1
|
Hasegawa S, Higuchi S, Minami Y, Sakai M, Matsui Y, Shirotani S, Haruki S, Yagishita D, Shoda M, Yamaguchi J. Impact of prolonged QTc interval on mortality risk with hypertrophic cardiomyopathy. J Arrhythm 2025; 41:e13199. [PMID: 39817027 PMCID: PMC11730705 DOI: 10.1002/joa3.13199] [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] [Received: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/18/2025] Open
Abstract
Background The association between corrected QT (QTc) interval and life-threatening cardiac events in patients with hypertrophic cardiomyopathy (HCM) remains unclear. This study sought to investigate whether the prolonged QTc was associated with HCM-related death in patients with HCM. Methods We included 445 patients with HCM (mean age 51 ± 16 years, 67% men). The QTc interval was measured at the time of the initial evaluation and the patients were classified into those with and without QTc prolongation, which was defined as a QTc interval >450 ms. HCM-related death was defined as a combined endpoint of sudden death or potentially lethal arrhythmic events, heart failure-related death, and stroke-related death. Results Prolonged QTc interval was found in 120 patients (26.4%) at the time of enrollment. Over a median (IQR) follow-up period of 8.1 (4.6-11.9) years, a total of 67 patients (15.1%) experienced HCM-related deaths including 57 (12.8%) with the endpoint of sudden death or potentially lethal arrhythmic events. In a multivariable analysis that included prolonged QTc interval and the risk factors for life-threatening events, prolonged QTc interval was independently associated with an HCM-related death (adjusted hazard ratio [HR]: 1.91; 95% confidence interval [CI]: 1.16-3.16; p = .011) and this trend also persisted for the combined endpoint of sudden death or potentially lethal arrhythmic events (adjusted HR: 2.01: 95% CI: 1.17-3.46; p = .012). Conclusions In this cohort of patients with HCM, QTc prolongation may be associated with HCM-related death, including the endpoint of sudden death or potentially lethal arrhythmic events.
Collapse
Affiliation(s)
- Shun Hasegawa
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Satoshi Higuchi
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
- Clinical Research Division for Heart Rhythm Management, Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Yuichiro Minami
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Masayuki Sakai
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Yuko Matsui
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Shota Shirotani
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Shintaro Haruki
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Daigo Yagishita
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
- Clinical Research Division for Heart Rhythm Management, Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | - Morio Shoda
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
- Clinical Research Division for Heart Rhythm Management, Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| | | |
Collapse
|
2
|
Gandon-Renard M, Val-Blasco A, Oughlis C, Gerbaud P, Lefebvre F, Gomez S, Journé C, Courilleau D, Mercier-Nomé F, Pereira L, Benitah JP, Gómez AM, Mercadier JJ. Dual effect of cardiac FKBP12.6 overexpression on excitation-contraction coupling and the incidence of ventricular arrhythmia depending on its expression level. J Mol Cell Cardiol 2024; 188:15-29. [PMID: 38224852 DOI: 10.1016/j.yjmcc.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 01/17/2024]
Abstract
FKBP12.6, a binding protein to the immunosuppressant FK506, which also binds the ryanodine receptor (RyR2) in the heart, has been proposed to regulate RyR2 function and to have antiarrhythmic properties. However, the level of FKBP12.6 expression in normal hearts remains elusive and some controversies still persist regarding its effects, both in basal conditions and during β-adrenergic stimulation. We quantified FKBP12.6 in the left ventricles (LV) of WT (wild-type) mice and in two novel transgenic models expressing distinct levels of FKBP12.6, using a custom-made specific anti-FKBP12.6 antibody and a recombinant protein. FKBP12.6 level in WT LV was very low (0.16 ± 0.02 nmol/g of LV), indicating that <15% RyR2 monomers are bound to the protein. Mice with 14.1 ± 0.2 nmol of FKBP12.6 per g of LV (TG1) had mild cardiac hypertrophy and normal function and were protected against epinephrine/caffeine-evoked arrhythmias. The ventricular myocytes showed higher [Ca2+]i transient amplitudes than WT myocytes and normal SR-Ca2+ load, while fewer myocytes showed Ca2+ sparks. TG1 cardiomyocytes responded to 50 nM Isoproterenol increasing these [Ca2+]i parameters and producing RyR2-Ser2808 phosphorylation. Mice with more than twice the TG1 FKBP12.6 value (TG2) showed marked cardiac hypertrophy with calcineurin activation and more arrhythmias than WT mice during β-adrenergic stimulation, challenging the protective potential of high FKBP12.6. RyR2R420Q CPVT mice overexpressing FKBP12.6 showed fewer proarrhythmic events and decreased incidence and duration of stress-induced bidirectional ventricular tachycardia. Our study, therefore, quantifies for the first time endogenous FKBP12.6 in the mouse heart, questioning its physiological relevance, at least at rest due its low level. By contrast, our work demonstrates that with caution FKBP12.6 remains an interesting target for the development of new antiarrhythmic therapies.
Collapse
Affiliation(s)
- Marine Gandon-Renard
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Almudena Val-Blasco
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Célia Oughlis
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Pascale Gerbaud
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Florence Lefebvre
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Susana Gomez
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Clément Journé
- Fédération de Recherche en Imagerie Multimodale (FRIM), Université Paris Cité, 75018 Paris, France
| | | | - Françoise Mercier-Nomé
- UMS-IPSIT, Université Paris-Saclay, 91400 Orsay, France; Inflammation, Microbiome and Immunosurveillance, Inserm UMR-996, Université Paris-Saclay, 92140 Clamart, France
| | - Laetitia Pereira
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Jean-Pierre Benitah
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France
| | - Ana Maria Gómez
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France.
| | - Jean-Jacques Mercadier
- Signalling and Cardiovascular Pathophysiology, Inserm UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France; Université Paris Cité, Paris, France.
| |
Collapse
|
3
|
Cheng D, Wei X, Zhang Y, Zhang Q, Xu J, Yang J, Yu J, Stalin A, Liu H, Wang J, Zhong D, Pan L, Zhao W, Chen Y. The Strength of hERG Inhibition by Erythromycin at Different Temperatures Might Be Due to Its Interacting Features with the Channels. Molecules 2023; 28:5176. [PMID: 37446837 DOI: 10.3390/molecules28135176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Erythromycin is one of the few compounds that remarkably increase ether-a-go-go-related gene (hERG) inhibition from room temperature (RT) to physiological temperature (PT). Understanding how erythromycin inhibits the hERG could help us to decide which compounds are needed for further studies. The whole-cell patch clamp technique was used to investigate the effects of erythromycin on hERG channels at different temperatures. While erythromycin caused a concentration-dependent inhibition of cardiac hERG channels, it also shifted the steady-state activation and steady-state inactivation of the channel to the left and significantly accelerated the onset of inactivation at both temperatures, although temperature itself caused a profound change in the dynamics of hERG channels. Our data also suggest that the binding pattern to S6 of the channels changes at PT. In contrast, cisapride, a well-known hERG blocker whose inhibition is not affected by temperature, does not change its critical binding sites after the temperature is raised to PT. Our data suggest that erythromycin is unique and that the shift in hERG inhibition may not apply to other compounds.
Collapse
Affiliation(s)
- Dongrong Cheng
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Xiaofeng Wei
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Yanting Zhang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Qian Zhang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jianwei Xu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jiaxin Yang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Junjie Yu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610064, China
| | - Huan Liu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jintao Wang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Dian Zhong
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Lanying Pan
- Shuren International Medical College, Zhejiang Shuren University, Hangzhou 310009, China
| | - Wei Zhao
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Yuan Chen
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| |
Collapse
|
4
|
Bastos RF, Tuleski GLR, Franco LFC, Sousa MG. Tpeak—Tend, a novel electrocardiographic marker in cats with hypertrophic cardiomyopathy—a brief communication. Vet Res Commun 2022; 47:559-565. [DOI: 10.1007/s11259-022-10004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022]
|
5
|
Gossios T, Savvatis K, Zegkos T, Ntelios D, Rouskas P, Parcharidou D, Karvounis H, Efthimiadis GK. Deciphering hypertrophic cardiomyopathy with electrocardiography. Heart Fail Rev 2021; 27:1313-1323. [PMID: 34286451 DOI: 10.1007/s10741-021-10147-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2021] [Indexed: 11/30/2022]
Abstract
The comprehensive assessment of patients with hypertrophic cardiomyopathy is a complex process, with each step concurrently focusing on confirmation of the diagnosis, differentiation between sarcomeric and non-sarcomeric disease (phenocopy), and prognostication. Novel modalities such as genetic testing and advanced imaging have allowed for substantial advancements in the understanding of this condition and facilitate patient management. However, their availability is at present not universal, and interpretation requires a high level of expertise. In this setting, electrocardiography, a fast and widely available method, still retains a significant role in everyday clinical assessment of this population. In our review, we follow a stepwise approach for the interpretation of each electrocardiographic segment, discussing clinical implications of electrocardiographic patterns in sarcomeric disease, their value in the differential diagnosis from phenocopies, and impact on patient management. Outlining the substantial amount of information to be obtained from a simple tracing, we exhibit how electrocardiography is likely to remain an integral diagnostic tool in the future as well.
Collapse
Affiliation(s)
- Thomas Gossios
- Cardiology Department, NHS Foundation Trust, Guy's and St Thomas Westminster Bridge Road, London, SE1 7EH, UK. .,Inherited Cardiac Conditions Unit, Barts Heart Centre, St Bartholomew's Hospital, London, UK. .,Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece.
| | - Konstantinos Savvatis
- Inherited Cardiac Conditions Unit, Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Thomas Zegkos
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Dimitrios Ntelios
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Pavlos Rouskas
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Despoina Parcharidou
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Haralambos Karvounis
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgios K Efthimiadis
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
6
|
Elmén L, Volpato CB, Kervadec A, Pineda S, Kalvakuri S, Alayari NN, Foco L, Pramstaller PP, Ocorr K, Rossini A, Cammarato A, Colas AR, Hicks AA, Bodmer R. Silencing of CCR4-NOT complex subunits affects heart structure and function. Dis Model Mech 2020; 13:dmm044727. [PMID: 32471864 PMCID: PMC7390626 DOI: 10.1242/dmm.044727] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
The identification of genetic variants that predispose individuals to cardiovascular disease and a better understanding of their targets would be highly advantageous. Genome-wide association studies have identified variants that associate with QT-interval length (a measure of myocardial repolarization). Three of the strongest associating variants (single-nucleotide polymorphisms) are located in the putative promotor region of CNOT1, a gene encoding the central CNOT1 subunit of CCR4-NOT: a multifunctional, conserved complex regulating gene expression and mRNA stability and turnover. We isolated the minimum fragment of the CNOT1 promoter containing all three variants from individuals homozygous for the QT risk alleles and demonstrated that the haplotype associating with longer QT interval caused reduced reporter expression in a cardiac cell line, suggesting that reduced CNOT1 expression might contribute to abnormal QT intervals. Systematic siRNA-mediated knockdown of CCR4-NOT components in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) revealed that silencing CNOT1 and other CCR4-NOT genes reduced their proliferative capacity. Silencing CNOT7 also shortened action potential duration. Furthermore, the cardiac-specific knockdown of Drosophila orthologs of CCR4-NOT genes in vivo (CNOT1/Not1 and CNOT7/8/Pop2) was either lethal or resulted in dilated cardiomyopathy, reduced contractility or a propensity for arrhythmia. Silencing CNOT2/Not2, CNOT4/Not4 and CNOT6/6L/twin also affected cardiac chamber size and contractility. Developmental studies suggested that CNOT1/Not1 and CNOT7/8/Pop2 are required during cardiac remodeling from larval to adult stages. To summarize, we have demonstrated how disease-associated genes identified by GWAS can be investigated by combining human cardiomyocyte cell-based and whole-organism in vivo heart models. Our results also suggest a potential link of CNOT1 and CNOT7/8 to QT alterations and further establish a crucial role of the CCR4-NOT complex in heart development and function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lisa Elmén
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Claudia B Volpato
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Anaïs Kervadec
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Santiago Pineda
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sreehari Kalvakuri
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nakissa N Alayari
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Karen Ocorr
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alessandra Rossini
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Anthony Cammarato
- Johns Hopkins University, Division of Cardiology, 720 Rutland Ave., Baltimore, MD 21205, USA
| | - Alexandre R Colas
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Andrew A Hicks
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Rolf Bodmer
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
7
|
Sudden Cardiac Death and Copy Number Variants: What Do We Know after 10 Years of Genetic Analysis? Forensic Sci Int Genet 2020; 47:102281. [PMID: 32248082 DOI: 10.1016/j.fsigen.2020.102281] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/02/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
Over the last ten years, analysis of copy number variants has increasingly been applied to the study of arrhythmogenic pathologies associated with sudden death, mainly due to significant advances in the field of massive genetic sequencing. Nevertheless, few published reports have focused on the prevalence of copy number variants associated with sudden cardiac death. As a result, the frequency of these genetic alterations in arrhythmogenic diseases as well as their genetic interpretation and clinical translation has not been established. This review summarizes the current available data concerning copy number variants in sudden cardiac death-related diseases.
Collapse
|
8
|
Martinez-Matilla M, Blanco-Verea A, Santori M, Ansede-Bermejo J, Ramos-Luis E, Gil R, Bermejo AM, Lotufo-Neto F, Hirata MH, Brisighelli F, Paramo M, Carracedo A, Brion M. Genetic susceptibility in pharmacodynamic and pharmacokinetic pathways underlying drug-induced arrhythmia and sudden unexplained deaths. Forensic Sci Int Genet 2019; 42:203-212. [PMID: 31376648 DOI: 10.1016/j.fsigen.2019.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/14/2019] [Accepted: 07/14/2019] [Indexed: 12/21/2022]
Abstract
Drug-induced arrhythmia is an adverse drug reaction that can be potentially fatal since it is mostly related to drug-induced QT prolongation, a known risk factor for Torsade de Pointes and sudden cardiac death (SCD). Several risk factors have been described in association to these drug-induced events, such as preexistent cardiac disease and genetic variation. Our objective was to study the genetic susceptibility in pharmacodynamic and pharmacokinetic pathways underlying suspected drug-induced arrhythmias and sudden unexplained deaths in 32 patients. The genetic component in the pharmacodynamic pathway was studied by analysing 96 genes associated with higher risk of SCD through massive parallel sequencing. Pharmacokinetic-mediated genetic susceptibility was investigated by studying the genes encoding cytochrome P450 enzymes using medium-throughput genotyping. Pharmacodynamic analysis showed three probably pathogenic variants and 45 variants of uncertain significance in 28 patients, several of them previously described in relation to mild or late onset cardiomyopathies. These results suggest that genetic variants in cardiomyopathy genes, in addition to those related with channelopathies, could be relevant to drug-induced cardiotoxicity and contribute to the arrhythmogenic phenotype. Pharmacokinetic analysis showed three patients that could have an altered metabolism of the drugs they received involving CYP2C19 and/or CYP2D6, probably contributing to the arrhythmogenic phenotype. The study of genetic variants in both pharmacodynamic and pharmacokinetic pathways may be a useful strategy to understand the multifactorial mechanism of drug-induced events in both clinical practice and forensic field. However, it is necessary to comprehensively study and evaluate the contribution of the genetic susceptibility to drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- M Martinez-Matilla
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela (A Coruña), Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain.
| | - A Blanco-Verea
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela (A Coruña), Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - M Santori
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - J Ansede-Bermejo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Centro Nacional de Genotipado-CeGen-USC-PRB3-ISCIII, Santiago de Compostela, Spain
| | - E Ramos-Luis
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela (A Coruña), Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - R Gil
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela (A Coruña), Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - A M Bermejo
- Instituto de Ciencias Forenses "Luis Concheiro" (INCIFOR), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - F Lotufo-Neto
- Psiquiatry Institute - Faculty of Medicine at University of São Paulo, São Paulo, Brazil
| | - M H Hirata
- Institute Dante Pazzanese of Cardiology, São Paulo, Brazil; School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - F Brisighelli
- Institute of Public Health, Section of Legal Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - M Paramo
- Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - A Carracedo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Centro Nacional de Genotipado-CeGen-USC-PRB3-ISCIII, Santiago de Compostela, Spain
| | - M Brion
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela (A Coruña), Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Centro Nacional de Genotipado-CeGen-USC-PRB3-ISCIII, Santiago de Compostela, Spain
| |
Collapse
|
9
|
Mascia G, Arbelo E, Solimene F, Giaccardi M, Brugada R, Brugada J. The long-QT syndrome and exercise practice: The never-ending debate. J Cardiovasc Electrophysiol 2018; 29:489-496. [DOI: 10.1111/jce.13410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Giuseppe Mascia
- Electrophysiology Unit; Clinica Montevergine; Mercogliano Italy
| | - Elena Arbelo
- Arrhythmia Section, Cardiology Department, Hospital Clínic, IDIBAPS; University of Barcelona; Barcelona Spain
| | | | - Marzia Giaccardi
- Cardiology and Electrophysiology Unit, Department of Internal Medicine; Santa Maria Nuova Hospital; Florence Italy
| | - Ramon Brugada
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica Girona (IDIBGI); University of Girona; Girona Spain
| | - Josep Brugada
- Arrhythmia Section, Cardiology Department, Hospital Clínic, IDIBAPS; University of Barcelona; Barcelona Spain
| |
Collapse
|
10
|
Mademont-Soler I, Mates J, Yotti R, Espinosa MA, Pérez-Serra A, Fernandez-Avila AI, Coll M, Méndez I, Iglesias A, del Olmo B, Riuró H, Cuenca S, Allegue C, Campuzano O, Picó F, Ferrer-Costa C, Álvarez P, Castillo S, Garcia-Pavia P, Gonzalez-Lopez E, Padron-Barthe L, Díaz de Bustamante A, Darnaude MT, González-Hevia JI, Brugada J, Fernandez-Aviles F, Brugada R. Additional value of screening for minor genes and copy number variants in hypertrophic cardiomyopathy. PLoS One 2017; 12:e0181465. [PMID: 28771489 PMCID: PMC5542623 DOI: 10.1371/journal.pone.0181465] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited heart disease. Next-generation sequencing (NGS) is the preferred genetic test, but the diagnostic value of screening for minor and candidate genes, and the role of copy number variants (CNVs) deserves further evaluation. METHODS Three hundred and eighty-seven consecutive unrelated patients with HCM were screened for genetic variants in the 5 most frequent genes (MYBPC3, MYH7, TNNT2, TNNI3 and TPM1) using Sanger sequencing (N = 84) or NGS (N = 303). In the NGS cohort we analyzed 20 additional minor or candidate genes, and applied a proprietary bioinformatics algorithm for detecting CNVs. Additionally, the rate and classification of TTN variants in HCM were compared with 427 patients without structural heart disease. RESULTS The percentage of patients with pathogenic/likely pathogenic (P/LP) variants in the main genes was 33.3%, without significant differences between the Sanger sequencing and NGS cohorts. The screening for 20 additional genes revealed LP variants in ACTC1, MYL2, MYL3, TNNC1, GLA and PRKAG2 in 12 patients. This approach resulted in more inconclusive tests (36.0% vs. 9.6%, p<0.001), mostly due to variants of unknown significance (VUS) in TTN. The detection rate of rare variants in TTN was not significantly different to that found in the group of patients without structural heart disease. In the NGS cohort, 4 patients (1.3%) had pathogenic CNVs: 2 deletions in MYBPC3 and 2 deletions involving the complete coding region of PLN. CONCLUSIONS A small percentage of HCM cases without point mutations in the 5 main genes are explained by P/LP variants in minor or candidate genes and CNVs. Screening for variants in TTN in HCM patients drastically increases the number of inconclusive tests, and shows a rate of VUS that is similar to patients without structural heart disease, suggesting that this gene should not be analyzed for clinical purposes in HCM.
Collapse
Affiliation(s)
- Irene Mademont-Soler
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jesus Mates
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Raquel Yotti
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Maria Angeles Espinosa
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Alexandra Pérez-Serra
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ana Isabel Fernandez-Avila
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Monica Coll
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Irene Méndez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Anna Iglesias
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Bernat del Olmo
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Helena Riuró
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Sofía Cuenca
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Catarina Allegue
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - Ferran Picó
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | | | | | | | - Pablo Garcia-Pavia
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Inherited Cardiac Diseases Unit. Department of Cardiology. Hospital Universitario Puerta de Hierro, Francisco de Vitoria University, Madrid, Spain
| | - Esther Gonzalez-Lopez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Inherited Cardiac Diseases Unit. Department of Cardiology. Hospital Universitario Puerta de Hierro, Francisco de Vitoria University, Madrid, Spain
| | - Laura Padron-Barthe
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Inherited Cardiac Diseases Unit. Department of Cardiology. Hospital Universitario Puerta de Hierro, Francisco de Vitoria University, Madrid, Spain
| | | | | | | | - Josep Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Arrhythmia Unit, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Francisco Fernandez-Aviles
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense, Madrid, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Cardiovascular Genetics Unit, Hospital Universitari Dr. Josep Trueta, Girona, Spain
| |
Collapse
|
11
|
Weissler-Snir A, Gollob MH, Chauhan V, Care M, Spears DA. Evaluation of Prolonged QT Interval: Structural Heart Disease Mimicking Long QT Syndrome. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2017; 40:417-424. [PMID: 28155223 DOI: 10.1111/pace.13040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/20/2016] [Accepted: 01/08/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND In about 20-25% of patients with congenital long QT syndrome (LQTS) a causative pathogenic mutation is not found. The aim of this study was to explore the prevalence of alternative cardiac diagnoses among patients exhibiting prolongation of QT interval with negative genetic testing for LQTS genes. METHODS We conducted a retrospective analysis of 239 consecutive patients who were evaluated in the inherited arrhythmia clinic at the Toronto General Hospital between July 2013 and December 2015 for possible LQTS. A detailed review of the patients' charts, electrocardiograms, and imaging was carried out. RESULTS The analysis included 56 gene-negative patients and 61 gene-positive patients. Of the gene-negative group, 25% had structural heart disease compared to only 1.6% of gene-positive patients (P < 0.001). Structural heart disease was more likely if only one abnormal QTc parameter was found in the course of the evaluation (35.2% vs 9.1%, P = 0.01). The most common structural cardiac pathology was bileaflet mitral valve prolapse (8.9%). No gene-positive patient had episodes of nonsustained ventricular tachycardia, compared to seven of the gene-negative patients (0% vs 12.5%, P = 0.005). CONCLUSIONS Structural pathology was detected in a quarter of gene-negative patients evaluated for possible LQTS. Hence, cardiac imaging and Holter monitoring should be strongly encouraged to rule out structural heart disease in this population.
Collapse
Affiliation(s)
- Adaya Weissler-Snir
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Michael H Gollob
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Vijay Chauhan
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Melanie Care
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Danna A Spears
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Tp-e interval and Tp-e/QTc ratio as novel surrogate markers for prediction of ventricular arrhythmic events in hypertrophic cardiomyopathy. Anatol J Cardiol 2017; 18:48-53. [PMID: 28315570 PMCID: PMC5512198 DOI: 10.14744/anatoljcardiol.2017.7581] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) as a common genetic heart disease characterized by ventricular hypertrophy and myocardial fibrosis is significantly associated with a higher risk of fatal ventricular arrhythmic events (VAEs). We aimed to assess the interval between the peak and the end of the electrocardiographic T wave (Tp-e) and Tp-e/corrected QT (QTc) ratio as candidate markers of ventricular arrhythmias in patients with HCM. METHODS In this single-center, prospective study, a total of 66 patients with HCM and 88 controls were enrolled. The patients were divided into two groups: those with VAEs (n=26) and those without VAEs (n=40). Tp-e interval and Tp-e/QTc ratio were measured using a 12-lead electrocardiogram. RESULTS Tp-e interval was significantly longer and Tp-e/QTc ratio were significantly higher in HCM patients than in the controls. In correlation analysis, maximal left ventricular (LV) thickness also has a significant positive correlation with Tp-e interval (r=0.422, p<0.001) and Tp-e/QTc ratio (r=0.348, p<0.001). Finally, multivariable regression analysis showed that a history of syncope, Tp-e interval [OR (odds ratio): 1.060; 95% confidence interval (CI): 1.005-1.117); p=0.012], Tp-e/QTc ratio (OR: 1.148; 95% CI: 1.086-1.204); p=0.049], and maximal LV thickness were independent predictors of VAEs in patients with HCM. CONCLUSION Our findings suggested that prolonged Tp-e interval and increased Tp-e/QTc ratio may be good surrogate markers for the prediction of VAEs in HCM.
Collapse
|
13
|
Early remodeling of repolarizing K + currents in the αMHC 403/+ mouse model of familial hypertrophic cardiomyopathy. J Mol Cell Cardiol 2017; 103:93-101. [PMID: 28089740 DOI: 10.1016/j.yjmcc.2017.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 11/20/2022]
Abstract
Familial hypertrophic cardiomyopathy (HCM), linked to mutations in myosin, myosin-binding proteins and other sarcolemmal proteins, is associated with increased risk of life threatening ventricular arrhythmias, and a number of animal models have been developed to facilitate analysis of disease progression and mechanisms. In the experiments here, we use the αMHC403/+ mouse line in which one αMHC allele harbors a common HCM mutation (in βMHC, Arg403 Gln). Here, we demonstrate marked prolongation of QT intervals in young adult (10-12week) male αMHC403/+ mice, well in advance of the onset of measurable left ventricular hypertrophy. Electrophysiological recordings from myocytes isolated from the interventricular septum of these animals revealed significantly (P<0.001) lower peak repolarizing voltage-gated K+ (Kv) current (IK,peak) amplitudes, compared with cells isolated from wild type (WT) littermate controls. Analysis of Kv current waveforms revealed that the amplitudes of the inactivating components of the total outward Kv current, Ito,f, Ito,s and IK,slow, were significantly lower in αMHC403/+, compared with WT, septum cells, whereas Iss amplitudes were similar. The amplitudes/densities of IK,peak and IK,slow were also lower in αMHC403/+, compared with WT, LV wall and LV apex myocytes, whereas Ito,f was attenuated in αMHC403/+ LV wall, but not LV apex, cells. These regional differences in the remodeling of repolarizing Kv currents in the αMHC403/+ mice would be expected to increase the dispersion of ventricular repolarization and be proarrhythmic. Quantitative RT-PCR analysis revealed reductions in the expression of transcripts encoding several K+ channel subunits in the interventricular septum, LV free wall and LV apex of (10-12week) αMHC403/+ mice, although this transcriptional remodeling was not correlated with the observed decreases in K+ current amplitudes.
Collapse
|
14
|
Ceyhan-Birsoy O, Pugh TJ, Bowser MJ, Hynes E, Frisella AL, Mahanta LM, Lebo MS, Amr SS, Funke BH. Next generation sequencing-based copy number analysis reveals low prevalence of deletions and duplications in 46 genes associated with genetic cardiomyopathies. Mol Genet Genomic Med 2015; 4:143-51. [PMID: 27066507 PMCID: PMC4799872 DOI: 10.1002/mgg3.187] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/19/2015] [Accepted: 10/22/2015] [Indexed: 12/04/2022] Open
Abstract
Background Diagnostic testing for genetic cardiomyopathies has undergone dramatic changes in the last decade with next generation sequencing (NGS) expanding the number of genes that can be interrogated simultaneously. Exon resolution copy number analysis is increasingly incorporated into routine diagnostic testing via cytogenomic arrays and more recently via NGS. While NGS is an attractive option for laboratories that have no access to array platforms, its higher false positive rate requires weighing the added cost incurred by orthogonal confirmation against the magnitude of the increase in diagnostic yield. Although copy number variants (CNVs) have been reported in various cardiomyopathy genes, their contribution has not been systematically studied. Methods We performed single exon resolution NGS‐based deletion/duplication analysis for up to 46 cardiomyopathy genes in >1400 individuals with cardiomyopathies including HCM, DCM, ARVC, RCM, and LVNC. Results and Conclusion Clinically significant deletions and duplications were identified in only 9 of 1425 (0.63%) individuals. The majority of those (6/9) represented intragenic events. We conclude that the added benefit of exon level deletion/duplication analysis is low for currently known cardiomyopathy genes and may not outweigh the increased cost and complexity of incorporating it into routine diagnostic testing for these disorders.
Collapse
Affiliation(s)
- Ozge Ceyhan-Birsoy
- Laboratory for Molecular MedicinePartners HealthCare Personalized MedicineCambridgeMassachusetts; Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusetts
| | - Trevor J Pugh
- Department of Medical Biophysics Princess Margaret Cancer Centre University Health Network University of Toronto Toronto Ontario Canada
| | - Mark J Bowser
- Laboratory for Molecular Medicine Partners HealthCare Personalized Medicine Cambridge Massachusetts
| | - Elizabeth Hynes
- Laboratory for Molecular Medicine Partners HealthCare Personalized Medicine Cambridge Massachusetts
| | - Ashley L Frisella
- Laboratory for Molecular Medicine Partners HealthCare Personalized Medicine Cambridge Massachusetts
| | - Lisa M Mahanta
- Laboratory for Molecular Medicine Partners HealthCare Personalized Medicine Cambridge Massachusetts
| | - Matt S Lebo
- Laboratory for Molecular MedicinePartners HealthCare Personalized MedicineCambridgeMassachusetts; Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusetts
| | - Sami S Amr
- Laboratory for Molecular MedicinePartners HealthCare Personalized MedicineCambridgeMassachusetts; Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusetts
| | - Birgit H Funke
- Laboratory for Molecular MedicinePartners HealthCare Personalized MedicineCambridgeMassachusetts; Department of PathologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusetts
| |
Collapse
|
15
|
Debonnaire P, Katsanos S, Joyce E, VAN DEN Brink OVW, Atsma DE, Schalij MJ, Bax JJ, Delgado V, Marsan NA. QRS Fragmentation and QTc Duration Relate to Malignant Ventricular Tachyarrhythmias and Sudden Cardiac Death in Patients with Hypertrophic Cardiomyopathy. J Cardiovasc Electrophysiol 2015; 26:547-55. [PMID: 25648421 DOI: 10.1111/jce.12629] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND QRS fragmentation (fQRS) and prolonged QTc interval on surface ECG are prognostic in various cardiomyopathies other than hypertrophic cardiomyopathy (HCM). The association between fQRS and prolonged QTc duration with occurrence of ventricular tachyarrhythmias or sudden cardiac death (VTA/SCD) in patients with HCM was explored. METHODS AND RESULTS One hundred and ninety-five clinical HCM patients were studied. QTc duration was derived applying Bazett's formula; fQRS was defined as presence of various RSR' patterns, R or S notching and/or >1 additional R wave in any non-aVR lead in patients without pacing or (in)complete bundle branch block. The endpoints comprised SCD, ECG documented sustained VTA (tachycardia or fibrillation) or appropriate implantable cardioverter defibrillator (ICD) therapies (antitachycardia pacing [ATP] or shock) for VTA in ICD recipients (n = 58 [30%]). QT prolonging drugs recipients were excluded. After a median follow-up of 5.7 years (IQR 2.7-9.1), 26 (13%) patients experienced VTA or SCD. Patients with fQRS in ≥3 territories (inferior, lateral, septal, and/or anterior) (p = 0.004) or QTc ≥460 ms (p = 0.009) had worse cumulative survival free of VTA/SCD than patients with fQRS in <3 territories or QTc <460 ms. fQRS in ≥3 territories (ß 4.5, p = 0.020, 95%CI 1.41-14.1) and QTc ≥460 ms (ß 2.7, p = 0.037, 95%CI 1.12-6.33) were independently associated with VTA/SCD. Likelihood ratio test indicated assessment of fQRS and QTc on top of conventional SCD risk factors provides incremental predictive value for VTA/SCD (p = 0.035). CONCLUSIONS Both fQRS in ≥3 territories and QTc duration are associated with VTA/SCD in HCM patients, independently of and incremental to conventional SCD risk factors.
Collapse
Affiliation(s)
- Philippe Debonnaire
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands.,Sint-Jan Hospital Bruges, Department of Cardiology, Bruges, Belgium
| | - Spyridon Katsanos
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | - Emer Joyce
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | | | - Douwe E Atsma
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | - Martin J Schalij
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | - Jeroen J Bax
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | - Victoria Delgado
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| | - Nina Ajmone Marsan
- Leiden University Medical Centre, Department of Cardiology, Leiden, the Netherlands
| |
Collapse
|
16
|
Wen SN, Liu N, Li SN, Wu XY, Salim M, Kang JP, Ning M, Wu JH, Ruan YF, Yu RH, Long DY, Tang RB, Sang CH, Jiang CX, Bai R, Du X, Dong JZ, Liu XH, Ma CS. QTc Interval Prolongation Predicts Arrhythmia Recurrence After Catheter Ablation of Atrial Fibrillation in Patients With Hypertrophic Cardiomyopathy. Circ J 2015; 79:1024-30. [PMID: 25739859 DOI: 10.1253/circj.cj-14-1290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND In hypertrophic cardiomyopathy (HCM) patients complicated with atrial fibrillation (AF), catheter ablation has been recommended as a treatment option. Meanwhile, prolongation of QTc interval has been linked to an increased AF incidence in the general population and to poor outcomes in HCM patients. However, whether QTc prolongation predicts arrhythmia recurrence after AF ablation in the HCM population remains unknown. METHODS AND RESULTS Thirty-nine HCM patients undergoing primary AF ablation were enrolled. The ablation strategy included bilateral pulmonary vein isolation (PVI) for paroxysmal AF (n=27) and PVI plus left atrial roof, mitral isthmus and tricuspid isthmus linear ablations for persistent AF (n=12). Pre-procedural QTc was corrected by using the Bazett's formula. At a 14.8-month follow up, 23 patients experienced atrial tachyarrhythmia recurrence. Recurrent patients had longer QTc than non-recurrent patients (461.0±28.8 ms vs. 434.3±18.2 ms, P=0.002). QTc and left atrial diameter (LAD) were independent predictors of recurrence. The cut-off value of QTc 448 ms predicted arrhythmia recurrence with a sensitivity of 73.9% and a specificity of 81.2%. A combination of LAD and QTc (global chi-squared=13.209) was better than LAD alone (global chi-squared=6.888) or QTc alone (global chi-squared=8.977) in predicting arrhythmia recurrence after AF ablation in HCM patients. CONCLUSIONS QTc prolongation is an independent predictor of arrhythmia recurrence in HCM patients undergoing AF ablation, and might be useful for identifying those patients likely to have a better outcome following the procedure.
Collapse
Affiliation(s)
- Song-Nan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Grall S, Biere L, Clerfond G, Mateus V, Prunier F, Furber A. ECG characteristics according to the presence of late gadolinium enhancement on cardiac MRI in hypertrophic cardiomyopathy. Open Heart 2014; 1:e000101. [PMID: 25332813 PMCID: PMC4189227 DOI: 10.1136/openhrt-2014-000101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/29/2014] [Accepted: 07/15/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Late gadolinium enhancement (LGE) on cardiac MRI (CMR) has been described as an independent predictive factor of cardiovascular events among patients with hypertrophic cardiomyopathy (HCM). LGE and Q waves are considered as myocardial scar markers but their relation in the context of HCM is poorly established and has to be more supported. The objective of the study was to compare ECG findings in the presence or absence of LGE. METHODS 42 patients with HCM confirmed by CMR were included in the study. ECG abnormalities including abnormal Q waves and five ECG scores of left ventricular hypertrophy were assessed and compared according to LGE presence and its extension. Some CMR features, such as septal to posterior wall thickness ratio, were also studied according to the presence of LGE and the presence of abnormal Q waves. RESULTS Abnormal Q waves were more prevalent in the LGE (+) group (60% vs 12%; p=0.002), but there was no correlation between location of Q waves on ECG and territory of LGE on CMR. Among patients with LGE, quantitative analysis of LGE was not different in the presence or absence of Q waves. In contrast to the LGE mass, septal to posterior wall thickness was higher in patients with abnormal Q waves (2.3±0.7 vs 1.6±0.5; p=0.012). CONCLUSIONS Although abnormal Q waves were more prevalent in the presence of LGE, no correlation was found with the LGE location and extent. These data suggest that abnormal electrical activation of the hypertrophied ventricular septum represented by a high septal to posterior wall thickness ratio seems to be an important mechanism of abnormal Q waves in HCM.
Collapse
Affiliation(s)
- Sylvain Grall
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| | - Loïc Biere
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| | - Guillaume Clerfond
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| | - Victor Mateus
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| | - Fabrice Prunier
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| | - Alain Furber
- Université d'Angers, Laboratoire “Cardioprotection, Remodelage et Thrombose”, Angers, France
- Centre Hospitalier Universitaire d'Angers, Service de Cardiologie, Angers, France
| |
Collapse
|
18
|
Polak S, Wiśniowska B, Fijorek K, Glinka A, Mendyk A. In vitro-in vivo extrapolation of drug-induced proarrhythmia predictions at the population level. Drug Discov Today 2013; 19:275-81. [PMID: 24140591 DOI: 10.1016/j.drudis.2013.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 09/16/2013] [Accepted: 10/09/2013] [Indexed: 01/25/2023]
Abstract
Drug cardiotoxicity is a serious issue for patients, regulators, pharmaceutical companies and health service payers because they are all affected by its consequences. Despite the wide range of data they generate, existing approaches for cardiac safety testing might not be adequate and sufficiently cost-effective, probably as a result of the complexity of the problem. For this reason, translational tools (based on biophysically detailed, mathematical models) allowing for in vitro-in vivo extrapolation are gaining increasing interest. This current review describes approaches that can be used for cardiac safety assessment at the population level, by accounting for various sources of variability including kinetics of the compound of interest.
Collapse
Affiliation(s)
- Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland; Simcyp Limited, Blades Enterprise Centre, John Street, Sheffield, UK.
| | - Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
| | - Kamil Fijorek
- Department of Statistics, Faculty of Management, Cracow University of Economics, Rakowicka 27 Street, 31-510 Kraków, Poland
| | - Anna Glinka
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
| | - Aleksander Mendyk
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
| |
Collapse
|
19
|
Cheng Y, Wan X, McElfresh TA, Chen X, Gresham KS, Rosenbaum DS, Chandler MP, Stelzer JE. Impaired contractile function due to decreased cardiac myosin binding protein C content in the sarcomere. Am J Physiol Heart Circ Physiol 2013; 305:H52-65. [PMID: 23666674 DOI: 10.1152/ajpheart.00929.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutations in cardiac myosin binding protein C (MyBP-C) are a common cause of familial hypertrophic cardiomyopathy (FHC). The majority of MyBP-C mutations are expected to reduce MyBP-C expression; however, the consequences of MyBP-C deficiency on the regulation of myofilament function, Ca²⁺ homeostasis, and in vivo cardiac function are unknown. To elucidate the effects of decreased MyBP-C expression on cardiac function, we employed MyBP-C heterozygous null (MyBP-C+/-) mice presenting decreases in MyBP-C expression (32%) similar to those of FHC patients carrying MyBP-C mutations. The levels of MyBP-C phosphorylation were reduced 53% in MyBP-C+/- hearts compared with wild-type hearts. Skinned myocardium isolated from MyBP-C+/- hearts displayed decreased cross-bridge stiffness at half-maximal Ca²⁺ activations, increased steady-state force generation, and accelerated rates of cross-bridge recruitment at low Ca²⁺ activations (<15% and <25% of maximum, respectively). Protein kinase A treatment abolished basal differences in rates of cross-bridge recruitment between MyBP-C+/- and wild-type myocardium. Intact ventricular myocytes from MyBP-C+/- hearts displayed abnormal sarcomere shortening but unchanged Ca²⁺ transient kinetics. Despite a lack of left ventricular hypertrophy, MyBP-C+/- hearts exhibited elevated end-diastolic pressure and decreased peak rate of LV pressure rise, which was normalized following dobutamine infusion. Furthermore, electrocardiogram recordings in conscious MyBP-C+/- mice revealed prolonged QRS and QT intervals, which are known risk factors for cardiac arrhythmia. Collectively, our data show that reduced MyBP-C expression and phosphorylation in the sarcomere result in myofilament dysfunction, contributing to contractile dysfunction that precedes compensatory adaptations in Ca²⁺ handling, and chamber remodeling. Perturbations in mechanical and electrical activity in MyBP-C+/- mice could increase their susceptibility to cardiac dysfunction and arrhythmia.
Collapse
Affiliation(s)
- Y Cheng
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Gray B, Ingles J, Medi C, Semsarian C. Prolongation of the QTc Interval Predicts Appropriate Implantable Cardioverter-Defibrillator Therapies in Hypertrophic Cardiomyopathy. JACC-HEART FAILURE 2013; 1:149-55. [DOI: 10.1016/j.jchf.2013.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 11/30/2022]
|
21
|
Parvatiyar MS, Landstrom AP, Figueiredo-Freitas C, Potter JD, Ackerman MJ, Pinto JR. A mutation in TNNC1-encoded cardiac troponin C, TNNC1-A31S, predisposes to hypertrophic cardiomyopathy and ventricular fibrillation. J Biol Chem 2012; 287:31845-55. [PMID: 22815480 DOI: 10.1074/jbc.m112.377713] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Defined as clinically unexplained hypertrophy of the left ventricle, hypertrophic cardiomyopathy (HCM) is traditionally understood as a disease of the cardiac sarcomere. Mutations in TNNC1-encoded cardiac troponin C (cTnC) are a relatively rare cause of HCM. Here, we report clinical and functional characterization of a novel TNNC1 mutation, A31S, identified in a pediatric HCM proband with multiple episodes of ventricular fibrillation and aborted sudden cardiac death. Diagnosed at age 5, the proband is family history-negative for HCM or sudden cardiac death, suggesting a de novo mutation. TnC-extracted cardiac skinned fibers were reconstituted with the cTnC-A31S mutant, which increased Ca(2+) sensitivity with no effect on the maximal contractile force generation. Reconstituted actomyosin ATPase assays with 50% cTnC-A31S:50% cTnC-WT demonstrated Ca(2+) sensitivity that was intermediate between 100% cTnC-A31S and 100% cTnC-WT, whereas the mutant increased the activation of the actomyosin ATPase without affecting the inhibitory qualities of the ATPase. The secondary structure of the cTnC mutant was evaluated by circular dichroism, which did not indicate global changes in structure. Fluorescence studies demonstrated increased Ca(2+) affinity in isolated cTnC, the troponin complex, thin filament, and to a lesser degree, thin filament with myosin subfragment 1. These results suggest that this mutation has a direct effect on the Ca(2+) sensitivity of the myofilament, which may alter Ca(2+) handling and contribute to the arrhythmogenesis observed in the proband. In summary, we report a novel mutation in the TNNC1 gene that is associated with HCM pathogenesis and may predispose to the pathogenesis of a fatal arrhythmogenic subtype of HCM.
Collapse
Affiliation(s)
- Michelle S Parvatiyar
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | | | | | | | | | | |
Collapse
|
22
|
Steffel J, Hürlimann D, Namdar M, Despotovic D, Kobza R, Wolber T, Holzmeister J, Haegeli L, Brunckhorst C, Lüscher TF, Jenni R, Duru F. Long-term follow-up of patients with isolated left ventricular noncompaction: role of electrocardiography in predicting poor outcome. Circ J 2011; 75:1728-34. [PMID: 21617326 DOI: 10.1253/circj.cj-10-1217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Abnormal baseline electrocardiograms (ECGs) are common in patients with isolated left ventricular noncompaction (IVNC). Whether certain electrocardiographic parameters are associated with a poor clinical outcome, however, remains elusive. The present study was therefore designed to comprehensively assess the predictive value of baseline ECG findings in patients newly diagnosed with IVNC. METHODS AND RESULTS 74 patients diagnosed with IVNC were included in the analysis. During follow-up, 8 patients (11%) died of a cardiovascular cause or underwent heart transplantation (primary outcome measure). On univariate analysis, several variables, including repolarization abnormalities (ST segment elevation/depression, T-wave inversion) in the inferior leads (5-year estimator: 67.1 ± 10.7% vs. 98 ± 2.2%; P = 0.001), an increase in PQ- (hazard ratio (HR) 1.032, P=0.004) and QTc-duration (HR 1.037, P=0.001), were predictive of cardiovascular death or heart transplantation. On multivariate analysis, only PQ- and QTc-duration and the presence of repolarization abnormalities in the inferior leads remained significantly predictive of a poor outcome. CONCLUSIONS PQ duration, QTc duration, and repolarization abnormalities in the inferior leads are independently predictive of a poor prognosis in IVNC. Further prospective studies are required to conclusively investigate the usefulness of baseline ECG parameters for risk stratification in patients with IVNC.
Collapse
Affiliation(s)
- Jan Steffel
- Clinic for Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Johnson JN, Grifoni C, Bos JM, Saber-Ayad M, Ommen SR, Nistri S, Cecchi F, Olivotto I, Ackerman MJ. Prevalence and clinical correlates of QT prolongation in patients with hypertrophic cardiomyopathy. Eur Heart J 2011; 32:1114-20. [PMID: 21345853 DOI: 10.1093/eurheartj/ehr021] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIMS Congenital or acquired QT prolongation is a risk factor for life-threatening arrhythmias. In patients with hypertrophic cardiomyopathy (HCM), the QT interval may be intrinsically prolonged. However, the prevalence, cause, and significance of QT prolongation among patients with HCM are unknown. METHODS AND RESULTS After exclusion of patients on QT-prolonging drugs, a blinded, retrospective analysis of electrocardiograms, echocardiograms, and genotype status in 479 unrelated patients with HCM [201 females, age at diagnosis 41 ± 18 years, maximal left ventricular wall thickness (MLVWT) 22 ± 6 mm] from two independent centres was performed. The mean QTc was 440 ± 28 ms. The QTc exceeded 480 ms in 13% of patients. Age, gender, family history of HCM or sudden cardiac arrest, and genotype status had no association with QTc. Patients with a QTc over 480 ms were more symptomatic at diagnosis (P < 0.001), had a higher MLVWT (P = 0.03), were more obstructive (P < 0.001), and were more likely to have undergone septal reduction therapy (P = 0.02). There was a weak but significant direct linear relationship between QTc and peak outflow gradient (r(2) = 0.05, P < 0.0001). CONCLUSIONS Compared with <1 in 200 otherwise healthy adults, QT prolongation (QTc > 480 ms) was present in 1 out of 8 patients with HCM. The QTc was partly reflective of the degree of cardiac hypertrophy and left ventricular outflow tract obstruction. Because of its pro-arrhythmic potential and its potential relevance to management and risk stratification, routine QTc assessment should be performed in patients with HCM, particularly when concomitant use of QT-prolonging medications is considered.
Collapse
Affiliation(s)
- Jonathan N Johnson
- Department of Pediatrics/Division of Pediatric Cardiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Congenital long QT syndrome (LQTS) affects an estimated 1 in 2500 people and typically presents with syncope, seizures or sudden death. Whereas someone exhibiting marked prolongation of the QT interval with QTc exceeding 500 ms who was just externally defibrillated from torsades de pointes while swimming poses negligible diagnostic challenge as to the unequivocal probability of LQTS, the certainty is considerably less for the otherwise asymptomatic person who happens to host a QTc value coined "borderline" (QTc > or = 440 ms). Although a normal QT interval imparts a much lower risk of life-threatening events, it does not preclude a patient from nevertheless harbouring a potentially lethal LQTS-causing genetic mutation. Indeed, genetic testing exerts significant diagnostic, prognostic and therapeutic implications. However, the 12-lead ECG remains the universal initial diagnostic test in the evaluation of LQTS and is subject to miscalculation, misinterpretation and mishandling. This review discusses the components of accurate QTc measurement and diagnosis, re-examines what is known about factors affecting QT interval measurement, and clarifies current recommendations regarding diagnosis of so-called "borderline" QT interval prolongation. The current guideline recommendations for the athlete with LQTS are also summarised.
Collapse
Affiliation(s)
- J N Johnson
- Department of Pediatrics/Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota, USA
| | - M J Ackerman
- Department of Pediatrics/Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Medicine/Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
25
|
Uchiyama K, Hayashi K, Fujino N, Konno T, Sakamoto Y, Sakata K, Kawashiri MA, Ino H, Yamagishi M. Impact of QT variables on clinical outcome of genotyped hypertrophic cardiomyopathy. Ann Noninvasive Electrocardiol 2009; 14:65-71. [PMID: 19149795 DOI: 10.1111/j.1542-474x.2008.00275.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Although QT variables such as its interval and/or dispersion can be clinical markers of ventricular tachyarrhythmia, few data exist regarding the role of QT variables in genotyped hypertrophic cardiomyopathy (HCM). Therefore, we analyzed QT variables in genotyped subjects with or without left ventricular hypertrophy (LVH). METHODS QT variables were analyzed in 111 mutation and 43 non-mutation carriers who were divided into three groups: A, those without ECG abnormalities and echocardiographically determined LVH (wall thickness > or =13 mm); B, those with ECG abnormalities but LVH; and C, those with ECG abnormalities and LVH. We also examined clinical outcome of enrolled patients. RESULTS Maximal LV wall thickness in group C (19.0 +/- 4.3 mm, mean +/-SD) was significantly greater than that in group A (9.2 +/- 1.8) and group B (10.4 +/- 1.8). Under these conditions, maximum QTc interval and QT dispersion were significantly longer in group C than those in group A (438 +/- 38 ms vs 406 +/- 30 and 64 +/- 31 vs 44 +/- 18, respectively; P < 0.05). QTc interval and QT dispersion in group B (436 +/- 50 and 64 +/- 22 ms) were also significantly greater than those in group A. During follow-up periods, four sudden cardiac deaths and one ventricular fibrillation were observed in group C, and two nonlethal ventricular tachyarrhythmias were observed in group B. CONCLUSIONS Patients with HCM-related gene mutation accompanying any ECG abnormalities frequently exhibited impaired QT variables even without LVH. We suggest that careful observation should be considered for those genotyped subjects.
Collapse
Affiliation(s)
- Katsuharu Uchiyama
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Atmaca M, Yavuzkir M, Mermi O, Topuz M, Kanmaz E, Tezcan E. Effect of sertindole on QTc interval in patients with schizophrenia. Neurosci Lett 2008; 442:1-3. [PMID: 18639380 DOI: 10.1016/j.neulet.2008.05.065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 05/20/2008] [Accepted: 05/20/2008] [Indexed: 10/22/2022]
Abstract
Sertindole has been marketed and offered daily clinical practice only for 9 months in our country, so no data has been its QTc prolongation potential. In the present study, we performed a clinical trial to investigate the effects of sertindole on QTc in patients with schizophrenia. The study comprised 21 patients with schizophrenia. Sertindole was administered in the following dosing regime: treatment was initiated with 4 mg/day sertindole. From day 3 to day 6, the dose was increased to 8 mg/day, and up to day 9, it was raised to 12 mg/day. The protocol allowed up to dose of 20mg/day according to effectiveness and tolerability. QTc values were determined at beginning, months 3 and 6. In addition, Positive and Negative Syndrome Scale (PANSS) were scored concomitantly. At the beginning of 6-month period, the mean QTc interval of patients was 391.7+/-19.2 ms. At the end of this period, it was 402.8+/-23.8 ms. Although the mean QTc interval changing was significant throughout 6-month period, of the patients, at any evaluation point, only 1 female (451 ms) and 1 male (433 ms) had borderline prolongation at month 3 for both, without any exceeding the dangerous limits. In summary, our results suggest that sertindole is tolerable and despite dose-related QT prolongation, sertindole had not the proarrhythmic profile. Future studies with larger sample evaluating the effects of treatment are required.
Collapse
Affiliation(s)
- Murad Atmaca
- Department of Psychiatry, Firat University, School of Medicine, Elazig, Turkey.
| | | | | | | | | | | |
Collapse
|
27
|
Shin JG, Kang WK, Shon JH, Arefayene M, Yoon YR, Kim KA, Kim DI, Kim DS, Cho KH, Woosley RL, Flockhart DA. Possible interethnic differences in quinidine-induced QT prolongation between healthy Caucasian and Korean subjects. Br J Clin Pharmacol 2006; 63:206-15. [PMID: 17096683 PMCID: PMC2000575 DOI: 10.1111/j.1365-2125.2006.02793.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS The aim of this study was to evaluate the pharmacokinetics and pharmacodynamics of quinidine-induced QT prolongation in healthy Caucasian and Korean subjects to investigate interethnic differences in susceptibility to drug-induced arrhythmia. METHODS A randomized, double-blind crossover study was conducted in 24 (12 male and 12 female) Korean and 13 (seven male and six female) Caucasian subjects. After a 20 min infusion of quinidine (4 mg kg(-1)) or saline, the serum concentration of quinidine and the QT interval corrected by Bazett's formula (QTc) were monitored. The dynamic data were analyzed by means of a population modelling approach using NONMEM. RESULTS There were no statistical differences in the pharmacokinetic profiles of quinidine between ethnic groups. The QTc values in Caucasians were higher than those in Koreans at the same quinidine concentrations, especially at higher quinidine concentrations and in female subjects. According to an E(max) model [equation: see text], the population modelling approach revealed that E0 (ms) was related to gender (408 + [34*(1 - Sex)]; 1 for male and 0 for female), DeltaE(max) (ms) was related to ethnicity ((136*f(ETHN)) + C(female): f(ETHN) = 1 for Koreans and 1.26 for Caucasians; C(female) was 106 only for Caucasian females), and EC50 was estimated to be 3.13 microm. CONCLUSIONS These results suggest that Korean subjects were less sensitive to quinidine-induced QT prolongation than Caucasian subjects, and that this trend was particularly true for females. Further population-based studies are merited to characterize more completely the ethnic differences in drug-induced QT prolongation between Asians and other ethnic groups.
Collapse
Affiliation(s)
- Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Centre, Inje University College of Medicine, Busan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Faltaos DW, Urien S, Carreau V, Chauvenet M, Hulot JS, Giral P, Bruckert E, Lechat P. Use of an indirect effect model to describe the LDL cholesterol-lowering effect by statins in hypercholesterolaemic patients. Fundam Clin Pharmacol 2006; 20:321-30. [PMID: 16671968 DOI: 10.1111/j.1472-8206.2006.00404.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Statins are the most commonly prescribed agents for the treatment of hypercholesterolaemia. This is due to their efficacy in reducing low-density lipoprotein cholesterol (LDL) level which is the primary goal of the treatment especially for patients with multiple risk factors or with established coronary heart diseases. The purpose of this study was to develop a pharmacokinetic/pharmacodynamic (PK/PD) model that describes the LDL-lowering process in patients with hypercholesterolaemia treated with atorvastatin, fluvastatin or simvastatin. A total of 100 patients were studied retrospectively. They received atorvastatin (n = 57), fluvastatin (n = 26) or simvastatin (n = 17). As no pharmacokinetic data were available, the absorption rate was fixed to 1/h and atorvastatin, simvastatin and fluvastatin elimination half-lives were fixed to 14, 2 and 2.5 h respectively. A total of 309 LDL levels were measured and the data were analysed by nonmem v. The time course of the LDL-lowering effect of statins was described by an indirect-response model with precursor (LDL synthesis, input rate K(in)) and response (circulating LDL, input and output rates K) compartments. The following parameters were estimated: LDL input rate (K(in)) 0.14 +/- 0.015 g/L/day (mean +/- SD); inhibition fraction of K(in) (INH) 0.21 +/- 0.017; and dose producing 50% increase of LDL removal (D50), 26 +/- 7.8, 1.3 +/- 0.48 and 15 +/- 5.25 mg for atorvastatin, simvastatin and fluvastatin, respectively. Gender, bodyweight, age, calories/day, sugar/day, lipids/day, hyperlipidaemia types and waist/hip circumference, renal and hepatic functions had no effect on the pharmacodynamic parameters. The pharmacodynamic parameters for the three statins were accurately estimated. The PK/PD model developed successfully predicted the time course of the LDL-lowering effect of statins.
Collapse
Affiliation(s)
- Demiana William Faltaos
- Pharmacology Department, Pitié-Salpêtrière University Hospital, Assistance-Publique Hôpitaux de Paris, Paris 6 University, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Kaneda T, Shimizu M, Ino H, Yamaguchi M, Terai H, Fujino N, Nagata M, Sakata K, Mabuchi H. Changes in cardiac tissue characterization in carriers with gene mutations associated with hypertrophic cardiomyopathy. Int J Cardiol 2005; 104:170-5. [PMID: 16168810 DOI: 10.1016/j.ijcard.2004.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Revised: 10/24/2004] [Accepted: 11/07/2004] [Indexed: 11/27/2022]
Abstract
BACKGROUND Early detection in patients with hypertrophic cardiomyopathy (HCM) is very important. Integrated backscatter (IB) provides a useful noninvasive measure of the acoustic properties of the myocardium, and may detect early myocardial changes. METHODS Thirty-four carriers who had gene mutations causing HCM were studied. The patients were divided into three groups as follows: (1) 21 patients with wall hypertrophy (Group A), (2) 7 patients with ECG abnormalities but without wall hypertrophy (Group B), and (3) 6 carriers with neither ECG abnormalities nor wall hypertrophy (Group C). All subjects underwent ECG, conventional echocardiography and acoustic densitometry. In addition, we studied subjects < or =20 years old from Groups B and C (Group B-2 and Group C-2, respectively), and compared them with control subjects with no cardiac disorders who were < or =20 years old. RESULTS In Group A, cyclic variations of integrated backscatter (CV-IB) in the interventricular septum and left ventricular posterior wall were significantly smaller than in Group C. The amplitude of IB in the interventricular septum and left ventricular posterior wall in Group A was significantly higher than those in Group C. Even in Group B, CV-IB in the interventricular septum was significantly smaller than those in Group C. Among patients < or =20 years old, CV-IB in the interventricular septum was significantly smaller in Group B-2 than in control subjects, while that in Group C-2 did not differ from that in control subjects. CONCLUSIONS Changes in tissue characterization were found in the hearts of HCM gene carriers even in the absence of wall hypertrophy. These results suggest that tissue changes detectable by the acoustic densitometry methods may occur in the hearts of HCM gene carriers without wall hypertrophy, and that they may be detectable at the time of appearance of ECG abnormalities.
Collapse
Affiliation(s)
- Tomoya Kaneda
- Molecular Genetics of Cardiovascular Disorders, Division of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, Takara-machi 13-1, Kanazawa 920-8640, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|