as well as adopting personalised therapeutic measures for the prevention of a lethal arrhythmic episode.
Collapse
Affiliation(s)
- Juan Carlos Bonilla
- Departamento de Patología, Hospital de San José, Hospital Infantil Universitario de San José, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia.
| | - Rafael Parra-Medina
- Departamento de Patología, Hospital de San José, Hospital Infantil Universitario de San José, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia; Instituto de Investigación, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia
| | - Juan José Chaves
- Departamento de Patología, Hospital de San José, Hospital Infantil Universitario de San José, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia
| | - Oscar Campuzano
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica Girona (IDIBGI), Universidad de Girona, Girona, España; Department de Ciencias Médicas, Facultad de Medicina, Universidad de Girona, Girona, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares-CIBERCV, Madrid, España
| | | | - Ramón Brugada
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica Girona (IDIBGI), Universidad de Girona, Girona, España; Department de Ciencias Médicas, Facultad de Medicina, Universidad de Girona, Girona, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares-CIBERCV, Madrid, España; Servicio de Cardiología, Hospital Josep Trueta, Girona, España
| | - Josep Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares-CIBERCV, Madrid, España; Institut Clínic Cardiovascular (ICCV), Hospital Clínic, Universidad de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| |
Collapse
|
82
|
Hasdemir C, Juang JJM, Kose S, Kocabas U, Orman MN, Payzin S, Sahin H, Celen C, Ozcan EE, Chen CYJ, Gunduz R, Turan OE, Senol O, Burashnikov E, Antzelevitch C. Coexistence of atrioventricular accessory pathways and drug-induced type 1 Brugada pattern. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2018; 41:1078-1092. [PMID: 29953624 DOI: 10.1111/pace.13414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/30/2018] [Accepted: 05/13/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Atrial arrhythmias, particularly atrioventricular nodal reentrant tachycardia, can coexist with drug-induced type 1 Brugada electrocardiogram (ECG) pattern (DI-Type1-BrP). The present study was designed to determine the prevalence of DI-Type1-BrP in patients with atrioventricular accessory pathways (AV-APs) and to investigate the clinical, electrocardiographic, electrophysiologic, and genetic characteristics of these patients. METHODS One-hundred twenty-four consecutive cases of AV-APs and 84 controls underwent an ajmaline challenge test to unmask DI-Type1-BrP. Genetic screening and analysis was performed in 55 of the cases (19 with and 36 without DI-Type1-BrP). RESULTS Patients with AV-APs were significantly more likely than controls to have a Type1-BrP unmasked (16.1 vs 4.8%, P = 0.012). At baseline, patients with DI-Type1-BrP had higher prevalence of chest pain, QR/rSr' pattern in V1 and QRS notching/slurring in V2 and aVL during preexcitation, rSr' pattern in V1 -V2 , and QRS notching/slurring in aVL during orthodromic atrioventricular reentrant tachycardia (AVRT) compared to patients without DI-Type1-BrP. Abnormal QRS configuration (QRS notching/slurring and/or fragmentation) in V2 during preexcitation was present in all patients with DI-Type1 BrP. The prevalence of spontaneous preexcited atrial fibrillation (AF) and history of AF were similar (15% vs 18.3%, P = 0.726) in patients with and without DI-Type1-BrP, respectively. The prevalence of mutations in Brugada-susceptibility genes was higher (36.8% vs 8.3%, P = 0.02) in patients with DI-Type1-BrP compared to patients without DI-Type1-BrP. CONCLUSIONS DI-Type1-BrP is relatively common in patients with AV-APs. We identify 12-lead ECG characteristics during preexcitation and orthodromic AVRT that point to an underlying type1-BrP, portending an increased probability for development of malignant arrhythmias.
Collapse
Affiliation(s)
- Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | | | | | - Umut Kocabas
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Mehmet N Orman
- Department of Biostatistics and Medical Informatics, Ege University School of Medicine, Izmir, Turkey
| | - Serdar Payzin
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Hatice Sahin
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Candan Celen
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Emin E Ozcan
- Department of Cardiology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Ching-Yu Julius Chen
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood, PA, USA.,Lankenau Heart Institute, Wynnewood, PA, USA.,Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
83
|
Loss-of-function mutations of SCN10A encoding Na V1.8 α subunit of voltage-gated sodium channel in patients with human kidney stone disease. Sci Rep 2018; 8:10453. [PMID: 29992996 PMCID: PMC6041274 DOI: 10.1038/s41598-018-28623-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/26/2018] [Indexed: 01/18/2023] Open
Abstract
Human kidney stone disease (KSD) causes significant morbidity and public health burden worldwide. The etiology of KSD is heterogeneous, ranging from monogenic defects to complex interaction between genetic and environmental factors. However, the genetic defects causing KSD in the majority of affected families are still unknown. Here, we report the discovery of mutations of SCN10A, encoding NaV1.8 α subunit of voltage-gated sodium channel, in families with KSD. The region on chromosome 3 where SCN10A locates was initially identified in a large family with KSD by genome-wide linkage analysis and exome sequencing. Two mutations (p.N909K and p.K1809R) in the same allele of SCN10A co-segregated with KSD in the affected family. Additional mutation (p.V1149M) of SCN10A was identified in another affected family, strongly supporting the causal role of SCN10A for KSD. The amino acids at these three positions, N909, K1809, and V1149, are highly conserved in vertebrate evolution, indicating their structural and functional significances. NaV1.8 α subunit mRNA and protein were found to express in human kidney tissues. The mutant proteins expressed in cultured cells were unstable and causing reduced current density as analyzed by whole-cell patch-clamp technique. Thus, loss-of-function mutations of SCN10A were associated with KSD in the families studied.
Collapse
|
84
|
Di Diego JM, Antzelevitch C. J wave syndromes as a cause of malignant cardiac arrhythmias. Pacing Clin Electrophysiol 2018; 41:684-699. [PMID: 29870068 PMCID: PMC6281786 DOI: 10.1111/pace.13408] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/05/2018] [Indexed: 12/19/2022]
Abstract
The J wave syndromes, including the Brugada (BrS) and early repolarization (ERS) syndromes, are characterized by the manifestation of prominent J waves in the electrocardiogram appearing as an ST segment elevation and the development of life-threatening cardiac arrhythmias. BrS and ERS differ with respect to the magnitude and lead location of abnormal J waves and are thought to represent a continuous spectrum of phenotypic expression termed J wave syndromes. Despite over 25 years of intensive research, risk stratification and the approach to therapy of these two inherited cardiac arrhythmia syndromes are still rapidly evolving. Our objective in this review is to provide an integrated synopsis of the clinical characteristics, risk stratifiers, as well as the molecular, ionic, cellular, and genetic mechanisms underlying these two syndromes that have captured the interest and attention of the cardiology community over the past two decades.
Collapse
Affiliation(s)
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood PA
- Lankenau Heart Institute, Wynnewood, PA
- Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia PA
| |
Collapse
|
85
|
Sieira J, Brugada P. The definition of the Brugada syndrome. Eur Heart J 2018; 38:3029-3034. [PMID: 29020354 DOI: 10.1093/eurheartj/ehx490] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/07/2017] [Indexed: 12/30/2022] Open
Abstract
Brugada syndrome (BS) is an inherited disease characterized by a coved-type ST-segment elevation in the right precordial leads and increased risk of sudden cardiac death (SCD), in the absence of structural abnormalities. The cornerstone of BS diagnosis and definition, is its characteristic ECG pattern that can be present spontaneously or unmasked by drugs. Brugada syndrome was first described 25 years ago; paradoxically, in an era of great technological development, a new syndrome was described with a technology developed almost a century before. Great scientific knowledge has been gathered since the description of the syndrome. The better understanding of its pathophysiology and genetic basis has led to several modifications in its definition. Despite these facts, the essential, the description of the specific ECG pattern has remained almost unchanged since the initial report. In this article, we present the definition of the BS, the rationale behind it and our thoughts about its future.
Collapse
Affiliation(s)
- Juan Sieira
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel, Postgraduate program in Cardiac Electrophysiology and Pacing, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pedro Brugada
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel, Postgraduate program in Cardiac Electrophysiology and Pacing, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
86
|
Abstract
INTRODUCTION Brugada syndrome (BrS) is an inherited cardiac arrhythmia syndrome characterized by ST-segment elevation in right precordial ECG leads and associated with sudden cardiac death in young adults. The ECG manifestations of BrS are often concealed but can be unmasked by sodium channel blockers and fever. Areas covered: Implantation of a cardioverter defibrillator (ICD) is first-line therapy for BrS patients presenting with prior cardiac arrest or documented VT. A pharmacological approach to therapy is recommended in cases of electrical storm, as an adjunct to ICD and as preventative therapy. The goal of pharmacological therapy is to produce an inward shift to counter the genetically-induced outward shift of ion channel current flowing during the early phases of the ventricular epicardial action potential. This is accomplished by augmentation of ICa using □□adrenergic agents or phosphodiesterase III inhibitors or via inhibition of Ito. Radiofrequency ablation of the right ventricular outward flow tract epicardium is effective in suppressing arrhythmogenesis in BrS patients experiencing frequent appropriate ICD-shocks. Expert commentary: Understanding of the pathophysiology and approach to therapy of BrS has advanced considerably in recent years, but there remains an urgent need for development of cardio-selective and ion-channel-specific Ito blockers for treatment of BrS.
Collapse
Affiliation(s)
- Mariana Argenziano
- a Cardiovascular Research , Lankenau Institute for Medical Research , Wynnewood , PA , USA
| | - Charles Antzelevitch
- a Cardiovascular Research , Lankenau Institute for Medical Research , Wynnewood , PA , USA.,b Cardiovascular Research , Lankenau Heart Institute , Wynnewood , PA , USA.,c Department of Medicine and Pharmacology and Experimental Therapeutics , Sidney Kimmel Medical College of Thomas Jefferson University , Philadelphia , PA , USA
| |
Collapse
|
87
|
SCN10A-Dependent Late I
Na
Current. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018; 11:e002167. [DOI: 10.1161/circgen.118.002167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
88
|
Macri V, Brody JA, Arking DE, Hucker WJ, Yin X, Lin H, Mills RW, Sinner MF, Lubitz SA, Liu CT, Morrison AC, Alonso A, Li N, Fedorov VV, Janssen PM, Bis JC, Heckbert SR, Dolmatova EV, Lumley T, Sitlani CM, Cupples LA, Pulit SL, Newton-Cheh C, Barnard J, Smith JD, Van Wagoner DR, Chung MK, Vlahakes GJ, O'Donnell CJ, Rotter JI, Margulies KB, Morley MP, Cappola TP, Benjamin EJ, Muzny D, Gibbs RA, Jackson RD, Magnani JW, Herndon CN, Rich SS, Psaty BM, Milan DJ, Boerwinkle E, Mohler PJ, Sotoodehnia N, Ellinor PT. Common Coding Variants in SCN10A Are Associated With the Nav1.8 Late Current and Cardiac Conduction. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e001663. [PMID: 29752399 PMCID: PMC6377236 DOI: 10.1161/circgen.116.001663] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/02/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Genetic variants at the SCN5A/SCN10A locus are strongly associated with electrocardiographic PR and QRS intervals. While SCN5A is the canonical cardiac sodium channel gene, the role of SCN10A in cardiac conduction is less well characterized. METHODS We sequenced the SCN10A locus in 3699 European-ancestry individuals to identify variants associated with cardiac conduction, and replicated our findings in 21,000 individuals of European ancestry. We examined association with expression in human atrial tissue. We explored the biophysical effect of variation on channel function using cellular electrophysiology. RESULTS We identified 2 intronic single nucleotide polymorphisms in high linkage disequilibrium (r 2=0.86) with each other to be the strongest signals for PR (rs10428132, β=-4.74, P=1.52×10-14) and QRS intervals (rs6599251, QRS β=-0.73; P=1.2×10-4), respectively. Although these variants were not associated with SCN5A or SCN10A expression in human atrial tissue (n=490), they were in high linkage disequilibrium (r 2≥0.72) with a common SCN10A missense variant, rs6795970 (V1073A). In total, we identified 7 missense variants, 4 of which (I962V, P1045T, V1073A, and L1092P) were associated with cardiac conduction. These 4 missense variants cluster in the cytoplasmic linker of the second and third domains of the SCN10A protein and together form 6 common haplotypes. Using cellular electrophysiology, we found that haplotypes associated with shorter PR intervals had a significantly larger percentage of late current compared with wild-type (I962V+V1073A+L1092P, 20.2±3.3%, P=0.03, and I962V+V1073A, 22.4±0.8%, P=0.0004 versus wild-type 11.7±1.6%), and the haplotype associated with the longest PR interval had a significantly smaller late current percentage (P1045T, 6.4±1.2%, P=0.03). CONCLUSIONS Our findings suggest an association between genetic variation in SCN10A, the late sodium current, and alterations in cardiac conduction.
Collapse
Affiliation(s)
- Vincenzo Macri
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., J.C.B., S.R.H., C.M.S., N.S.)
| | - Dan E Arking
- University of Washington, Seattle. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (D.E.A.)
| | - William J Hucker
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Cardiac Arrhythmia Service (W.J.H., S.A.L., D.J.M., P.T.E.)
| | - Xiaoyan Yin
- Massachusetts General Hospital, Boston. NHLBI's & Boston University's Framingham Heart Study, MA (X.Y., H.L., L.A.C.)
- Department of Biostatistics (X.Y., L.A.C., C.-T.L.)
| | - Honghuang Lin
- Massachusetts General Hospital, Boston. NHLBI's & Boston University's Framingham Heart Study, MA (X.Y., H.L., L.A.C.)
- School of Public Health, Boston University, MA. Computational Biomedicine Section (H.L.)
| | - Robert W Mills
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
| | - Moritz F Sinner
- Department of Medicine, Boston University School of Medicine, MA. German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Germany and Department of Medicine I, University Hospital Munich, Ludwig-Maximilian's University, Munich, Germany (M.F.S.)
| | - Steven A Lubitz
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Cardiac Arrhythmia Service (W.J.H., S.A.L., D.J.M., P.T.E.)
| | - Ching-Ti Liu
- Department of Biostatistics (X.Y., L.A.C., C.-T.L.)
| | - Alanna C Morrison
- Human Genetics Center, University of Texas Health Science Center at Houston (A.C.M., E.B.)
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Ning Li
- Department of Physiology & Cell Biology and Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (N.L., V.V.F., P.M.J., P.J.M.)
| | - Vadim V Fedorov
- Department of Physiology & Cell Biology and Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (N.L., V.V.F., P.M.J., P.J.M.)
| | - Paul M Janssen
- Department of Physiology & Cell Biology and Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (N.L., V.V.F., P.M.J., P.J.M.)
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., J.C.B., S.R.H., C.M.S., N.S.)
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., J.C.B., S.R.H., C.M.S., N.S.)
- Department of Epidemiology (S.R.H., T.L.)
| | - Elena V Dolmatova
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., J.C.B., S.R.H., C.M.S., N.S.)
| | - L Adrienne Cupples
- Massachusetts General Hospital, Boston. NHLBI's & Boston University's Framingham Heart Study, MA (X.Y., H.L., L.A.C.)
- Department of Biostatistics (X.Y., L.A.C., C.-T.L.)
| | - Sara L Pulit
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Department of Statistics, University of Auckland, New Zealand (S.L.P.)
| | - Christopher Newton-Cheh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Center for Genomic Medicine (C.N.-C.)
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA (C.N.-C.)
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute (J.B.)
| | - Jonathan D Smith
- Department of Cardiovascular Medicine, Heart and Vascular Institute (J.D.S., D.R.V.W., M.K.C.)
- Department of Cellular and Molecular Medicine Biology, Lerner Research Institute (J.D.S.)
| | - David R Van Wagoner
- Department of Cardiovascular Medicine, Heart and Vascular Institute (J.D.S., D.R.V.W., M.K.C.)
- Department of Molecular Cardiology, Lerner Research Institute (D.R.V.W., M.K.C.)
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart and Vascular Institute (J.D.S., D.R.V.W., M.K.C.)
- Department of Molecular Cardiology, Lerner Research Institute (D.R.V.W., M.K.C.)
| | | | | | - Jerome I Rotter
- Cleveland Clinic, OH. Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute & Department of Pediatrics, Harbor-UCLA Medical Center, Torrance (J.I.R.)
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
- Department of Medicine, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
| | - Michael P Morley
- Penn Cardiovascular Institute, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
- Department of Medicine, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
| | - Thomas P Cappola
- Penn Cardiovascular Institute, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
- Department of Medicine, Perelman School of Medicine (K.B.M., M.P.M., T.P.C.)
| | - Emelia J Benjamin
- Department of Epidemiology (E.J.B.)
- Preventive Medicine Section (E.J.B.)
- Cardiology Section (E.J.B.)
| | - Donna Muzny
- University of Pennsylvania, Philadelphia. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (D.M.M., R.A.G., E.B.)
| | - Richard A Gibbs
- University of Pennsylvania, Philadelphia. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (D.M.M., R.A.G., E.B.)
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes and Metabolism, College of Medicine, The Ohio State University, Columbus (R.D.J.)
| | - Jared W Magnani
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute (J.W.M.)
| | - Caroline N Herndon
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (C.N.H., P.T.E.)
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville (S.S.R.)
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle; and Kaiser Permanente Washington Health Research Institute, Seattle, WA. (B.M.P.)
| | - David J Milan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Cardiac Arrhythmia Service (W.J.H., S.A.L., D.J.M., P.T.E.)
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston (A.C.M., E.B.)
- University of Pennsylvania, Philadelphia. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (D.M.M., R.A.G., E.B.)
| | - Peter J Mohler
- Department of Physiology & Cell Biology and Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (N.L., V.V.F., P.M.J., P.J.M.)
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., J.C.B., S.R.H., C.M.S., N.S.)
- Division of Cardiology (N.S.)
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown (V.M., W.J.H., R.W.M., S.A.L., E.V.D., S.L.P., C.N.-C., D.J.M., P.T.E.)
- Cardiac Arrhythmia Service (W.J.H., S.A.L., D.J.M., P.T.E.)
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (C.N.H., P.T.E.)
| |
Collapse
|
89
|
Abou Ziki MD, Seidelmann SB, Smith E, Atteya G, Jiang Y, Fernandes RG, Marieb MA, Akar JG, Mani A. Deleterious protein-altering mutations in the SCN10A voltage-gated sodium channel gene are associated with prolonged QT. Clin Genet 2018; 93:741-751. [PMID: 28407228 PMCID: PMC5640462 DOI: 10.1111/cge.13036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/27/2017] [Accepted: 04/09/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Long QT syndrome (LQT) is a pro-arrhythmogenic condition with life-threatening complications. Fifteen genes have been associated with congenital LQT, however, the genetic causes remain unknown in more than 20% of cases. MATERIALS AND METHODS Eighteen patients with history of palpitations, pre-syncope, syncope and prolonged QT were referred to the Yale Cardiovascular Genetics Program. All subjects underwent whole-exome sequencing (WES) followed by confirmatory Sanger sequencing. Mutation burden analysis was carried out using WES data from 16 subjects with no identifiable cause of LQT. RESULTS Deleterious and novel SCN10A mutations were identified in 3 of the 16 patients (19%) with idiopathic LQT. These included 2 frameshifts and 1 missense variants (p.G810fs, p.R1259Q, and p.P1877fs). Further analysis identified 2 damaging SCN10A mutations with allele frequencies of approximately 0.2% (p.R14L and p.R1268Q) in 2 independent cases. None of the SCN10A mutation carriers had mutations in known arrhythmia genes. Damaging SCN10A mutations (p.R209H and p.R485C) were also identified in the 2 subjects on QT prolonging medications. CONCLUSION Our findings implicate SCN10A in LQT. The presence of frameshift mutations suggests loss-of-function as the underlying disease mechanism. The common association with atrial fibrillation suggests a unique mechanism of disease for this LQT gene.
Collapse
Affiliation(s)
- Maen D. Abou Ziki
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Emily Smith
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Gourg Atteya
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Yuexin Jiang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Rodolfo Gil Fernandes
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Mark A. Marieb
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Joseph G. Akar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Deparetment of Genetics, Yale University School of Medicine, New Haven, CT, 06510
| |
Collapse
|
90
|
Sonoda K, Ohno S, Ozawa J, Hayano M, Hattori T, Kobori A, Yahata M, Aburadani I, Watanabe S, Matsumoto Y, Makiyama T, Horie M. Copy number variations of SCN5A in Brugada syndrome. Heart Rhythm 2018; 15:1179-1188. [PMID: 29574140 DOI: 10.1016/j.hrthm.2018.03.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Loss-of-function mutations in SCN5A are associated in ∼20% of Brugada syndrome (BrS) patients. Copy number variations (CNVs) have been shown to be associated with several inherited arrhythmia syndromes. OBJECTIVE The purpose of this study was to investigate SCN5A CNVs among BrS probands. METHODS The study cohort consisted of 151 BrS probands who were symptomatic or had a family history of BrS, sudden death, syncope, or arrhythmic diseases. We performed sequence analysis of SCN5A by the Sanger method. For detecting CNVs in SCN5A, we performed multiplex ligation-dependent probe amplification analysis of the 151 BrS probands. RESULTS We identified pathogenic SCN5A mutations in 20 probands by the Sanger method. In 140 probands in whom multiplex ligation-dependent probe amplification was successfully performed, 4 probands were found to present different CNVs (deletion in 3 and duplication in 1). Three of them had fatal arrhythmia events; the remaining 1 was asymptomatic but had a family history. Mean age at diagnosis was 23 ± 14 years. All of the baseline 12-lead electrocardiograms showed PQ-interval prolongation. The characteristics of these 4 probands with CNVs were similar to those of the probands with mutations leading to premature truncation of the protein or missense mutations causing peak INa reduction >90%. CONCLUSION We identified SCN5A CNVs in 2.9% of BrS probands who were symptomatic or had a family history.
Collapse
Affiliation(s)
- Keiko Sonoda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Molecular Biology, National Cerebral and Cardiovacular Center, Suita, Japan
| | - Seiko Ohno
- Department of Molecular Biology, National Cerebral and Cardiovacular Center, Suita, Japan; Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan; Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Junichi Ozawa
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mamoru Hayano
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tetsuhisa Hattori
- Department of Molecular Biology, National Cerebral and Cardiovacular Center, Suita, Japan; Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Atsushi Kobori
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Mitsuhiko Yahata
- Department of Cardiovascular Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Isao Aburadani
- Department of Cardiovascular Medicine, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Seiichi Watanabe
- Department of Pediatrics, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Yuichi Matsumoto
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Takeru Makiyama
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
91
|
Abstract
INTRODUCTION AND OBJECTIVES The importance of sodium channels for the normal electrical activity of the heart is emphasized by the fact that mutations (inherited or de novo) in genes that encode for these channels or their associated proteins cause arrhythmogenic syndromes such as the Brugada syndrome and the long QT syndrome (LQTS). The aim of this study is to conduct a review of the literature on the mutations in the sodium channel complex responsible for heart disease and the implications of a close relationship between genetics and the clinical aspects of the main cardiac channelopathies, namely at the level of diagnosis, risk stratification, prognosis, screening of family members and treatment. METHODS The online Pubmed® database was used to search for articles published in this field in indexed journals. The MeSH database was used to define the following query: "Mutation [Mesh] AND Sodium Channels [Mesh] AND Heart Diseases [Mesh]", and articles published in the last 15 years, written in English or Portuguese and referring to research in human beings were included. CONCLUSIONS In the past few years, significant advances have been made to clarify the genetic and molecular basis of these syndromes. A greater understanding of the underlying pathophysiological mechanisms showed the importance of the relationship between genotype and phenotype and led to progress in the clinical approach to these patients. However, it is still necessary to improve diagnostic capacity, optimize risk stratification, and develop new specific treatments according to the genotype-phenotype binomial.
Collapse
|
92
|
Asvestas D, Tse G, Baranchuk A, Bazoukis G, Liu T, Saplaouras A, Korantzopoulos P, Goga C, Efremidis M, Sideris A, Letsas KP. High risk electrocardiographic markers in Brugada syndrome. IJC HEART & VASCULATURE 2018; 18:58-64. [PMID: 29876505 PMCID: PMC5988483 DOI: 10.1016/j.ijcha.2018.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022]
Abstract
Several clinical, electrocardiographic (ECG) and electrophysiological markers have been proposed to provide optimal risk stratification in patients with Brugada syndrome (BrS). Of the different markers, only a spontaneous type 1 ECG pattern has clearly shown a sufficiently high predictive value. This review article highlights specific ECG markers based on depolarization and/or repolarization that have been associated with an increased risk of arrhythmic events in patients with BrS.
Collapse
Affiliation(s)
- Dimitrios Asvestas
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | - Gary Tse
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR, PR China
- Li Ka Shing Institute of Health Sciences, 30-32 Ngan Shing St, Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Adrian Baranchuk
- Division of Cardiology, Queen's University, Kingston General Hospital, Kingston, Ontario, Canada
| | - George Bazoukis
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | - Tong Liu
- Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, PR China
| | - Athanasios Saplaouras
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | | | - Christina Goga
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | - Michael Efremidis
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | - Antonios Sideris
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| | - Konstantinos P. Letsas
- Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Greece
| |
Collapse
|
93
|
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
|
94
|
Fonseca DJ, Vaz da Silva MJ. Cardiac channelopathies: The role of sodium channel mutations. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.repce.2017.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
95
|
Munroe PB, Addison S, Abrams DJ, Sebire NJ, Cartwright J, Donaldson I, Cohen MM, Mein C, Tinker A, Harmer SC, Aziz Q, Terry A, Struebig M, Warren HR, Vadgama B, Fowler DJ, Peebles D, Taylor AM, Lally PJ, Thayyil S. Postmortem Genetic Testing for Cardiac Ion Channelopathies in Stillbirths. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018; 11:e001817. [PMID: 29874177 DOI: 10.1161/circgen.117.001817] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 11/07/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although stillbirth is a significant health problem worldwide, the definitive cause of death remains elusive in many cases, despite detailed autopsy. In this study of partly explained and unexplained stillbirths, we used next-generation sequencing to examine an extended panel of 35 candidate genes known to be associated with ion channel disorders and sudden cardiac death. METHODS AND RESULTS We examined tissue from 242 stillbirths (≥22 weeks), including those where no definite cause of death could be confirmed after a full autopsy. We obtained high-quality DNA from 70 cases, which were then sequenced for a custom panel of 35 genes, 12 for inherited long- and short-QT syndrome genes (LQT1-LQT12 and SQT1-3), and 23 additional candidate genes derived from genome-wide association studies. We examined the functional significance of a selected variant by patch-clamp electrophysiological recording. No predicted damaging variants were identified in KCNQ1 (LQT1) or KCNH2 (LQT2). A rare putative pathogenic variant was found in KCNJ2(LQT7) in 1 case, and several novel variants of uncertain significance were observed. The KCNJ2 variant (p. R40Q), when assessed by whole-cell patch clamp, affected the function of the channel. There was no significant evidence of enrichment of rare predicted damaging variants within any of the candidate genes. CONCLUSIONS Although a causative link is unclear, 1 putative pathogenic and variants of uncertain significance variant resulting in cardiac channelopathies was identified in some cases of otherwise unexplained stillbirth, and these variants may have a role in fetal demise. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01120886.
Collapse
Affiliation(s)
- Patricia B Munroe
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.).
| | - Shea Addison
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Dominic J Abrams
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Neil J Sebire
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - James Cartwright
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Ian Donaldson
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Marta M Cohen
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Charles Mein
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Andrew Tinker
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Stephen C Harmer
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Qadeer Aziz
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Anna Terry
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Monika Struebig
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Helen R Warren
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Bhumita Vadgama
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Darren J Fowler
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Donald Peebles
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Andrew M Taylor
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Peter J Lally
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.)
| | - Sudhin Thayyil
- From the Clinical Pharmacology (P.B.M., S.A., J.C., A.T., S.C.H., Q.A., H.R.W.) and National Institute for Health Research Barts Cardiovascular Biomedical Research Unit (P.B.M., A.T., H.R.W.), William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, United Kingdom; Genome Centre, Queen Mary University of London, United Kingdom (I.D., C.M., A.T., M.S., B.V.); Centre for Perinatal Neuroscience, Imperial College London, United Kingdom (S.A., P.J.L., S.T.); Paediatric Cardiology, Children's Hospital Boston, MA (D.J.A.); Histopathology, Great Ormond Street Hospital, London, United Kingdom (N.J.S.); Histopathology, Sheffield Children's Hospital, United Kingdom (M.M.C.); Histopathology, Southampton General Hospital, United Kingdom (D.J.F.); Institute for Women's Health, San Antonio, TX (D.P.); and Institute for Cardiovascular Science, University College London, United Kingdom (A.M.T.).
| |
Collapse
|
96
|
Abstract
Pain is an increasing clinical challenge affecting about half the population, with a substantial number of people suffering daily intense pain. Such suffering can be linked to the dramatic rise in opioid use and associated deaths in the United States. There is a pressing need for new analgesics with limited side effects. Here, we summarize what we know about the genetics of pain and implications for drug development. We make the case that chronic pain is not one but a set of disease states, with peripheral drive a key element in most. We argue that understanding redundancy and plasticity, hallmarks of the nervous system, is critical in developing analgesic drug strategies. We describe the exploitation of monogenic pain syndromes and genetic association studies to define analgesic targets, as well as issues associated with animal models of pain. We appraise present-day screening technologies and describe recent approaches to pain treatment that hold promise.
Collapse
Affiliation(s)
- Jane E Sexton
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom;
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom;
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom;
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
97
|
Savio-Galimberti E, Argenziano M, Antzelevitch C. Cardiac Arrhythmias Related to Sodium Channel Dysfunction. Handb Exp Pharmacol 2018; 246:331-354. [PMID: 28965168 DOI: 10.1007/164_2017_43] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The voltage-gated cardiac sodium channel (Nav1.5) is a mega-complex comprised of a pore-forming α subunit and 4 ancillary β-subunits together with numerous protein partners. Genetic defects in the form of rare variants in one or more sodium channel-related genes can cause a loss- or gain-of-function of sodium channel current (INa) leading to the manifestation of various disease phenotypes, including Brugada syndrome, long QT syndrome, progressive cardiac conduction disease, sick sinus syndrome, multifocal ectopic Purkinje-related premature contractions, and atrial fibrillation. Some sodium channelopathies have also been shown to be responsible for sudden infant death syndrome (SIDS). Although these genetic defects often present as pure electrical diseases, recent studies point to a contribution of structural abnormalities to the electrocardiographic and arrhythmic manifestation in some cases, such as dilated cardiomyopathy. The same rare variants in SCN5A or related genes may present with different clinical phenotypes in different individuals and sometimes in members of the same family. Genetic background and epigenetic and environmental factors contribute to the expression of these overlap syndromes. Our goal in this chapter is to review and discuss what is known about the clinical phenotype and genotype of each cardiac sodium channelopathy, and to briefly discuss the underlying mechanisms.
Collapse
Affiliation(s)
| | - Mariana Argenziano
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA.
| |
Collapse
|
98
|
Massively Parallel Sequencing of Genes Implicated in Heritable Cardiac Disorders: A Strategy for a Small Diagnostic Laboratory. Med Sci (Basel) 2017; 5:medsci5040022. [PMID: 29099038 PMCID: PMC5753651 DOI: 10.3390/medsci5040022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/20/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022] Open
Abstract
Sudden cardiac death (SCD) in people before the age of 35 years is a devastating event for any family. The causes of SCD in the young can be broadly divided into two groups: heritable cardiac disorders that affect the heart structure (cardiomyopathies) and primary electrical disorders (cardiac ion channelopathies). Genetic testing is vital as those suffering from cardiac ion channelopathies have structurally normal hearts, and those with cardiomyopathies may only show subtle abnormalities in the heart and these signs may not be detected during an autopsy. Post-mortem genetic testing of SCD victims is important to identify the underlying genetic cause. This is important as family cascade screening may be undertaken to identify those who may be at risk and provide vital information about risk stratification and clinical management. The development of massively parallel sequencing (MPS) has made it possible for the simultaneous screening of multiple patients for hundreds of genes. In light of this, we opted to develop an MPS approach for SCD analysis that would allow us to screen for mutations in genes implicated in cardiomyopathies and cardiac ion channelopathies. The rationale behind this panel was to limit it to genes carrying the greatest mutation load. If no likely pathogenic gene variant were found then testing could cascade to whole exome/genome sequencing as a gene-discovery exercise. The overarching aim was to design and validate a custom-cardiac panel that satisfies the diagnostic requirements of LabPLUS (Auckland City Hospital, Auckland, NZ) and the guidelines provided by the Royal College of Pathologists of Australasia and the Association for Clinical Genetic Science.
Collapse
|
99
|
Abstract
Brugada syndrome (BrS) is one of the commonest inherited primary arrhythmia syndromes typically presenting with arrhythmic syncope or sudden cardiac death (SCD) due to polymorphic ventricular tachycardia and ventricular fibrillation precipitated by vagotonia or fever in apparently healthy adults, less frequently in children. The prevalence of the syndrome (0.01%-0.3%) varies among regions and ethnicities, being the highest in Southeast Asia. BrS is diagnosed by the "coved type" ST-segment elevation≥2mm followed by a negative T-wave in ≥1 of the right precordial leads V1-V2. The typical electrocardiogram in BrS is often concealed by fluctuations between normal, non-diagnostic and diagnostic ST-segment pattern in the same patient, thus hindering the diagnosis. Presently, the majority of BrS patients is incidentally diagnosed, and may remain asymptomatic for their lifetime. However, BrS is responsible for 4-12% of all SCDs and for ~20% of SCDs in patients with structurally normal hearts. Arrhythmic risk is the highest in SCD survivors and in patients with spontaneous BrS electrocardiogram and arrhythmic syncope, but risk stratification for SCD in asymptomatic subjects has not yet been fully defined. Recent achievements have expanded our understanding of the genetics and electrophysiological mechanisms underlying BrS, while radiofrequency catheter ablation may be an effective new approach to treat ventricular tachyarrhythmias in BrS patients with arrhythmic storms. The present review summarizes our contemporary understanding and recent advances in the inheritance, pathophysiology, clinical assessment and treatment of BrS patients.
Collapse
Affiliation(s)
- Marija M Polovina
- School of Medicine, Belgrade University, Belgrade, Serbia; Cardiology Clinic, Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Bojan Banko
- Centre for Radiology and MRI, Clinical Centre of Serbia, Belgrade, Serbia
| | - Gregory Y H Lip
- School of Medicine, Belgrade University, Belgrade, Serbia; University of Birmingham, Institute of Cardiovascular Science, City Hospital, Birmingham, United Kingdom
| | - Tatjana S Potpara
- School of Medicine, Belgrade University, Belgrade, Serbia; Cardiology Clinic, Clinical Centre of Serbia, Belgrade, Serbia.
| |
Collapse
|
100
|
Ghouse J, Skov MW, Bigseth RS, Ahlberg G, Kanters JK, Olesen MS. Distinguishing pathogenic mutations from background genetic noise in cardiology: The use of large genome databases for genetic interpretation. Clin Genet 2017; 93:459-466. [PMID: 28589536 DOI: 10.1111/cge.13066] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
Advances in clinical genetic testing have led to increased insight into the human genome, including how challenging it is to interpret rare genetic variation. In some cases, the ability to detect genetic mutations exceeds the ability to understand their clinical impact, limiting the advantage of these technologies. Obstacles in genomic medicine are many and include: understanding the level of certainty/uncertainty behind pathogenicity determination, the numerous different variant interpretation-guidelines used by clinical laboratories, delivering the certain or uncertain result to the patient, helping patients evaluate medical decisions in light of uncertainty regarding the consequence of the findings. Through publication of large publicly available exome/genome databases, researchers and physicians are now able to highlight dubious variants previously associated with different cardiac traits. Also, continuous efforts through data sharing, international collaborative efforts to develop disease-gene-specific guidelines, and computational analyses using large data, will indubitably assist in better variant interpretation and classification. This article discusses the current, and quickly changing, state of variant interpretation resources within cardiovascular genetic research, e.g., publicly available databases and ways of how cardiovascular genetic counselors and geneticists can aid in improving variant interpretation in cardiology.
Collapse
Affiliation(s)
- J Ghouse
- Laboratory of Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - M W Skov
- Laboratory of Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - R S Bigseth
- Laboratory of Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - G Ahlberg
- Laboratory of Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - J K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M S Olesen
- Laboratory of Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|