1
|
Comollo TW, Zou X, Zhang C, Kesters D, Hof T, Sampson KJ, Kass RS. Exploring mutation specific beta blocker pharmacology of the pathogenic late sodium channel current from patient-specific pluripotent stem cell myocytes derived from long QT syndrome mutation carriers. Channels (Austin) 2022; 16:173-184. [PMID: 35949058 PMCID: PMC9373745 DOI: 10.1080/19336950.2022.2106025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The congenital long QT syndrome (LQTS), one of the most common cardiac channelopathies, is characterized by delayed ventricular repolarization underlying prolongation of the QT interval of the surface electrocardiogram. LQTS is caused by mutations in genes coding for cardiac ion channels or ion channel-associated proteins. The major therapeutic approach to LQTS management is beta blocker therapy which has been shown to be effective in treatment of LQTS variants caused by mutations in K+ channels. However, this approach has been questioned in the treatment of patients identified as LQTS variant 3(LQT3) patients who carry mutations in SCN5A, the gene coding for the principal cardiac Na+ channel. LQT3 mutations are gain of function mutations that disrupt spontaneous Na+ channel inactivation and promote persistent or late Na+ channel current (INaL) that delays repolarization and underlies QT prolongation. Clinical investigation of patients with the two most common LQT3 mutations, the ΔKPQ and the E1784K mutations, found beta blocker treatment a useful therapeutic approach for managing arrhythmias in this patient population. However, there is little experimental data that reveals the mechanisms underlying these antiarrhythmic actions. Here, we have investigated the effects of the beta blocker propranolol on INaL expressed by ΔKPQ and E1784K channels in induced pluripotent stem cells derived from patients carrying these mutations. Our results indicate that propranolol preferentially inhibits INaL expressed by these channels suggesting that the protective effects of propranolol in treating LQT3 patients is due in part to modulation of INaL.
Collapse
Affiliation(s)
- Thomas W. Comollo
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Xinle Zou
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Chuangeng Zhang
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Divya Kesters
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Thomas Hof
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Kevin J. Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Robert S. Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA,CONTACT Robert S. Kass
| |
Collapse
|
2
|
Yang Q, Berkman AM, Ezekian JE, Rosamilia M, Rosenfeld JA, Liu P, Landstrom AP. Determining the Likelihood of Disease Pathogenicity Among Incidentally Identified Genetic Variants in Rare Dilated Cardiomyopathy-Associated Genes. J Am Heart Assoc 2022; 11:e025257. [PMID: 36129056 DOI: 10.1161/jaha.122.025257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background As utilization of clinical exome sequencing (ES) has expanded, criteria for evaluating the diagnostic weight of incidentally identified variants are critical to guide clinicians and researchers. This is particularly important in genes associated with dilated cardiomyopathy (DCM), which can cause heart failure and sudden death. We sought to compare the frequency and distribution of incidentally identified variants in DCM-associated genes between a clinical referral cohort with those in control and known case cohorts to determine the likelihood of pathogenicity among those undergoing genetic testing for non-DCM indications. Methods and Results A total of 39 rare, non-TTN DCM-associated genes were identified and evaluated from a clinical ES testing referral cohort (n=14 005, Baylor Genetic Laboratories) and compared with a DCM case cohort (n=9442) as well as a control cohort of population variants (n=141 456) derived from the gnomAD database. Variant frequencies in each cohort were compared. Signal-to-noise ratios were calculated comparing the DCM and ES cohort with the gnomAD cohort. The likely pathogenic/pathogenic variant yield in the DCM cohort (8.2%) was significantly higher than in the ES cohort (1.9%). Based on signal-to-noise and correlation analysis, incidental variants found in FLNC, RBM20, MYH6, DSP, ABCC9, JPH2, and NEXN had the greatest chance of being DCM-associated. Conclusions The distribution of pathogenic variants between the ES cohort and the DCM case cohort was gene specific, and variants found in the ES cohort were similar to variants found in the control cohort. Incidentally identified variants in specific genes are more associated with DCM than others.
Collapse
Affiliation(s)
- Qixin Yang
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC.,Department of Cardiology The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou China
| | - Amy M Berkman
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Jordan E Ezekian
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Michael Rosamilia
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics Baylor College of Medicine and Baylor Genetics Laboratories Houston TX
| | - Pengfei Liu
- Department of Molecular and Human Genetics Baylor College of Medicine and Baylor Genetics Laboratories Houston TX
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
3
|
Kim WC, Lemire E, Nosib S, Nosib S. A Novel Frameshift Mutation, KCNH2 [p.Asp896ArgfsX79], Leading to Malignant Ventricular Arrhythmia, Identified After Treatment of Gastrointestinal Bleeding. CJC Open 2021; 3:1383-1387. [PMID: 34901807 PMCID: PMC8640589 DOI: 10.1016/j.cjco.2021.06.005] [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: 03/17/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
A novel frameshift mutation in the KCNH2 gene for long QT syndrome type 2 (LQTS2) was identified after torsades des pointes ventricular tachycardia in a 49-year-old patient managed with octreotide and nadolol for an acute variceal bleed. In spite of removal of offending medications, and correction of underlying electrolyte abnormalities, the patient’s QT interval remained prolonged—at 521 ms—raising the suspicion of an underlying channelopathy. Genetic studies confirmed heterozygosity for a novel frameshift mutation for the KCNH2 gene, D896Rfs X79. We explore the pathogenicity and clinical impact of this variant mutation.
Collapse
Affiliation(s)
- Wan Cheol Kim
- Department of Medicine, University of Dalhousie, Halifax, Nova Scotia, Canada
| | - Edmond Lemire
- Division of Medical Genetics, University of Saskatchewan, Royal University Hospital, Saskatoon, Saskatchewan, Canada
| | - Siddarth Nosib
- College of Medicine, University of Central Lancashire, Preston, United Kingdom
| | - Shravankumar Nosib
- Department of Medicine, University of Saskatchewan, Royal University Hospital, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
4
|
Correa A, Haider SW, Aronow WS. Precision medicine in cardiac electrophysiology: where we are and where we need to go. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:165-180. [DOI: 10.1080/23808993.2020.1754127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Ashish Correa
- Department of Cardiology, Mount Sinai Morningside (formerly Mount St. Luke’s), Icahn School of Medicine, New York, NY, USA
| | - Syed Waqas Haider
- Department of Cardiology, Mount Sinai Morningside (formerly Mount St. Luke’s), Icahn School of Medicine, New York, NY, USA
| | - Wilbert S. Aronow
- Department of Cardiology, Westchester Medical Center, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
5
|
Giudicessi JR, Ackerman MJ. Calcium Revisited: New Insights Into the Molecular Basis of Long-QT Syndrome. Circ Arrhythm Electrophysiol 2018; 9:CIRCEP.116.002480. [PMID: 27390209 DOI: 10.1161/circep.116.002480] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022]
Affiliation(s)
- John R Giudicessi
- From the Internal Medicine Residency and Clinician-Investigator Programs, Department of Medicine (J.R.G.) and Departments of Cardiovascular Diseases, Pediatrics (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (M.J.A.), Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN
| | - Michael J Ackerman
- From the Internal Medicine Residency and Clinician-Investigator Programs, Department of Medicine (J.R.G.) and Departments of Cardiovascular Diseases, Pediatrics (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (M.J.A.), Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN.
| |
Collapse
|
6
|
Duras E, İrdem A, Özkaya O. Long QT syndrome diagnosed in two sisters with propionic acidemia: a case report. J Pediatr Endocrinol Metab 2017; 30:1133-1136. [PMID: 28820736 DOI: 10.1515/jpem-2016-0469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 07/17/2017] [Indexed: 12/30/2022]
Abstract
Propionic acidemia (PA) is a rare autosomal recessive metabolic disorder caused by deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). This disorder mostly progresses with episodes of metabolic acidosis. Cardiomyopathy is among the cardiac complications known to occur during metabolic decompensation episodes. However, several recent papers emphasized the association of PA and long QT syndrome (LQTS) which may lead to extremely serious and fatal consequences. In this report, we describe two sisters with PA who have prolonged QT duration that were incidentally detected in an outpatient setting. LQTS was verified by electrocardiogram, stress test and 24 h rhythm holter monitoring. By this report, we want to emphasize the importance of early diagnosis of LQTS in asymptomatic patients with PA to prevent fatal complications.
Collapse
|
7
|
Fernández-Falgueras A, Sarquella-Brugada G, Brugada J, Brugada R, Campuzano O. Cardiac Channelopathies and Sudden Death: Recent Clinical and Genetic Advances. BIOLOGY 2017; 6:7. [PMID: 28146053 PMCID: PMC5372000 DOI: 10.3390/biology6010007] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
Sudden cardiac death poses a unique challenge to clinicians because it may be the only symptom of an inherited heart condition. Indeed, inherited heart diseases can cause sudden cardiac death in older and younger individuals. Two groups of familial diseases are responsible for sudden cardiac death: cardiomyopathies (mainly hypertrophic cardiomyopathy, dilated cardiomyopathy, and arrhythmogenic cardiomyopathy) and channelopathies (mainly long QT syndrome, Brugada syndrome, short QT syndrome, and catecholaminergic polymorphic ventricular tachycardia). This review focuses on cardiac channelopathies, which are characterized by lethal arrhythmias in the structurally normal heart, incomplete penetrance, and variable expressivity. Arrhythmias in these diseases result from pathogenic variants in genes encoding cardiac ion channels or associated proteins. Due to a lack of gross structural changes in the heart, channelopathies are often considered as potential causes of death in otherwise unexplained forensic autopsies. The asymptomatic nature of channelopathies is cause for concern in family members who may be carrying genetic risk factors, making the identification of these genetic factors of significant clinical importance.
Collapse
Affiliation(s)
| | | | - Josep Brugada
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona 08950, Spain.
| | - Ramon Brugada
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
- Familial Cardiomyopathies Unit, Hospital Josep Trueta, Girona 17007, Spain.
| | - Oscar Campuzano
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
| |
Collapse
|
8
|
Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ, Kass RS. Molecular Pathophysiology of Congenital Long QT Syndrome. Physiol Rev 2017; 97:89-134. [PMID: 27807201 PMCID: PMC5539372 DOI: 10.1152/physrev.00008.2016] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ion channels represent the molecular entities that give rise to the cardiac action potential, the fundamental cellular electrical event in the heart. The concerted function of these channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syndrome. We examine mechanisms of dysfunction associated with three critical cardiac currents that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the study of mutations that cause congenital Long QT Syndrome, the scientific community has advanced understanding of ion channel structure-function relationships, physiology, and pharmacological response to clinically employed and experimental pharmacological agents. Our understanding of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-driven clinical management of the disease has improved patient care as precision medicine becomes even more a reality.
Collapse
Affiliation(s)
- M S Bohnen
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - G Peng
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - S H Robey
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - C Terrenoire
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - V Iyer
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - K J Sampson
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - R S Kass
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| |
Collapse
|
9
|
Veltmann C, Barajas-Martinez H, Wolpert C, Borggrefe M, Schimpf R, Pfeiffer R, Cáceres G, Burashnikov E, Antzelevitch C, Hu D. Further Insights in the Most Common SCN5A Mutation Causing Overlapping Phenotype of Long QT Syndrome, Brugada Syndrome, and Conduction Defect. J Am Heart Assoc 2016; 5:JAHA.116.003379. [PMID: 27381756 PMCID: PMC5015375 DOI: 10.1161/jaha.116.003379] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Phenotypic overlap of type 3 long QT syndrome (LQT3), Brugada syndrome (BrS), cardiac conduction disease (CCD), and sinus node dysfunction (SND) is observed with SCN5A mutations. SCN5A‐E1784K is the most common mutation associated with BrS and LQTS3. The present study examines the genotype–phenotype relationship in a large family carrying SCN5A‐E1784K and SCN5A‐H558R polymorphism. Methods and Results Clinical work‐up, follow‐up, and genetic analysis were performed in 35 family members. Seventeen were SCN5A‐E1784K positive. They also displayed QTc prolongation, and either BrS, CCD, or both. One carrier exhibited SND. The presence of SCN5A‐H558R did not significantly alter the phenotype of SCN5A‐E1784K carriers. Fourteen SCN5A‐E1784K patients underwent implantable cardioverter‐defibrillator (ICD) implantation; 4 developed VF and received appropriate ICD shocks after 8±3 months of follow‐up. One patient without ICD also developed VF after 6.7 years. These 5 cases carried both SCN5A‐E1784K and SCN5A‐H558R. Functional characterization was achieved by expressing SCN5A variants in TSA201 cells. Peak (INa,P) or late (INa,L) sodium currents were recorded using whole‐cell patch‐clamp techniques. Co‐expression of SCN5A‐E1784K and SCN5A‐WT reduced INa,P to 70.03% of WT, shifted steady‐state inactivation by −11.03 mV, and increased INa,L from 0.14% to 1.86% of INa,P. Similar changes were observed when SCN5A‐E1784K was co‐expressed with SCN5A‐H558R. Conclusions We demonstrate a strong genotype‐phenotype correlation with complete penetrance for BrS, LQTS, or CCD in the largest family harboring SCN5A‐E1784K mutation described so far. Phenotype of LQTS is present during all decades of life, whereas CCD develops with increasing age. Phenotypic overlap may explain the high event rate in carriers.
Collapse
Affiliation(s)
- Christian Veltmann
- Rhythmology and Electrophysiology, Department of Cardiology and Angiology Hannover Medical School, Hannover, Germany
| | | | - Christian Wolpert
- Klinik für Innere Medizin, Cardiology Klinikum Ludwigsburg, Ludwigsburg, Germany
| | - Martin Borggrefe
- 1st Department of Medicine-Cardiology, University Medical Centre Mannheim, Mannheim, Germany DZHK partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Rainer Schimpf
- 1st Department of Medicine-Cardiology, University Medical Centre Mannheim, Mannheim, Germany DZHK partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Ryan Pfeiffer
- Molecular Genetic Department, Masonic Medical Research Laboratory, Utica, NY, USA
| | - Gabriel Cáceres
- Molecular Genetic Department, Masonic Medical Research Laboratory, Utica, NY, USA
| | - Elena Burashnikov
- Molecular Genetic Department, Masonic Medical Research Laboratory, Utica, NY, USA
| | | | - Dan Hu
- Molecular Genetic Department, Masonic Medical Research Laboratory, Utica, NY, USA
| |
Collapse
|
10
|
Spears DA, Gollob MH. Genetics of inherited primary arrhythmia disorders. APPLICATION OF CLINICAL GENETICS 2015; 8:215-33. [PMID: 26425105 PMCID: PMC4583121 DOI: 10.2147/tacg.s55762] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A sudden unexplained death is felt to be due to a primary arrhythmic disorder when no structural heart disease is found on autopsy, and there is no preceding documentation of heart disease. In these cases, death is presumed to be secondary to a lethal and potentially heritable abnormality of cardiac ion channel function. These channelopathies include congenital long QT syndrome, catecholaminergic polymorphic ventricular tachycardia, Brugada syndrome, and short QT syndrome. In certain cases, genetic testing may have an important role in supporting a diagnosis of a primary arrhythmia disorder, and can also provide prognostic information, but by far the greatest strength of genetic testing lies in the screening of family members, who may be at risk. The purpose of this review is to describe the basic genetic and molecular pathophysiology of the primary inherited arrhythmia disorders, and to outline a rational approach to genetic testing, management, and family screening.
Collapse
Affiliation(s)
- Danna A Spears
- Division of Cardiology - Electrophysiology, University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - Michael H Gollob
- Division of Cardiology - Electrophysiology, University Health Network, Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
11
|
Qureshi SF, Ali A, John P, Jadhav AP, Venkateshwari A, Rao H, Jayakrishnan MP, Narasimhan C, Shenthar J, Thangaraj K, Nallari P. Mutational analysis of SCN5A gene in long QT syndrome. Meta Gene 2015; 6:26-35. [PMID: 26401487 PMCID: PMC4561237 DOI: 10.1016/j.mgene.2015.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 07/05/2015] [Accepted: 07/31/2015] [Indexed: 12/20/2022] Open
Abstract
The SCN5A gene encodes for the INa channel implicated in long QT syndrome type-3 (LQTS-type-3). Clinical symptoms of this type are lethal as most patients had a sudden death during sleep. Screening of SCN5A in South Indian cohort by PCR-SSCP analyses revealed five polymorphisms — A29A (exon-2), H558R (exon-12), E1061E and S1074R (exon-17) and IVS25 + 65G > A (exon-25) respectively. In-silico and statistical analyses were performed on all the polymorphisms. Exon-2 of SCN5A gene revealed A282G polymorphism (rs6599230), resulting in alanine for alanine (A29A) silent substitution in the N-terminus of SCN5A protein. Exon-12 showed A1868G polymorphism (H558R — rs1805124) and its ‘AA’ genotype and ‘A’ allele frequency were found to be higher in LQTS patients pointing towards its role in LQTS etiology. Two polymorphisms A3378G (E1061E) and the novel C3417A (S1074R) were identified as compound heterozygotes/genetic compounds in exon-17 of SCN5A located in the DIIS6–DIIIS1 domain of the SCN5A transmembrane protein. IVS25 + 65G > A was identified in intron-25 of SCN5A. The ‘G’ allele was identified as the risk allele. Variations were identified in in-silico analyses which revealed that these genetic compounds may lead to downstream signaling variations causing aberrations in sodium channel functions leading to prolonged QTc. The compound heterozygotes of SCN5A gene polymorphisms revealed a significant association which may be deleterious/lethal leading to an aberrant sodium ion channel causing prolonged QTc. Screening of SCN5A in South Indian cohort showed 4 reported and 1 novel polymorphism. Compound heterozygotes found to have a significant association with LQTS Genetic compounds maybe deleterious leading to an aberrant sodium ion channel
Collapse
Affiliation(s)
- Sameera Fatima Qureshi
- Dept. of Genetics, University College of Science, Osmania University, Hyderabad 500007, Andhra Pradesh, India
| | - Altaf Ali
- Dept. of Genetics, University College of Science, Osmania University, Hyderabad 500007, Andhra Pradesh, India
| | - Princy John
- Dept. of Genetics, University College of Science, Osmania University, Hyderabad 500007, Andhra Pradesh, India
| | - Amol P Jadhav
- Dept. of Genetics, University College of Science, Osmania University, Hyderabad 500007, Andhra Pradesh, India
| | - Ananthapur Venkateshwari
- Institute of Genetics and Hospital for Genetic Diseases, Osmania University, Begumpet, Hyderabad 500016, Andhra Pradesh, India
| | - Hygriv Rao
- Krishna Institute of Medical Sciences, 1-8-31/1, Minister Road, Secunderabad 500 003, Andhra Pradesh, India
| | - M P Jayakrishnan
- Institute of Maternal and Child Health, Calicut Medical College, Calicut 8, Kerala, India
| | | | - Jayaprakash Shenthar
- Sri Jayadeva Institute of Cardiovascular Science and Research, Jayanagar 9th Block, Bannerghatta Road, Bangalore 560069, Karnataka, India
| | - Kumarasamy Thangaraj
- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, Andhra Pradesh, India
| | - Pratibha Nallari
- Dept. of Genetics, University College of Science, Osmania University, Hyderabad 500007, Andhra Pradesh, India
| |
Collapse
|
12
|
Ackerman MJ. Genetic purgatory and the cardiac channelopathies: Exposing the variants of uncertain/unknown significance issue. Heart Rhythm 2015; 12:2325-31. [PMID: 26144349 DOI: 10.1016/j.hrthm.2015.07.002] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Indexed: 10/23/2022]
Abstract
Merriam-Webster's online dictionary defines purgatory as "an intermediate state after death for expiatory purification" or more specifically as "a place or state of punishment wherein according to Roman Catholic doctrine the souls of those who die in God׳s grace may make satisfaction for past sins and so become fit for heaven." Alternatively, it is defined as "a place or state of temporary suffering or misery." Either way, purgatory is a place where you are stuck, and you don't want to be stuck there. It is in this context that the term genetic purgatory is introduced. Genetic purgatory is a place where the genetic test-ordering physician and patients and their families are stuck when a variant of uncertain/unknown significance (VUS) has been elucidated. It is in this dark place where suffering and misery are occurring because of unenlightened handling of a VUS, which includes using the VUS for predictive genetic testing and making radical treatment recommendations based on the presence or absence of a so-called maybe mutation. Before one can escape from this miserable place, one must first recognize that one is stuck there. Hence, the purpose of this review article is to fully expose the VUS issue as it relates to the cardiac channelopathies and make the cardiologists/geneticists/genetic counselors who order such genetic tests believers in genetic purgatory. Only then can one meaningfully attempt to get out of that place and seek to promote a VUS to disease-causative mutation status or demote it to an utterly innocuous and irrelevant variant.
Collapse
Affiliation(s)
- Michael J Ackerman
- Departments of Medicine, Pediatrics, and Molecular Pharmacology & Experimental Therapeutics, Divisions of Cardiovascular Medicine, Pediatric Cardiology, and the Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
13
|
Effect of celastrol on growth inhibition of prostate cancer cells through the regulation of hERG channel in vitro. BIOMED RESEARCH INTERNATIONAL 2015; 2015:308475. [PMID: 25866772 PMCID: PMC4383143 DOI: 10.1155/2015/308475] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/28/2014] [Indexed: 11/17/2022]
Abstract
Objective. To explore the antiprostate cancer effects of Celastrol on prostate cancer cells' proliferation, apoptosis, and cell cycle distribution, as well as the correlation to the regulation of hERG. Methods. DU145 cells were treated with various concentrations of Celastrol (0.25–16.0 μmol/L) for 0–72 hours. MTT assay was used to evaluate the inhibition effect of Celastrol on the growth of DU145 cells. Cell apoptosis was detected through both Annexin-V FITC/PI double-labeled cytometry and Hoechst 33258. Cell cycle regulation was examined by a propidium iodide method. Western blot and RT-PCR technologies were applied to assess the expression level of hERG in DU145 cells. Results. Celastrol presented striking growth inhibition and apoptosis induction potency on DU145 cells in vitro in a time- and dose-dependent manner. The IC50 value of Celastrol for 24 hours was 2.349 ± 0.213 μmol/L. Moreover, Celastrol induced DU145 cell apoptosis in a cell cycle-dependent manner, which means Celastrol could arrest DU145 cells in G0/G1 phase; accordingly, cells in S phase decreased gradually and no obvious changes were found in G2/M phase cells. Through transmission electron microscope, apoptotic bodies containing nuclear fragments were found in Celastrol-treated DU145 cells. Overexpression of hERG channel was found in DU145 cells, while Celastrol could downregulate it at both protein and mRNA level in a dose-dependent manner (P < 0.01). Conclusions. Celastrol exhibits its antiprostate cancer effects partially through the downregulation of the expression level of hERG channel in DU145 cells, suggesting that Celastrol may be a potential agent against prostate cancer with a mechanism of blocking the hERG channel.
Collapse
|
14
|
Abstract
Proper generation and conduction of the cardiac electrical impulse is essential for the continuous coordinated contraction of the heart. Dysregulation of cardiac electrical function may lead to cardiac arrhythmias, which constitute a huge medical and social burden. Identifying the genetic factors underlying cardiac electrical activity serves the double purpose of allowing the early identification of individuals at risk for arrhythmia and discovering new potential therapeutic targets for prevention. The aim of this review is to provide an overview of the genes and genetic loci linked thus far to cardiac electrical function and arrhythmia. These genes and loci have been primarily uncovered through studies on the familial rhythm disorders and through genome-wide association studies on electrocardiographic parameters in large sets of the general population. An overview of all genes and loci with their respective effect is given.
Collapse
Affiliation(s)
- Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands. Tel.: +31 20 5665962; Fax: +31 20 6976177;
| | | |
Collapse
|
15
|
Novel SCN5A mutations in two families with "Brugada-like" ST elevation in the inferior leads and conduction disturbances. J Interv Card Electrophysiol 2013; 37:131-40. [PMID: 23612926 DOI: 10.1007/s10840-013-9805-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 03/20/2013] [Indexed: 10/26/2022]
Abstract
AIMS Brugada syndrome (BrS) is an inherited cardiac disease characterized by ST segment elevation in V1-V3 ECG leads. Mutations SCN5A gene encoding for the cardiac voltage-gated Na(+) channel are found in some BrS patients, but also in family members with isolated conduction disturbances. However, some patients show coved ST elevation in the inferior or lateral leads whose association with SCN5A and familial conduction disturbances are poorly known. METHODS AND RESULTS Two novel SCN5A mutations, D1430N and Q1476X, were identified in two unrelated families comprising patients with Brugada-like ST elevation located in the inferior leads or isolated conduction disturbances. Wild-type (WT) and D1430N mutant channels were expressed in tsA201 cells. Patch clamp electrophysiological experiments revealed total absence of Na(+) current resulting from Nav1.5 mutant when compared to WT channels. Treatments known to restore trafficking defect (incubation at low temperature, with mexiletine or lidocaine) did not restore Na(+) current supporting that Nav1.5 mutation is not a defective trafficking mutation. Furthermore, immunocytolabelling indicates the membrane localisation of both WT and mutant channels confirming what we observed in our patch clamp experiments. This suggests that the mutation may induce a complete block of Na(+) permeation. The nonsense mutation Q1476X was leading to a premature stop codon and was not expressed. CONCLUSION Brugada-like ST elevation in the inferior ECG leads or isolated conduction disturbances were found in two unrelated families and associated with two novel SCN5A mutations. The missense and nonsense mutations are both resulting in a complete loss of ventricular Na(+) current explaining the phenotypes.
Collapse
|
16
|
Duehmke RM, Pearcey S, Stefaniak JD, Guzadhur L, Jeevaratnam K, Costopoulos C, Pedersen TH, Grace AA, Huang CLH. Altered re-excitation thresholds and conduction of extrasystolic action potentials contribute to arrhythmogenicity in murine models of long QT syndrome. Acta Physiol (Oxf) 2012; 206:164-77. [PMID: 22510251 DOI: 10.1111/j.1748-1716.2012.02443.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/18/2011] [Accepted: 04/03/2012] [Indexed: 11/26/2022]
Abstract
AIM QT interval prolongation reflecting delayed action potential (AP) repolarization is associated with polymorphic ventricular tachycardia and early after depolarizations potentially initiating extrasystolic APs if of sufficient amplitude. The current experiments explored contributions of altered re-excitation thresholds for, and conduction of, such extrasystolic APs to arrhythmogenesis in Langendorff-perfused, normokalaemic, control wild-type hearts and two experimental groups modelling long QT (LQT). The two LQT groups consisted of genetically modified, Scn5a(+/ΔKPQ) and hypokalaemic wild-type murine hearts. METHODS Hearts were paced from their right ventricles and monophasic AP electrode recordings obtained from their left ventricular epicardia, with recording and pacing electrodes separated by 1 cm. An adaptive programmed electrical stimulation protocol applied pacing (S1) stimulus trains followed by premature (S2) extrastimuli whose amplitudes were progressively increased with progressive decrements in S1S2 interval to maintain stimulus capture. Such protocols culminated in either arrhythmic or refractory endpoints. RESULTS Arrhythmic outcomes were associated with (1) lower conduction velocities in their initiating extrasystolic APs than refractory outcomes and (2) higher conduction velocities in the LQT groups than in controls. Furthermore, (3) the endpoints were reached at longer S1S2 coupling intervals and with smaller stimulus amplitudes in the LQT groups compared with controls. This was despite (4) similar relationships between conduction velocity and S1S2 coupling interval and between re-excitation thresholds and S1S2 coupling interval in all three experimental groups. CONCLUSIONS Arrhythmias induced by extrasystolic APs in the LQT groups thus occur under conditions of higher conduction velocity and greater sensitivity to extrastimuli than in controls.
Collapse
Affiliation(s)
| | - S. Pearcey
- Physiological Laboratory; University of Cambridge; Cambridge; UK
| | - J. D. Stefaniak
- Physiological Laboratory; University of Cambridge; Cambridge; UK
| | - L. Guzadhur
- Physiological Laboratory; University of Cambridge; Cambridge; UK
| | | | - C. Costopoulos
- Department of Biochemistry; University of Cambridge; Cambridge; UK
| | - T. H. Pedersen
- Department of Biomedicine; Aarhus University; Aarhus; Denmark
| | - A. A. Grace
- Department of Biochemistry; University of Cambridge; Cambridge; UK
| | | |
Collapse
|
17
|
Song W, Shou W. Cardiac sodium channel Nav1.5 mutations and cardiac arrhythmia. Pediatr Cardiol 2012; 33:943-9. [PMID: 22460359 PMCID: PMC3393812 DOI: 10.1007/s00246-012-0303-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 03/09/2012] [Indexed: 12/19/2022]
Abstract
As a major cardiac voltage-gated sodium channel isoform in the heart, the Nav1.5 channel is essential for cardiac action potential initiation and subsequent propagation throughout the heart. Mutations of Nav1.5 have been linked to a variety of cardiac diseases such as long QT syndrome (LQTs), Brugada syndrome, cardiac conduction defect, atrial fibrillation, and dilated cardiomyopathy. The mutagenesis approach and heterologous expression systems are most frequently used to study the function of this channel. This review focuses primarily on recent findings of Nav1.5 mutations associated with type 3 long QT syndrome (LQT3) in particular. Understanding the functional changes of the Nav1.5 mutation may offer critical insight into the mechanism of long QT3 syndrome. In addition, this review provides the updated information on the current progress of using various experimental model systems to study primarily the long QT3 syndrome.
Collapse
Affiliation(s)
- Weihua Song
- Department of Pediatrics, Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | | |
Collapse
|
18
|
Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K+ Channels: Structure, Function, and Clinical Significance. Physiol Rev 2012; 92:1393-478. [DOI: 10.1152/physrev.00036.2011] [Citation(s) in RCA: 526] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the rapid component of the delayed rectifier K+ channel, Kv11.1, which are expressed in the heart, various brain regions, smooth muscle cells, endocrine cells, and a wide range of tumor cell lines. However, it is the role that Kv11.1 channels play in the heart that has been best characterized, for two main reasons. First, it is the gene product involved in chromosome 7-associated long QT syndrome (LQTS), an inherited disorder associated with a markedly increased risk of ventricular arrhythmias and sudden cardiac death. Second, blockade of Kv11.1, by a wide range of prescription medications, causes drug-induced QT prolongation with an increase in risk of sudden cardiac arrest. In the first part of this review, the properties of Kv11.1 channels, including biogenesis, trafficking, gating, and pharmacology are discussed, while the second part focuses on the pathophysiology of Kv11.1 channels.
Collapse
Affiliation(s)
- Jamie I. Vandenberg
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Matthew D. Perry
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Mark J. Perrin
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Stefan A. Mann
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Ying Ke
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Adam P. Hill
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
19
|
Perrin MJ, Gollob MH. The genetics of cardiac disease associated with sudden cardiac death: a paper from the 2011 William Beaumont Hospital Symposium on molecular pathology. J Mol Diagn 2012; 14:424-36. [PMID: 22749884 DOI: 10.1016/j.jmoldx.2012.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/03/2012] [Accepted: 04/13/2012] [Indexed: 11/17/2022] Open
Abstract
Sudden cardiac death due to ventricular arrhythmia most commonly occurs in the setting of coronary artery disease. However, a number of inherited syndromes have now been identified that carry a significant risk of sudden cardiac death and that are disproportionately represented in the young. Arrhythmia in such conditions may result from genetically mediated structural heart disease (eg, hypertrophic cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy) or from altered function of cardiac ion channels in the absence of overt structural disease (eg, Brugada syndrome and long QT syndrome). The past 15 years have seen considerable progress in our understanding of the genetic underpinnings of these disorders. With the advent of clinical genetic testing as a routine part of clinical care, a new knowledge base is required of practicing cardiologists and genetic testing facilities, particularly related to the rational ordering of genetic testing and the interpretation of results. This review addresses the latest findings in regard to the genetic causes of inherited syndromes associated with sudden cardiac death and summarizes recently published guidelines for the genetic testing of affected individuals and their families.
Collapse
Affiliation(s)
- Mark J Perrin
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | |
Collapse
|
20
|
|
21
|
Jiménez-Vargas JM, Restano-Cassulini R, Possani LD. Toxin modulators and blockers of hERG K(+) channels. Toxicon 2012; 60:492-501. [PMID: 22497787 DOI: 10.1016/j.toxicon.2012.03.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/27/2012] [Indexed: 12/24/2022]
Abstract
The K(+) channel encoded by the Ether-á-go-go-Related Gene (ERG) is expressed in different tissues of different animal species. There are at least three subtypes of this channel, being the sub-type 1 (ERG1) crucial in the repolarization phase of the cardiac action potential. Mutations in this gene can affect the properties of the channel producing the type II long QT syndrome (LQTS2) and many drugs are also known to affect this channel with a similar side effect. Various scorpion, spider and sea anemone toxins affect the ERG currents by blocking the ion-conducting pore from the external side or by modulating channel gating through binding to the voltage-sensor domain. By doing so, these toxins become very useful tools for better understanding the structural and functional characteristics of these ion channels. This review discusses the interaction between the ERG channels and the peptides isolated from venoms of these animals. Special emphasis is placed on scorpion toxins, although the effects of several spider venom toxins and anemone toxins will be also revised.
Collapse
Affiliation(s)
- J M Jiménez-Vargas
- Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Av. Universidad 2001, P.O. Box 501-3, Cuernavaca 62210, Mexico.
| | | | | |
Collapse
|
22
|
Hamilton RM. Tinker, tailor: Foretelling the future in long QT syndrome. Heart Rhythm 2011; 8:1544-5. [DOI: 10.1016/j.hrthm.2011.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Indexed: 10/18/2022]
|
23
|
Roberts JD, Gollob MH. The genetic and clinical features of cardiac channelopathies. Future Cardiol 2010; 6:491-506. [PMID: 20608822 DOI: 10.2217/fca.10.27] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sudden cardiac death, secondary to malignant ventricular arrhythmias, has traditionally been associated with structural heart disease. An important exception includes a group of clinical entities referred to as 'channelopathies' that develop secondary to genetic mutations, which alter cardiac ion channel activity. Otherwise healthy individuals affected by these forms of primary electrical disease are vulnerable to fatal arrhythmic events from a very young age. At present, there are four distinct conditions that are classified as cardiac channelopathies, namely congenital long-QT syndrome, Brugada syndrome, catecholaminergic polymorphic ventricular tachycardia and short-QT syndrome. Our growing insight into the genetics of these conditions has led to an improved understanding of the molecular pathophysiology responsible for the malignant arrhythmias characterizing these disorders. However, despite our knowledge of these conditions, the success of medical therapy remains modest and the prevention of sudden cardiac death may necessitate insertion of an implantable cardioverter-defibrillator. The young age of affected patients makes this a particularly undesirable treatment strategy and emphasizes the importance of translating our insight into the molecular pathophysiology defining these conditions into more effective forms of therapy.
Collapse
Affiliation(s)
- Jason D Roberts
- Arrhythmia Research Laboratory, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | | |
Collapse
|
24
|
Collins RT, Aziz PF, Gleason MM, Kaplan PB, Shah MJ. Abnormalities of cardiac repolarization in Williams syndrome. Am J Cardiol 2010; 106:1029-33. [PMID: 20854969 DOI: 10.1016/j.amjcard.2010.05.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 05/14/2010] [Accepted: 05/14/2010] [Indexed: 11/26/2022]
Abstract
Williams syndrome (WS) affects 1 in 8,000 live births and has a high risk of sudden death. No previous studies have evaluated corrected QT (QTc) prolongation in WS. Retrospective review of all patients with WS evaluated at our institution from January 1, 1980 to December 31, 2007 was performed. WS was diagnosed by a medical geneticist and/or by fluorescence in situ hybridization. Patients with ≥1 electrocardiogram (ECG) with sinus rhythm and measurable intervals were included. Normal control ECGs were identified from a large clinical database. Corrected JT (JTc) interval was calculated when QRS and QTc intervals were prolonged. QTc interval ≥460 ms and JTc interval >340 ms were defined as prolonged. Prevalence comparisons were made using Fisher's exact test. Statistical probability of <0.05 was considered significant. Of 270 patients identified, 188 had ECGs for review. Complete data were present in 499 of 517 ECGs (patients' mean age 10.3 ± 9.9 years); 1,522 normal ECGs of age-similar patients composed the control group. QTc prolongation prevalences were 2.0% in controls and 13.6% in WS (p <0.0001); in those, JTc prolongation prevalences were 1.8% in controls and 11.7% in WS (p <0.0001). Four patients died during follow-up; 2 had QTc prolongation and 1 died during noncardiac surgery. Another patient with QTc prolongation sustained cardiac arrest during a procedure. In conclusion, cardiac repolarization is prolonged in WS. Presence of prolonged cardiac repolarization may contribute to the high incidence of periprocedural mortality in these patients. All patients with WS should be screened for cardiac repolarization abnormalities, especially before surgery.
Collapse
|
25
|
Lanfranchi PA, Ackerman MJ, Kara T, Shamsuzzaman ASM, Wolk R, Jurak P, Amin R, Somers VK. Gene-specific paradoxical QT responses during rapid eye movement sleep in women with congenital long QT syndrome. Heart Rhythm 2010; 7:1067-74. [PMID: 20470906 PMCID: PMC3332552 DOI: 10.1016/j.hrthm.2010.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Accepted: 05/08/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Patients with congenital long QT syndrome (LQTS) type 2 (LQT2) may develop arrhythmias during emotional stress, acoustic stimuli, or sleep. Women with LQT2 are more susceptible to fatal arrhythmias than are men. OBJECTIVE The purpose of this study was to examine the effects of sleep on RR and QT intervals in patients with LQT1, in those with LQT2, and in controls and to test the hypothesis that there is a gene-specific effect of sleep on the QT interval in LQT2 that may be especially evident in women with LQT2. METHODS Thirty-four subjects with genotyped LQTS and 18 healthy controls were studied. Among the 34 subjects with LQTS, 16 (10 women, age 32 +/- 3 years) had LQT1 and 18 (11 women, age 38 +/- 3 years) had LQT2. Subjects underwent standard polysomnography including ECG recordings. RR, QT, and QTc (Bazett and Fridericia formulas) were measured over recordings obtained during stable conditions during wakefulness, during stage 2 and stages 3-4 of non-rapid eye movement (NREM), and during rapid eye movement (REM) sleep. RESULTS LQT2 women showed a marked RR decrease and marked QT and QTc increase from NREM to REM sleep, changes that were not observed in either women or men with LQT1 or in men with LQT2. CONCLUSION Pronounced cardiac activation during REM and substantial QTc prolongation is noted in a sex- and gene-specific fashion among women with LQT2. REM-related changes in cardiac activation and ventricular repolarization may be implicated in sleep-related malignant arrhythmias in women with the LQT2 genotype.
Collapse
Affiliation(s)
- Paola A. Lanfranchi
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
- Department of Medicine, Division of Cardiology, Hôpital du Sacré-Cœur, Montréal, Quebec, Canada
- Sleep Disorders Center, Hôpital du Sacré-Cœur, Montréal, Quebec, Canada
| | - Michael J. Ackerman
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Tomas Kara
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Abu S. M. Shamsuzzaman
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Robert Wolk
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
- Cardiovascular/Metabolic Diseases, Pfizer Global Research & Development, Pfizer, Inc., Groton, Connecticut
| | - Pavel Jurak
- Institute of Scientific Instruments, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| | - Raouf Amin
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Virend K. Somers
- Department of Internal Medicine, Divisions of Hypertension and Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
26
|
Witte DP, Aronow BJ, Harmony JAK. Understanding Cardiac Development Through the Perspective of Gene Regulation and Gene Manipulation. ACTA ACUST UNITED AC 2010. [DOI: 10.1080/15513819609169282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Im SW, Lee MK, Lee HJ, Oh SI, Kim HL, Sung J, Cho SI, Seo JS, Kim JI. Analysis of genetic and non-genetic factors that affect the QTc interval in a Mongolian population: the GENDISCAN study. Exp Mol Med 2010; 41:841-8. [PMID: 19641374 DOI: 10.3858/emm.2009.41.11.090] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The QTc interval is a complex quantitative trait and a strong prognostic indicator of cardiovascular mortality in general, healthy people. The aim of this study was to identify non-genetic factors and quantitative trait loci that govern the QTc interval in an isolated Mongolian population. We used multiple regression analysis to determine the relationship between the QTc interval and non-genetic factors including height, blood pressure, and the plasma lipid level. Whole genome linkage analyses were performed to reveal quantitative trait loci for the QTc interval with 349 microsatellite markers from 1,080 Mongolian subjects. Among many factors previously known for association with the QTc interval, age, sex, heart rate, QRS duration of electrocardiogram and systolic blood pressure were also found to have influence on the QTc interval. A genetic effect for the QTc interval was identified based on familial correlation with a heritability value of 0.31. In a whole genome linkage analysis, we identified the four potential linkage regions 7q31-34, 5q21, 4q28, and 2q36.
Collapse
Affiliation(s)
- Sun-Wha Im
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The heart is a force-generating organ that responds to self-generated electrical stimuli from specialized cardiomyocytes. This function is modulated by sympathetic and parasympathetic activity. In order to contract and accommodate the repetitive morphological changes induced by the cardiac cycle, cardiomyocytes depend on their highly evolved and specialized cytoskeletal apparatus. Defects in components of the cytoskeleton affect the ability of the cell to compensate at both functional and structural levels in the long term. In addition to structural remodeling, the myocardium becomes increasingly susceptible to altered electrical activity, leading to arrhythmogenesis. The development of arrhythmias secondary to structural remodeling defects has been noted, although the detailed molecular mechanisms are still elusive. Here, the author reviews the current knowledge of the molecular and functional relationships between the cytoskeleton and ion channels, and discusses the future impact of new data on molecular cardiology research and clinical practice.
Collapse
Affiliation(s)
- Matteo Vatta
- Baylor College of Medicine, Pediatrics (Cardiology), Texas Children's Hospital, 6621 Fannin St, FC 430.09, Houston, TX 77030, USA.
| | | |
Collapse
|
29
|
Disease-Causing Polymorphisms in the Spectrum of Long QT Syndrome Mutations. J Am Coll Cardiol 2009; 54:820-1. [DOI: 10.1016/j.jacc.2009.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 06/30/2009] [Indexed: 11/20/2022]
|
30
|
Regional, age-dependent, and genotype-dependent differences in ventricular action potential duration and activation time in 410 Langendorff-perfused mouse hearts. Basic Res Cardiol 2009; 104:523-33. [DOI: 10.1007/s00395-009-0019-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 02/17/2009] [Accepted: 03/04/2009] [Indexed: 10/21/2022]
|
31
|
Tsurugi T, Nagatomo T, Abe H, Oginosawa Y, Takemasa H, Kohno R, Makita N, Makielski JC, Otsuji Y. Differential modulation of late sodium current by protein kinase A in R1623Q mutant of LQT3. Life Sci 2009; 84:380-7. [PMID: 19167409 PMCID: PMC2747087 DOI: 10.1016/j.lfs.2009.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 11/17/2008] [Accepted: 01/05/2009] [Indexed: 12/28/2022]
Abstract
AIMS In the type 3 long QT syndrome (LQT3), shortening of the QT interval by overdrive pacing is used to prevent life-threatening arrhythmias. However, it is unclear whether accelerated heart rate induced by beta-adrenergic agents produces similar effects on the late sodium current (I(Na)) to those by overdrive pacing therapy. We analyzed the beta-adrenergic-like effects of protein kinase A and fluoride on I(Na) in R1623Q mutant channels. MAIN METHODS cDNA encoding either wild-type (WT) or R1623Q mutant of hNa(v)1.5 was stably transfected into HEK293 cells. I(Na) was recorded using a whole-cell patch-clamp technique at 23 degrees C. KEY FINDINGS In R1623Q channels, 2 mM pCPT-AMP and 120 mM fluoride significantly delayed macroscopic current decay and increased relative amplitude of the late I(Na) in a time-dependent manner. Modulations of peak I(Na) gating kinetics (activation, inactivation, recovery from inactivation) by fluoride were similar in WT and R1623Q channels. The effects of fluoride were almost completely abolished by concomitant dialysis with a protein kinase inhibitor. We also compared the effect of pacing with that of beta-adrenergic stimulation by analyzing the frequency-dependence of the late I(Na). Fluoride augmented frequency-dependent reduction of the late I(Na), which was due to preferential delay of recovery of late I(Na). However, the increase in late I(Na) by fluoride at steady-state was more potent than the frequency-dependent reduction of late I(Na). SIGNIFICANCE Different basic mechanisms participate in the QT interval shortening by pacing and beta-adrenergic stimulation in the LQT3.
Collapse
Affiliation(s)
- Takuo Tsurugi
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Toshihisa Nagatomo
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Haruhiko Abe
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Yasushi Oginosawa
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Hiroko Takemasa
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Ritsuko Kohno
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| | - Naomasa Makita
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Jonathan C. Makielski
- Department of Medicine, Section of Cardiovascular Medicine, University of Wisconsin, Madison, WI 53792, USA
| | - Yutaka Otsuji
- Second Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu 807-8555, Japan
| |
Collapse
|
32
|
Sudandiradoss C, Sethumadhavan R. In silico investigations on functional and haplotype tag SNPs associated with congenital long QT syndromes (LQTSs). Genomic Med 2008; 2:55-67. [PMID: 19214780 PMCID: PMC2694858 DOI: 10.1007/s11568-009-9027-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 11/17/2008] [Accepted: 01/15/2009] [Indexed: 01/08/2023] Open
Abstract
Single-nucleotide polymorphisms (SNPs) play a major role in the understanding of the genetic basis of many complex human diseases. It is still a major challenge to identify the functional SNPs in disease-related genes. In this review, the genetic variation that can alter the expression and the function of the genes, namely KCNQ1, KCNH2, SCN5A, KCNE1 and KCNE2, with the potential role for the development of congenital long QT syndrome (LQTS) was analyzed. Of the total of 3,309 SNPs in all five genes, 27 non-synonymous SNPs (nsSNPs) in the coding region and 44 SNPs in the 5' and 3' un-translated regions (UTR) were identified as functionally significant. SIFT and PolyPhen programs were used to analyze the nsSNPs and FastSNP; UTR scan programs were used to compute SNPs in the 5' and 3' untranslated regions. Of the five selected genes, KCNQ1 has the highest number of 26 haplotype blocks and 6 tag SNPs with a complete linkage disequilibrium value. The gene SCN5A has ten haplotype blocks and four tag SNPs. Both KCNE1 and KCNE2 genes have only one haplotype block and four tag SNPs. Four haplotype blocks and two tag SNPs were obtained for KCNH2 gene. Also, this review reports the copy number variations (CNVs), expressed sequence tags (ESTs) and genome survey sequences (GSS) of the selected genes. These computational methods are in good agreement with experimental works reported earlier concerning LQTS.
Collapse
Affiliation(s)
- C. Sudandiradoss
- Bioinformatics Division, School of Biotechnology, Chemical and Biomedical Engineering, Vellore Institute of Technology, Vellore, TN 632014 India
| | - Rao Sethumadhavan
- Bioinformatics Division, School of Biotechnology, Chemical and Biomedical Engineering, Vellore Institute of Technology, Vellore, TN 632014 India
| |
Collapse
|
33
|
Perrin MJ, Subbiah RN, Vandenberg JI, Hill AP. Human ether-a-go-go related gene (hERG) K+ channels: function and dysfunction. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2008; 98:137-48. [PMID: 19027781 DOI: 10.1016/j.pbiomolbio.2008.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human Ether-a-go-go Related Gene (hERG) potassium channel plays a central role in regulating cardiac excitability and maintenance of normal cardiac rhythm. Mutations in hERG cause a third of all cases of congenital long QT syndrome, a disorder of cardiac repolarisation characterised by prolongation of the QT interval on the surface electrocardiogram, abnormal T waves, and a risk of sudden cardiac death due to ventricular arrhythmias. Additionally, the hERG channel protein is the molecular target for almost all drugs that cause the acquired form of long QT syndrome. Advances in understanding the structural basis of hERG gating, its traffic to the cell surface, and the molecular architecture involved in drug-block of hERG, are providing the foundation for rational treatment and prevention of hERG associated long QT syndrome. This review summarises the current knowledge of hERG function and dysfunction, and the areas of ongoing research.
Collapse
Affiliation(s)
- Mark J Perrin
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia
| | | | | | | |
Collapse
|
34
|
Nagaoka I, Shimizu W, Itoh H, Yamamoto S, Sakaguchi T, Oka Y, Tsuji K, Ashihara T, Ito M, Yoshida H, Ohno S, Makiyama T, Miyamoto Y, Noda T, Kamakura S, Akao M, Horie M. Mutation site dependent variability of cardiac events in Japanese LQT2 form of congenital long-QT syndrome. Circ J 2008; 72:694-9. [PMID: 18441445 DOI: 10.1253/circj.72.694] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND In the LQT2 form of long QT syndrome (LQTS), mutation sites are reported to correlate with clinical phenotypes in Caucasians, but the relationship in Asian patients remains unknown. The present study was designed to determine whether the location of KCNH2 mutations would influence the arrhythmic risk in LQT2 patients. METHODS AND RESULTS In 118 genetically-confirmed LQT2 patients (69 families, 62 KCNH2 mutations), the ECG parameters, Schwartz scores, and the incidence of cardiac events, defined as syncope, aborted cardiac arrest, and sudden cardiac death, were evaluated. To examine the effect of mutation sites, the participants were divided accordingly: pore (n=56) and non-pore (n=62) groups. The corrected QTend interval was significantly greater in the pore than in the non-pore group (QTc; 522+/-63 ms vs 490+/-49 ms, p=0.002). In this study, the clinical course of each of the probands did not differ according to the mutation sites, whereas non-probands carrying the pore site mutation experienced their first cardiac events at significantly younger age than those with the non-pore site mutation (log-rank, p=0.0005). CONCLUSIONS In a Japanese LQT2 cohort, family members with the pore site mutation were at higher arrhythmic risk than those with the non-pore site mutation.
Collapse
Affiliation(s)
- Iori Nagaoka
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jeyaraj D, Abernethy DP, Natarajan RN, Dettmer MM, Dikshteyn M, Meredith DM, Patel K, Allareddy RR, Lewis SA, Kaufman ES. IKr channel blockade to unmask occult congenital long QT syndrome. Heart Rhythm 2008; 5:2-7. [DOI: 10.1016/j.hrthm.2007.08.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Koo SH, Teo WS, Ching CK, Chan SH, Lee EJD. Mutation Screening in KCNQ1, HERG, KCNE1, KCNE2 and SCN5A Genes in a Long QT Syndrome Family. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2007. [DOI: 10.47102/annals-acadmedsg.v36n6p394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Introduction: Long QT syndrome (LQTS), an inherited cardiac arrhythmia, is a disorder of ventricular repolarisation characterised by electrocardiographic abnormalities and the onset of torsades de pointes leading to syncope and sudden death. Genetic polymorphisms in 5 wellcharacterised cardiac ion channel genes have been identified to be responsible for the disorder. The aim of this study is to identify disease-causing mutations in these candidate genes in a LQTS family.
Materials and Methods: The present study systematically screens the coding region of the LQTS-associated genes (KCNQ1, HERG, KCNE1, KCNE2 and SCN5A) for mutations using DNA sequencing analysis.
Results: The mutational analysis revealed 7 synonymous and 2 nonsynonymous polymorphisms in the 5 ion channel genes screened.
Conclusion: We did not identify any clear identifiable genetic marker causative of LQTS, suggesting the existence of LQTSassociated genes awaiting discovery.
Key words: Arrhythmia, Ion channels, Long QT syndrome
Collapse
|
37
|
Ehret GB, Desmeules JA, Broers B. Methadone-associated long QT syndrome: improving pharmacotherapy for dependence on illegal opioids and lessons learned for pharmacology. Expert Opin Drug Saf 2007; 6:289-303. [PMID: 17480178 DOI: 10.1517/14740338.6.3.289] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Methadone is used as the pharmacologic mainstay for substitution for illegal opiates and as analgesic for chronic or cancer-related pain. Given the benefits of methadone substitution for illicit opioids, the finding of an association between methadone and prolongation of cardiac depolarization (QT prolongation) and torsades de pointes is of great concern. QT prolongation can occur with many drugs and is a potentially lethal adverse drug reaction, necessitating risk monitoring and therapeutic alternatives in some patients. Recent studies suggest that QT prolongation with methadone is context dependent: occurrence is more frequent with high doses of methadone, concomitant administration of CYP3A4 inhibitors, hypokalemia, hepatic failure, administration of other QT prolonging drugs and pre-existing heart disease. The valued benefit of methadone substitution therapy on the one hand and the increased cardiovascular risk in particular situations on the other illustrate the difficulties in dealing with drug-induced QT prolongation in general.
Collapse
Affiliation(s)
- Georg B Ehret
- Johns Hopkins University School of Medicine, McKusick Nathans Institute of Genetic Medicine, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
38
|
Benditt DG, Sakaguchi S. Syncope. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
39
|
Bezzina CR, Wilde AAM. Genetic Basis for Cardiac Arrhythmias. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
40
|
Abstract
Inherited cardiac arrhythmia syndromes have received a lot of attention in recent years, particularly the molecular genetic basis, which has been unraveled to a great extent in the past years. Disease entities have been subdivided based on their causal gene defect, which, indeed, has been shown to impact on disease expression, clinical characteristics, prognosis and treatment. This particularly holds for the long QT syndrome. Studies in other, more recently described, disease entities, such as Brugada syndrome, catecholaminergic polymorphic ventricular arrhythmias and the short QT syndrome, are ongoing. For some of them the heterogenetic nature has just very recently been established. For these reasons, genetic testing has been introduced to clinical practice in several countries, which enables timely treatment of affected individuals and reassurance of those not inheriting the causal gene defect. Presymptomatic testing, however, is not without drawbacks. Psychosocial studies are needed in this field and should be promoted. It is likely that this development will further increase the knowledge of the (patho-) physiology of these disease entities, but also of more common arrhythmia syndromes.
Collapse
Affiliation(s)
- Arthur Am Wilde
- Experimental & Molecular Cardiology Group, Academic Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
41
|
Abstract
The long and short QT syndromes are heterogeneous diseases characterized by abnormal ventricular repolarization and episodes of syncope and/or life-threatening cardiac arrhythmias. Several disease-causing genes have been identified, including those encoding cardiac ion channel-composing proteins. The clinical determination of genotype offers a striking benefit: diagnosis, prediction of clinical phenotype, risk stratification, clinical and genetic counseling, and introduction of therapy. Genetic testing is of special importance for the genotyped patient's family members to prevent unexpected cardiac death. By means of recently advanced methodology in molecular genetics and electrophysiology it is expected that novel genes responsible for these disease entities will be identified.
Collapse
Affiliation(s)
- Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan.
| | | |
Collapse
|
42
|
Koo SH, Ho WF, Lee EJD. Genetic polymorphisms in KCNQ1, HERG, KCNE1 and KCNE2 genes in the Chinese, Malay and Indian populations of Singapore. Br J Clin Pharmacol 2006; 61:301-8. [PMID: 16487223 PMCID: PMC1885019 DOI: 10.1111/j.1365-2125.2005.02545.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIMS To determine the genetic variability of long QT syndrome (LQTS)-associated genes (KCNQ1, HERG, KCNE1 and KCNE2) among three distinct ethnic groups in the Singapore population. METHODS Genomic DNA samples from up to 265 normal healthy Chinese, 118 Malay and 139 Indian volunteer subjects were screened for genetic variations in the coding region of the LQTS-associated genes using denaturing high-performance liquid chromatography and sequencing analyses. RESULTS In total, 37 single nucleotide polymorphisms (SNPs) were identified in the coding exons of the LQTS-associated potassium ion channel genes, seven of which were novel nonsynonymous polymorphisms. SNPs 356G-->A (exon 1 of KCNQ1), 2624C-->T and 2893G-->A (exon 11 of HERG), 3164G-->A, 3322C-->G and 3460G-->A (exon 14 of HERG), and 79C-->T (exon 3 of KCNE2) resulted in Gly119Asp, Thr875Met, Gly965Arg, Arg1055Gln, Leu1108Val, Gly1154Ser and Arg27Cys amino acid substitutions, respectively. In addition, 16 intronic variants were detected. The functional consequence of these variants has not been studied and their association with risk of LQTS is unclear. CONCLUSIONS There exist multiple genetic polymorphisms of the LQTS-associated genes in the three distinct Asian populations. Though the functional significance of many of these SNPs is unknown, this interindividual and interethnic genetic variability may underlie the different susceptibilities of individuals to developing LQTS.
Collapse
Affiliation(s)
- Seok Hwee Koo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
43
|
Ching CK, Tan EC. Congenital long QT syndromes: clinical features, molecular genetics and genetic testing. Expert Rev Mol Diagn 2006; 6:365-74. [PMID: 16706739 DOI: 10.1586/14737159.6.3.365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Congenital long QT syndrome (LQTS) is a primary electrical disease characterized by a prolonged QT interval in the surface electrocardiogram and increased predisposition to a typical polymorphic ventricular tachycardia, termed Torsade de Pointes. Most patients with LQTS are asymptomatic and are diagnosed incidentally based on an electrocardiogram. Symptomatic patients may suffer from severe cardiac events, such as syncope and/or sudden cardiac death. Autosomal dominant forms are caused by heterozygous mutations in genes encoding the components of the ion channels. The autosomal recessive form with congenital deafness is also known as Jervell and Lang-Nielsen syndrome. It is caused by homozygous mutations or certain compound heterozygous mutations. Depending on the genetic defects, there are differences in the age of onset, severity of symptoms, and number of cardiac events and event triggers. With advances in gene technology, it is now feasible to perform genetic testing for LQTS, especially for those with family history. Identification of the mutation will lead to better management of symptoms and more targeted treatment, depending on the underlying genetic defect, resulting in a reduction of mortality and cardiac events.
Collapse
Affiliation(s)
- Chi-Keong Ching
- National Heart Centre, Department of Cardiology, National Heart Centre, Mistri Wing 17 Third Hospital Avenue, Singapore 168752, Republic of Singapore
| | | |
Collapse
|
44
|
Abstract
Propionic acidemia and long QT syndrome (LQTS) are rare disorders. In addition, both conditions are potentially lethal. The patient presented in this article was initially diagnosed with propionic acidemia. Incidentally, she was found to have LQTS on electrocardiogram and verified by stress test and epinephrine challenge. Although the patient was asymptomatic and arrhythmia free, we started her on atenolol. This is the first report of the association between LQTS and propionic acidemia.
Collapse
Affiliation(s)
- B Kakavand
- Division of Pediatric Cardiology, University of Kentucky, 800 Rose Street, MN 470, Lexington, KY, 40536-0298, USA.
| | - V A Schroeder
- Division of Pediatric Cardiology, University of Kentucky, 800 Rose Street, MN 470, Lexington, KY, 40536-0298, USA
| | - T G Di Sessa
- Division of Pediatric Cardiology, University of Kentucky, 800 Rose Street, MN 470, Lexington, KY, 40536-0298, USA
| |
Collapse
|
45
|
Shushi L, Kerem B, Goldmit M, Peretz A, Attali B, Medina A, Towbin JA, Kurokawa J, Kass RS, Benhorin J. Clinical, genetic, and electrophysiologic characteristics of a new PAS-domain HERG mutation (M124R) causing Long QT syndrome. Ann Noninvasive Electrocardiol 2005; 10:334-41. [PMID: 16029385 PMCID: PMC6932044 DOI: 10.1111/j.1542-474x.2005.00643.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES To describe the clinical, genetic, and electrophysiologic characteristics of a new PAS-domain HERG mutation (M124R) that has been identified in a single large Jewish family with Long QT syndrome (LQTS). BACKGROUND Many previously reported HERG mutations causing LQTS are located either in the C-terminus, or in the pore region. Relatively fewer clinical data are available on N-terminus (PAS-domain) mutation carriers. METHODS Clinical data were available in 76 family members (aged 1-93 years, 69 alive) over 18 years of follow-up, while electrocardiographic data were available in 57, and genetic data in 45 family members. Cellular electrophysiology was assessed in transfected Chinese Hamster Ovary (CHO) cells using the whole-cell patch-clamp technique. RESULTS Thirty-six family members were phenotypically categorized as nonaffected, 3 as equivocal, and 20 as affected. Mean QTc was 410+/-23, 440+/-10, and 498+/-41 ms, respectively, in these three subgroups. Eight out of 20 affected family members were symptomatic: five had only syncope, two had aborted cardiac arrest, and one sudden death. Genetic analyses identified the M124R point mutation in all affected members tested (n=16), while all those tested with nonaffected (n=26) and equivocal (n=3) phenotype did not carry the mutation. The M124R mutation reduced the HERG tail-current density by 65%, significantly accelerated the deactivation kinetics, and caused a negative shift in the voltage dependence of activation. CONCLUSIONS A new PAS-domain HERG mutation (M124R) was identified as causing LQTS in a large Jewish family, with high penetrance and frequent disease-related symptoms. This mutation markedly decreased the tail-current density and accelerated the deactivation kinetics of the HERG channel in transfected CHO cells.
Collapse
Affiliation(s)
- Liat Shushi
- Department of Genetics, The Life Sciences Institute
| | | | - Maya Goldmit
- Department of Genetics, The Life Sciences Institute
| | - Asher Peretz
- Departments of Physiology and Pharmacology, Sackler Medical School, Tel‐Aviv University, Israel
| | - Bernard Attali
- Departments of Physiology and Pharmacology, Sackler Medical School, Tel‐Aviv University, Israel
| | - Aron Medina
- Department of Cardiology, Bikur Cholim Hospital, The Hebrew University, Jerusalem, Israel
| | - Jeffrey A. Towbin
- Department of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Junko Kurokawa
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York
| | - Robert S. Kass
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York
| | - Jesaia Benhorin
- Department of Cardiology, Bikur Cholim Hospital, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
46
|
Berecki G, Zegers JG, Bhuiyan ZA, Verkerk AO, Wilders R, van Ginneken ACG. Long-QT syndrome-related sodium channel mutations probed by the dynamic action potential clamp technique. J Physiol 2005; 570:237-50. [PMID: 16254012 PMCID: PMC1464302 DOI: 10.1113/jphysiol.2005.096578] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Long-QT3 syndrome (LQT3) is linked to cardiac sodium channel gene (SCN5A) mutations. In this study, we used the 'dynamic action potential clamp' (dAPC) technique to effectively replace the native sodium current (I(Na)) of the Priebe-Beuckelmann human ventricular cell model with wild-type (WT) or mutant I(Na) generated in a human embryonic kidney (HEK)-293 cell that is voltage clamped by the free-running action potential of the ventricular cell. We recorded I(Na) from HEK cells expressing either WT or LQT3-associated Y1795C or A1330P SCN5A at 35 degrees C, and let this current generate and shape the action potential (AP) of subepicardial, mid-myocardial and subendocardial model cells. The HEK cell's endogenous background current was completely removed by a real-time digital subtraction procedure. With WT I(Na), AP duration (APD) was longer than with the original Priebe-Beuckelmann model I(Na), due to a late I(Na) component of approximately 30 pA that could not be revealed with conventional voltage-clamp protocols. With mutant I(Na), this late component was larger ( approximately 100 pA), producing a marked increase in APD ( approximately 70-80 ms at 1 Hz for the subepicardial model cell). The late I(Na) magnitude showed reverse frequency dependence, resulting in a significantly steeper APD-frequency relation in the mutant case. AP prolongation was more pronounced for the mid-myocardial cell type, resulting in increased APD dispersion for each of the mutants. For both mutants, a 2 s pause following rapid (2 Hz) pacing resulted in distorted AP morphology and beat-to-beat fluctuations of I(Na). Our dAPC data directly demonstrate the arrhythmogenic nature of LQT3-associated SCN5A mutations.
Collapse
Affiliation(s)
- Géza Berecki
- Experimental and Molecular Cardiology Group, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
47
|
Pérez Riera AR, Ferreira C, Dubner SJ, Schapachnik E, Soares JD, Francis J. Brief review of the recently described short QT syndrome and other cardiac channelopathies. Ann Noninvasive Electrocardiol 2005; 10:371-7. [PMID: 16029390 PMCID: PMC6932401 DOI: 10.1111/j.1542-474x.2005.00632.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
There are many diseases related to ion-channel disorders, so-called "channelopathies." Hereditary short QT syndrome is a clinical-electrocardiographic entity with autosomal-dominant mode of transmission and it is the most recently described channelopathy. The syndrome may affect infants, children, or young adults with strong positive family background of sudden cardiac death. Short QT syndrome is characterized by short QT and heart-rate-corrected QTc intervals. It is frequently associated with tall-, peaked-, and narrow-based T waves that are reminiscent of the typical "desert tent" T waves of hyperkalemia. There is a high tendency for paroxysmal atrial fibrillation due to the heterogeneous abbreviation of action potential duration and refractoriness of atrial myocytes. The arrhythmia can also be induced by programmed electrical stimulation. The safest treatment suggested is an implantable cardioverter defibrillator, though the possibilities of inappropriate shocks have caused some concern, especially in teenagers. The ability of quinidine to prolong the QT interval has the potential to be an effective therapy for patients with short QT syndrome. This is particularly important in developing countries, where the implantable cardioverter-defibrillator therapy is not always available. Since these patients are at risk of sudden cardiac death from birth, and implantable cardioverter-defibrillator implantation has a lot of limitations in very young children, the utility of quinidine has to be evaluated further. Clinicians need to be aware of this deadly electrocardiographic (ECG) pattern as it portends a high risk of sudden cardiac death in otherwise healthy subjects with structurally normal hearts.
Collapse
|
48
|
|
49
|
Towbin JA, Vatta M, Wang Z, Bowles NE, Li H. Emerging targets in the long QT syndromes and Brugada syndrome. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.3.3.423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Abstract
This article outlines the up-to-date understanding of the molecular basis of disorders that cause sudden death. Several arrhythmic disorders that cause sudden death have been well-described at the molecular level, including the long QT syndromes and Brugada syndrome; this article reviews the current scientific knowledge of these diseases. Hypertrophic cardiomyopathy, a myocardial disorder that causes sudden death also has been well-studied. Finally, a disorder in which myocardial abnormalities and rhythm abnormalities coexist, arrhythmogenic right ventricular dysplasia, is described.
Collapse
MESH Headings
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Arrhythmogenic Right Ventricular Dysplasia/genetics
- Arrhythmogenic Right Ventricular Dysplasia/physiopathology
- Cardiomyopathy, Hypertrophic, Familial/genetics
- Cardiomyopathy, Hypertrophic, Familial/physiopathology
- Child
- Death, Sudden, Cardiac/etiology
- ERG1 Potassium Channel
- Ether-A-Go-Go Potassium Channels
- Humans
- KCNQ Potassium Channels
- KCNQ1 Potassium Channel
- Long QT Syndrome/complications
- Long QT Syndrome/genetics
- Long QT Syndrome/therapy
- NAV1.5 Voltage-Gated Sodium Channel
- Potassium Channels/physiology
- Potassium Channels, Voltage-Gated
- Sodium Channels/physiology
- Syndrome
- Tachycardia, Ventricular/genetics
- Wolff-Parkinson-White Syndrome/physiopathology
Collapse
Affiliation(s)
- Jeffrey A Towbin
- Department of Pediatrics (Cardiology), Texas Children's Hospital and Baylor College of Medicine, 6621 Fannin Street, FC. 430.09, Houston, TX 77030, USA.
| |
Collapse
|