51
|
Ramakrishna H, O’Hare M, Mookadam F, Gutsche JT, Shah R, Augoustides JG. Sudden Cardiac Death and Disorders of the QT Interval: Anesthetic Implications and Focus on Perioperative Management. J Cardiothorac Vasc Anesth 2015; 29:1723-33. [DOI: 10.1053/j.jvca.2015.07.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Indexed: 12/19/2022]
|
52
|
Hassinen M, Haverinen J, Vornanen M. Molecular basis and drug sensitivity of the delayed rectifier (IKr) in the fish heart. Comp Biochem Physiol C Toxicol Pharmacol 2015. [PMID: 26215639 DOI: 10.1016/j.cbpc.2015.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Fishes are increasingly used as models for human cardiac diseases, creating a need for a better understanding of the molecular basis of fish cardiac ion currents. To this end we cloned KCNH6 channel of the crucian carp (Carassius carassius) that produces the rapid component of the delayed rectifier K(+) current (IKr), the main repolarising current of the fish heart. KCNH6 (ccErg2) was the main isoform of the Kv11 potassium channel family with relative transcript levels of 98.9% and 99.6% in crucian carp atrium and ventricle, respectively. KCNH2 (ccErg1), an orthologue to human cardiac Erg (Herg) channel, was only slightly expressed in the crucian carp heart. The native atrial IKr and the cloned ccErg2 were inhibited by similar concentrations of verapamil, terfenadine and KB-R7943 (P>0.05), while the atrial IKr was about an order of magnitude more sensitive to E-4031 than ccErg2 (P<0.05) suggesting that some accessory β-subunits may be involved. Sensitivity of the crucian carp atrial IKr to E-4031, terfenadine and KB-R7943 was similar to what has been reported for the Herg channel. In contrast, the sensitivity of the crucian carp IKr to verapamil was approximately 30 times higher than the previously reported values for the Herg current. In conclusion, the cardiac IKr is produced by non-orthologous gene products in fish (Erg2) and mammalian hearts (Erg1) and some marked differences exist in drug sensitivity between fish and mammalian Erg1/2 which need to be taken into account when using fish heart as a model for human heart.
Collapse
Affiliation(s)
- Minna Hassinen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland.
| | - Jaakko Haverinen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland
| | - Matti Vornanen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland
| |
Collapse
|
53
|
Liu WJ, Deng JX, Wang G, Gao KP, Lin ZX, Liu SY, Wang YH, Liu J. Manipulation of KCNE2 expression modulates action potential duration and Ito and IK in rat and mouse ventricular myocytes. Am J Physiol Heart Circ Physiol 2015; 309:H1288-302. [PMID: 26297229 DOI: 10.1152/ajpheart.00757.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 08/19/2015] [Indexed: 11/22/2022]
Abstract
In heterologous expression systems, KCNE2 has been demonstrated to interact with multiple α-subunits of voltage-dependent cation channels and modulate their functions. However, the physiological and pathological roles of KCNE2 in cardiomyocytes are poorly understood. The present study aimed to investigate the effects of bidirectional modulation of KCNE2 expression on action potential (AP) duration (APD) and voltage-dependent K+ channels in cardiomyocytes. Adenoviral gene delivery and RNA interference were used to either increase or decrease KCNE2 expression in cultured neonatal and adult rat or neonatal mouse ventricular myocytes. Knockdown of KCNE2 prolonged APD in both neonatal and adult myocytes, whereas overexpression of KCNE2 shortened APD in neonatal but not adult myocytes. Consistent with the alterations in APD, KCNE2 knockdown decreased transient outward K+ current ( Ito) densities in neonatal and adult myocytes, whereas KCNE2 overexpression increased Ito densities in neonatal but not adult myocytes. Furthermore, KCNE2 knockdown accelerated the rates of Ito activation and inactivation, whereas KCNE2 overexpression slowed Ito gating kinetics in neonatal but not adult myocytes. Delayed rectifier K+ current densities were remarkably affected by manipulation of KCNE2 expression in mouse but not rat cardiomyocytes. Simulation of the AP of a rat ventricular myocyte with a mathematical model showed that alterations in Ito densities and gating properties can result in similar APD alterations in KCNE2 overexpression and knockdown cells. In conclusion, endogenous KCNE2 in cardiomyocytes is important in maintaining cardiac electrical stability mainly by regulating Ito and APD. Perturbation of KCNE2 expression may predispose the heart to ventricular arrhythmia by prolonging APD.
Collapse
Affiliation(s)
- Wen-juan Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Jian-xin Deng
- Department of Endocrinology, The First Affiliated Hospital of Shenzhen University, Shenzhen No. 2 People's Hospital, Shenzhen, China
| | - Gang Wang
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Kai-ping Gao
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Ze-xun Lin
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Shuai-ye Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Yong-hui Wang
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China; and
| |
Collapse
|
54
|
Romero L, Trenor B, Yang PC, Saiz J, Clancy CE. In silico screening of the impact of hERG channel kinetic abnormalities on channel block and susceptibility to acquired long QT syndrome. J Mol Cell Cardiol 2015; 87:271-82. [DOI: 10.1016/j.yjmcc.2015.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
55
|
Abbott GW. KCNE1 and KCNE3: The yin and yang of voltage-gated K(+) channel regulation. Gene 2015; 576:1-13. [PMID: 26410412 DOI: 10.1016/j.gene.2015.09.059] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 09/03/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022]
Abstract
The human KCNE gene family comprises five genes encoding single transmembrane-spanning ion channel regulatory subunits. The primary function of KCNE subunits appears to be regulation of voltage-gated potassium (Kv) channels, and the best-understood KCNE complexes are with the KCNQ1 Kv α subunit. Here, we review the often opposite effects of KCNE1 and KCNE3 on Kv channel biology, with an emphasis on regulation of KCNQ1. Slow-activating IKs channel complexes formed by KCNQ1 and KCNE1 are essential for human ventricular myocyte repolarization, while constitutively active KCNQ1-KCNE3 channels are important in the intestine. Inherited sequence variants in human KCNE1 and KCNE3 cause cardiac arrhythmias but by different mechanisms, and each is important for hearing in unique ways. Because of their contrasting effects on KCNQ1 function, KCNE1 and KCNE3 have proved invaluable tools in the mechanistic understanding of how channel gating can be manipulated, and each may also provide a window into novel insights and new therapeutic opportunities in K(+) channel pharmacology. Finally, findings from studies of Kcne1(-/-) and Kcne3(-/-) mouse lines serve to illustrate the complexity of KCNE biology and KCNE-linked disease states.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Pharmacology and Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA; 360 Medical Surge II, Dept. of Pharmacology, School of Medicine, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
56
|
Fermini B, Hancox JC, Abi-Gerges N, Bridgland-Taylor M, Chaudhary KW, Colatsky T, Correll K, Crumb W, Damiano B, Erdemli G, Gintant G, Imredy J, Koerner J, Kramer J, Levesque P, Li Z, Lindqvist A, Obejero-Paz CA, Rampe D, Sawada K, Strauss DG, Vandenberg JI. A New Perspective in the Field of Cardiac Safety Testing through the Comprehensive In Vitro Proarrhythmia Assay Paradigm. ACTA ACUST UNITED AC 2015; 21:1-11. [PMID: 26170255 DOI: 10.1177/1087057115594589] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/11/2015] [Indexed: 12/31/2022]
Abstract
For the past decade, cardiac safety screening to evaluate the propensity of drugs to produce QT interval prolongation and Torsades de Pointes (TdP) arrhythmia has been conducted according to ICH S7B and ICH E14 guidelines. Central to the existing approach are hERG channel assays and in vivo QT measurements. Although effective, the present paradigm carries a risk of unnecessary compound attrition and high cost, especially when considering costly thorough QT (TQT) studies conducted later in drug development. The C: omprehensive I: n Vitro P: roarrhythmia A: ssay (CiPA) initiative is a public-private collaboration with the aim of updating the existing cardiac safety testing paradigm to better evaluate arrhythmia risk and remove the need for TQT studies. It is hoped that CiPA will produce a standardized ion channel assay approach, incorporating defined tests against major cardiac ion channels, the results of which then inform evaluation of proarrhythmic actions in silico, using human ventricular action potential reconstructions. Results are then to be confirmed using human (stem cell-derived) cardiomyocytes. This perspective article reviews the rationale, progress of, and challenges for the CiPA initiative, if this new paradigm is to replace existing practice and, in time, lead to improved and widely accepted cardiac safety testing guidelines.
Collapse
Affiliation(s)
| | - Jules C Hancox
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - Najah Abi-Gerges
- Translational Safety, Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca R&D, Macclesfield, UK AnaBios Corporation, San Diego, CA, USA
| | - Matthew Bridgland-Taylor
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca R&D, Macclesfield, UK
| | | | - Thomas Colatsky
- Division of Applied Regulatory Science, CDER, US Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - Bruce Damiano
- Global Safety Pharmacology, Discovery Sciences, Janssen Research & Development LLC, Spring House, PA, USA
| | - Gul Erdemli
- Center for Proteomic Chemistry, Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, USA
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences & Technology, AbbVie, North Chicago, IL, USA
| | - John Imredy
- Department of Safety Assessment, Merck & Co, Kenilworth, NJ, USA
| | - John Koerner
- Division of Cardiovascular and Renal Products, CDER, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - James Kramer
- ChanTest, A Charles River Company, Cleveland, OH, USA
| | - Paul Levesque
- Bristol Myers Squibb Research & Development, Princeton, NJ, USA
| | - Zhihua Li
- Division of Applied Regulatory Science, CDER, US Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - David Rampe
- Preclinical Safety, Sanofi, Bridgewater, NJ, USA
| | - Kohei Sawada
- Global Cardiovascular Assessment, Eisai Co., Ltd., Ibaraki, Japan
| | - David G Strauss
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, St Vincent's Clinical School, University of NSW, Darlinghurst, NSW, Australia
| |
Collapse
|
57
|
Lee J, Lee KI, Chung HM. Investigation of Transcriptional Gene Profiling in Normal Murine Hair Follicular Substructures Using Next-Generation Sequencing to Provide Potential Insights Into Skin Disease. Cell Transplant 2015; 25:377-99. [PMID: 25995029 DOI: 10.3727/096368915x688227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Skin diseases, including hair-related diseases and neoplasia, are a major public health problem. While their prevalence is increasing, their treatment options are limited. Researchers have tried to investigate the genes and signal pathways underlying hair follicles (HFs) to develop genetically targeted therapies through microarrays, which represent an appropriate modality for the analysis of small genomes. To enable the comprehensive transcriptome analysis of large and/or complex transcriptomes, we performed RNA-seq using next-generation sequencing (NGS). We isolated interfollicular keratinocytes (IFKs), HFs, and dermal fibroblasts including dermal papilla cells (DFs-DPCs) from normal C57BL/6 murine skin, transplanted combinations of these samples into nude mice, and followed the mice over time. Sustained hair growth was supported by HFs and DFs-DPCs. We then investigated the pathways and the relevant gene ontology associated with any identified differentially expressed genes (DEGs). In addition, in the culture and flow cytometry (FCM), the HFs had a more quiescent cell cycle pattern than did the IFKs and DFs-DPCs. Therefore, the representative cell cycle-related gene expression of IFKs, HFs, and DFs-DPCs was analyzed by NGS. Our study will allow researchers to further investigate the potential interactions and signaling pathways that are active in HF-related diseases and cancer and may aid in future bioengineering applications.
Collapse
Affiliation(s)
- Jaein Lee
- Department of Laboratory Medicine, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Republic of Korea
| | | | | |
Collapse
|
58
|
Nakajima T, Kaneko Y, Kurabayashi M. Unveiling specific triggers and precipitating factors for fatal cardiac events in inherited arrhythmia syndromes. Circ J 2015; 79:1185-92. [PMID: 25925977 DOI: 10.1253/circj.cj-15-0322] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Patients with inherited arrhythmia syndromes, such as long QT syndrome, Brugada syndrome, early repolarization syndrome, catecholaminergic polymorphic ventricular tachycardia, and their latent forms, are at risk for fatal arrhythmias. These diseases are typically associated with genetic mutations that perturb cardiac ionic currents. The analysis of cardiac events by genotype-phenotype correlation studies has revealed that fatal arrhythmias in some genotypes are triggered by physical or emotional stress, and those in the others are more likely to occur during sleep or at rest. Thus, the risk stratification and management of affected patients differ strikingly according to the genetic variant of the inherited arrhythmia syndrome. Risk stratification may be further refined by considering the precipitating factors, such as drugs, bradycardia, electrolyte disturbances, fever, and cardiac memory. Moreover, an increasing number of studies imply that the susceptibility of fatal arrhythmias in patients with acute coronary syndrome or takotsubo cardiomyopathy is at least partly ascribed to the genetic variants causing inherited arrhythmia syndromes. In this article, we review the recent advances in the understanding of the molecular genetics and genotype-phenotype correlations in inherited arrhythmia syndromes and consider the triggers and precipitating factors for fatal arrhythmias in these disorders. Further studies to explore the triggers and precipitating factors specific to the genotypes and diseases are needed for better clinical management.
Collapse
Affiliation(s)
- Tadashi Nakajima
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine
| | | | | |
Collapse
|
59
|
KChIP-like auxiliary subunits of Kv4 channels regulate excitability of muscle cells and control male turning behavior during mating in Caenorhabditis elegans. J Neurosci 2015; 35:1880-91. [PMID: 25653349 DOI: 10.1523/jneurosci.3429-14.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated Kv4 channels control the excitability of neurons and cardiac myocytes by conducting rapidly activating-inactivating currents. The function of Kv4 channels is profoundly modulated by K(+) channel interacting protein (KChIP) soluble auxiliary subunits. However, the in vivo mechanism of the modulation is not fully understood. Here, we identified three C. elegans KChIP-like (ceKChIP) proteins, NCS-4, NCS-5, and NCS-7. All three ceKChIPs alter electrical characteristics of SHL-1, a C. elegans Kv4 channel ortholog, currents by slowing down inactivation kinetics and shifting voltage dependence of activation to more hyperpolarizing potentials. Native SHL-1 current is completely abolished in cultured myocytes of Triple KO worms in which all three ceKChIP genes are deleted. Reexpression of NCS-4 partially restored expression of functional SHL-1 channels, whereas NCS-4(efm), a NCS-4 mutant with impaired Ca(2+)-binding ability, only enhanced expression of SHL-1 proteins, but failed to transport them from the Golgi apparatus to the cell membrane in body wall muscles of Triple KO worms. Moreover, translational reporter revealed that NCS-4 assembles with SHL-1 K(+) channels in male diagonal muscles. Deletion of either ncs-4 or shl-1 significantly impairs male turning, a behavior controlled by diagonal muscles during mating. The phenotype of the ncs-4 null mutant could be rescued by reexpression of NCS-4, but not NCS-4(efm), further emphasizing the importance of Ca(2+) binding to ceKChIPs in regulating native SHL-1 channel function. Together, these data reveal an evolutionarily conserved mechanism underlying the regulation of Kv4 channels by KChIPs and unravel critical roles of ceKChIPs in regulating muscle cell excitability and animal behavior in C. elegans.
Collapse
|
60
|
Williams VS, Cresswell CJ, Ruspi G, Yang T, Atak TC, McLoughlin M, Ingram CD, Ramirez AH, Roden D, Armstrong M. Multiplex ligation-dependent probe amplification copy number variant analysis in patients with acquired long QT syndrome. Europace 2015; 17:635-41. [PMID: 25564553 DOI: 10.1093/europace/euu288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/16/2014] [Indexed: 01/06/2023] Open
Abstract
AIMS Thirteen genetic loci map to families with congenital long QT syndrome (cLQT) and multiple single nucleotide mutations have been functionally implicated in cLQT. Studies have investigated copy number variations (CNVs) in the cLQT genes to ascertain their involvement in cLQT. In these studies 3-12% of cLQT patients who were mutation negative by all other methods carried CNVs in cLQT genes. Prolongation of the QT interval can also be acquired after exposure to certain drugs [acquired LQT (aLQT)]. Single nucleotide mutations in cLQT genes have also been associated with and functionally implicated in aLQT, but to date no studies have explored CNVs as an additional susceptibility factor in aLQT. The aim of this study was to explore the contribution of CNVs in determining susceptibility to aLQT. METHODS AND RESULTS In this study we screened the commonest cLQT genes (KCNQ1; KCNH2; SCN5A; KCNE1, and KCNE2) in a general population of healthy volunteers and in a cohort of subjects presenting with aLQT for CNVs using the multiplex ligation-dependent probe amplification method. Copy number variants were detected and confirmed in 1 of 197 of the healthy volunteers and in 1 of 90 subjects with aLQT. The CNV in the aLQT subject was functionally characterized and demonstrated impaired channel function. CONCLUSION Copy number variation is a possible additional risk factor for aLQT and should be considered for incorporation into pharmacogenetic screening of LQTS genes in addition to mutation detection to improve the safety of medication administration.
Collapse
Affiliation(s)
- Victoria S Williams
- AstraZeneca, Personalised Healthcare and Biomarkers, R&D Genetics, Alderley Park, UK
| | - Carl J Cresswell
- AstraZeneca, Personalised Healthcare and Biomarkers, R&D Genetics, Alderley Park, UK
| | - Gerhard Ruspi
- School of Biomedical Sciences, King's College London, London, UK
| | - Tao Yang
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Thomas C Atak
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Matthew McLoughlin
- AstraZeneca, Personalised Healthcare and Biomarkers, R&D Genetics, Alderley Park, UK
| | | | - Andrea H Ramirez
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Dan Roden
- Department of Medicine, Vanderbilt University, Nashville, TN, USA Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Martin Armstrong
- AstraZeneca, Personalised Healthcare and Biomarkers, R&D Genetics, Alderley Park, UK Shire AG, Business Park Terre-Bonne, Route de Crassier 7, 1262 Eysins, Switzerland
| |
Collapse
|
61
|
Nielsen JB, Bentzen BH, Olesen MS, David JP, Olesen SP, Haunsø S, Svendsen JH, Schmitt N. Gain-of-function mutations in potassium channel subunit KCNE2 associated with early-onset lone atrial fibrillation. Biomark Med 2014; 8:557-70. [PMID: 24796621 DOI: 10.2217/bmm.13.137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is the most common cardiac arrhythmia. Disturbances in cardiac potassium conductance are considered as one of the disease mechanisms in AF. We aimed to investigate if mutations in potassium-channel β-subunits KCNE2 and KCNE3 are associated with early-onset lone AF. METHODS & RESULTS The coding regions of KCNE2 and KCNE3 were bidirectionally sequenced in 192 unrelated patients diagnosed with early-onset lone AF (<40 years). Two nonsynonymous missense mutations were identified in KCNE2 (M23L and I57T). Both mutations were absent in a healthy control group (n=1500 alleles). Electrophysiological investigations were performed for both mutations in combination with candidate pore-forming α-subunits KV7.1, KV11.1, KV4.3 and KV1.5. A significant gain-of-function effect was observed upon coexpression with KV7.1 and KV7.1+KCNE1. Confocal imaging found no differences in subcellular localization. No disease-suspected mutations were identified in KCNE3. CONCLUSION We identified two KCNE2 gain-of-function missense mutations that seem to increase the susceptibility of early-onset lone AF. These results confirm previous findings indicating that gain-of-function in the slow delayed rectifier potassium current might be involved in the pathogenesis of AF.
Collapse
Affiliation(s)
- Jonas Bille Nielsen
- Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Petropoulou E, Jamshidi Y, Behr ER. The genetics of pro-arrhythmic adverse drug reactions. Br J Clin Pharmacol 2014; 77:618-25. [PMID: 23834499 DOI: 10.1111/bcp.12208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/26/2013] [Indexed: 12/19/2022] Open
Abstract
Ventricular arrhythmia induced by drugs (pro-arrythmia) is an uncommon event, whose occurrence is unpredictable but potentially fatal. The ability of a variety of medications to induce these arrhythmias is a significant problem facing the pharmaceutical industry. Genetic variants have been shown to play a role in adverse events and are also known to influence an individual's optimal drug dose. This review provides an overview of the current understanding of the role of genetic variants in modulating the risk of drug induced arrhythmias.
Collapse
Affiliation(s)
- Evmorfia Petropoulou
- Human Genetics Research Centre, Division of Biomedical Sciences, St George's Hospital Medical School, London, SW17 0RE, UK
| | | | | |
Collapse
|
63
|
Clarithromycin-induced QT prolongation in a patient with incidental parathyroidectomy. Cardiovasc Endocrinol 2014. [DOI: 10.1097/xce.0000000000000016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
64
|
KCNE2 modulates cardiac L-type Ca2+ channel. J Mol Cell Cardiol 2014; 72:208-18. [DOI: 10.1016/j.yjmcc.2014.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 03/05/2014] [Accepted: 03/18/2014] [Indexed: 11/19/2022]
|
65
|
Plant LD, Xiong D, Dai H, Goldstein SAN. Individual IKs channels at the surface of mammalian cells contain two KCNE1 accessory subunits. Proc Natl Acad Sci U S A 2014; 111:E1438-46. [PMID: 24591645 PMCID: PMC3986162 DOI: 10.1073/pnas.1323548111] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
KCNE1 (E1) β-subunits assemble with KCNQ1 (Q1) voltage-gated K(+) channel α-subunits to form IKslow (IKs) channels in the heart and ear. The number of E1 subunits in IKs channels has been an issue of ongoing debate. Here, we use single-molecule spectroscopy to demonstrate that surface IKs channels with human subunits contain two E1 and four Q1 subunits. This stoichiometry does not vary. Thus, IKs channels in cells with elevated levels of E1 carry no more than two E1 subunits. Cells with low levels of E1 produce IKs channels with two E1 subunits and Q1 channels with no E1 subunits--channels with one E1 do not appear to form or are restricted from surface expression. The plethora of models of cardiac function, transgenic animals, and drug screens based on variable E1 stoichiometry do not reflect physiology.
Collapse
Affiliation(s)
| | | | - Hui Dai
- Department of Biochemistry, Brandeis University, Waltham, MA, 02453
| | | |
Collapse
|
66
|
Wang D, Shah KR, Um SY, Eng LS, Zhou B, Lin Y, Mitchell AA, Nicaj L, Prinz M, McDonald TV, Sampson BA, Tang Y. Cardiac channelopathy testing in 274 ethnically diverse sudden unexplained deaths. Forensic Sci Int 2014; 237:90-9. [DOI: 10.1016/j.forsciint.2014.01.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/23/2013] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
|
67
|
Marzuillo P, Benettoni A, Germani C, Ferrara G, D'Agata B, Barbi E. Acquired long QT syndrome: a focus for the general pediatrician. Pediatr Emerg Care 2014; 30:257-261. [PMID: 24694881 DOI: 10.1097/pec.0000000000000108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acquired long QT syndrome (LQTS) is a disorder of cardiac repolarization most often due to specific drugs, hypokalemia, or hypomagnesemia that may precipitate torsade de pointes and cause sudden cardiac death. Common presentations of the LQTS are palpitations, presyncope, syncope, cardiac arrest, and seizures. An abnormal 12-lead electrocardiogram obtained while the patient is at rest is the key to diagnosis. The occurrence of drug-induced LQTS is unpredictable in any given individual, but a common observation is that most patients have at least 1 identifiable risk factor in addition to drug exposure. The cornerstone of the management of acquired LQTS includes the identification and discontinuation of any precipitating drug and the correction of metabolic abnormalities, such as hypokalemia or hypomagnesemia. Most of the episodes of torsade de pointes are short-lived and terminate spontaneously. We propose a management protocol that could be useful for the daily practice in the emergency pediatric department to reduce the risk of acquired QT prolongation.
Collapse
Affiliation(s)
- Pierluigi Marzuillo
- From the *Department of Women and Children and General and Specialized Surgery, Seconda Università degli Studi di Napoli, Naples and †Institute for Maternal and Child Health-IRCCS, Burlo Garofolo, University of Trieste, Trieste, Italy
| | | | | | | | | | | |
Collapse
|
68
|
Romero L, Trenor B, Yang PC, Saiz J, Clancy CE. In silico screening of the impact of hERG channel kinetic abnormalities on channel block and susceptibility to acquired long QT syndrome. J Mol Cell Cardiol 2014; 72:126-37. [PMID: 24631769 DOI: 10.1016/j.yjmcc.2014.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 01/08/2023]
Abstract
Accurate diagnosis of predisposition to long QT syndrome is crucial for reducing the risk of cardiac arrhythmias. In recent years, drug-induced provocative tests have proved useful to unmask some latent mutations linked to cardiac arrhythmias. In this study we expanded this concept by developing a prototype for a computational provocative screening test to reveal genetic predisposition to acquired long-QT syndrome (aLQTS). We developed a computational approach to reveal the pharmacological properties of IKr blocking drugs that are most likely to cause aLQTS in the setting of subtle alterations in IKr channel gating that would be expected to result from benign genetic variants. We used the model to predict the most potentially lethal combinations of kinetic anomalies and drug properties. In doing so, we also implicitly predicted ideal inverse therapeutic properties of K channel openers that would be expected to remedy a specific defect. We systematically performed "in silico mutagenesis" by altering discrete kinetic transition rates of the Fink et al. Markov model of human IKr channels, corresponding to activation, inactivation, deactivation and recovery from inactivation of IKr channels. We then screened and identified the properties of IKr blockers that caused acquired long QT and therefore unmasked mutant phenotypes for mild, moderate and severe variants. Mutant IKr channels were incorporated into the O'Hara et al. human ventricular action potential (AP) model and subjected to simulated application of a wide variety of IKr-drug interactions in order to identify the characteristics that selectively exacerbate the AP duration (APD) differences between wild-type and IKr mutated cells. Our results show that drugs with disparate affinities to conformation states of the IKr channel are key to amplify variants underlying susceptibility to acquired long QT syndrome, an effect that is especially pronounced at slow frequencies. Finally, we developed a mathematical formulation of the M54T MiRP1 latent mutation and simulated a provocative test. In this setting, application of dofetilide dramatically amplified the predicted QT interval duration in the M54T hMiRP1 mutation compared to wild-type.
Collapse
Affiliation(s)
- Lucia Romero
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Beatriz Trenor
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Pei-Chi Yang
- Department of Pharmacology, University of California, Davis, Genome Building Rm 3503, Davis, CA 95616-8636, USA.
| | - Javier Saiz
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Genome Building Rm 3503, Davis, CA 95616-8636, USA.
| |
Collapse
|
69
|
Christiansen M, Hedley PL, Theilade J, Stoevring B, Leren TP, Eschen O, Sørensen KM, Tybjærg-Hansen A, Ousager LB, Pedersen LN, Frikke-Schmidt R, Aidt FH, Hansen MG, Hansen J, Bloch Thomsen PE, Toft E, Henriksen FL, Bundgaard H, Jensen HK, Kanters JK. Mutations in Danish patients with long QT syndrome and the identification of a large founder family with p.F29L in KCNH2. BMC MEDICAL GENETICS 2014; 15:31. [PMID: 24606995 PMCID: PMC4007532 DOI: 10.1186/1471-2350-15-31] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/18/2014] [Indexed: 01/08/2023]
Abstract
Background Long QT syndrome (LQTS) is a cardiac ion channelopathy which presents clinically with palpitations, syncope or sudden death. More than 700 LQTS-causing mutations have been identified in 13 genes, all of which encode proteins involved in the execution of the cardiac action potential. The most frequently affected genes, covering > 90% of cases, are KCNQ1, KCNH2 and SCN5A. Methods We describe 64 different mutations in 70 unrelated Danish families using a routine five-gene screen, comprising KCNQ1, KCNH2 and SCN5A as well as KCNE1 and KCNE2. Results Twenty-two mutations were found in KCNQ1, 28 in KCNH2, 9 in SCN5A, 3 in KCNE1 and 2 in KCNE2. Twenty-six of these have only been described in the Danish population and 18 are novel. One double heterozygote (1.4% of families) was found. A founder mutation, p.F29L in KCNH2, was identified in 5 “unrelated” families. Disease association, in 31.2% of cases, was based on the type of mutation identified (nonsense, insertion/deletion, frameshift or splice-site). Functional data was available for 22.7% of the missense mutations. None of the mutations were found in 364 Danish alleles and only three, all functionally characterised, were recorded in the Exome Variation Server, albeit at a frequency of < 1:1000. Conclusion The genetic etiology of LQTS in Denmark is similar to that found in other populations. A large founder family with p.F29L in KCNH2 was identified. In 48.4% of the mutations disease causation was based on mutation type or functional analysis.
Collapse
Affiliation(s)
- Michael Christiansen
- Department of Clinical Biochemistry, Immunology and Genetics, Statens Serum Institut, Ørestads Boulevard 5, 2300S, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Crump SM, Abbott GW. Arrhythmogenic KCNE gene variants: current knowledge and future challenges. Front Genet 2014; 5:3. [PMID: 24478792 PMCID: PMC3900874 DOI: 10.3389/fgene.2014.00003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 01/04/2014] [Indexed: 11/13/2022] Open
Abstract
There are twenty-five known inherited cardiac arrhythmia susceptibility genes, all of which encode either ion channel pore-forming subunits or proteins that regulate aspects of ion channel biology such as function, trafficking, and localization. The human KCNE gene family comprises five potassium channel regulatory subunits, sequence variants in each of which are associated with cardiac arrhythmias. KCNE gene products exhibit promiscuous partnering and in some cases ubiquitous expression, hampering efforts to unequivocally correlate each gene to specific native potassium currents. Likewise, deducing the molecular etiology of cardiac arrhythmias in individuals harboring rare KCNE gene variants, or more common KCNE polymorphisms, can be challenging. In this review we provide an update on putative arrhythmia-causing KCNE gene variants, and discuss current thinking and future challenges in the study of molecular mechanisms of KCNE-associated cardiac rhythm disturbances.
Collapse
Affiliation(s)
- Shawn M Crump
- Bioelectricity Laboratory, Department of Pharmacology, Department of Physiology and Biophysics, School of Medicine, University of California Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Pharmacology, Department of Physiology and Biophysics, School of Medicine, University of California Irvine, CA, USA
| |
Collapse
|
71
|
Abstract
Abnormal functioning of cardiac ion channels can disrupt cardiac myocyte action potentials and thus cause potentially lethal cardiac arrhythmias. Ion channel dysfunction has been observed at all stages in channel ontogeny, from biogenesis to regulation, and arises from genetic or environmental factors, or both. Acquired arrhythmias - including those that are drug induced - are more common than solely inherited arrhythmias but, in some cases, also contain an identifiable genetic component. This interplay between the pharmacology and genetics - known as 'pharmacogenetics' - of cardiac ion channels and the systems that impact them presents both challenges and opportunities to academics, pharmaceutical companies and clinicians seeking to develop and utilize therapies for cardiac rhythm disorders. In this review, we discuss ion channel pharmacogenetics in the context of both causation and treatment of cardiac arrhythmias, focusing on the long QT syndromes.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Weill Medical College of Cornell University, Greenberg Division of Cardiology, Department of Medicine and Department of Pharmacology, 520 East 70th Street, New York, NY 10021, USA.
| | | |
Collapse
|
72
|
Kline CF, Mohler PJ. Weighing in on molecular anchors: the role of ankyrin polypeptides in human arrhythmia. Expert Rev Cardiovasc Ther 2014; 4:477-85. [PMID: 16918266 DOI: 10.1586/14779072.4.4.477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Loss-of-function gene variants which affect the biophysical properties of ion channel proteins have long been associated with the destabilization of cardiac electrical activity, leading to human arrhythmia and sudden cardiac death. However, recent studies have also demonstrated the importance of ion channel/transporter-anchoring molecules for normal cardiac function. Ankyrins are a family of membrane adaptor proteins whose role in metazoan physiology has been elucidated over the last quarter of a century, but with great strides taken in the last half decade with regard to cardiac cell physiology. The association of dysfunction in ankyrin-based cellular pathways with abnormal human cardiac function represents a surprising turn in the genetics of arrhythmias and sudden cardiac death, demonstrating an exciting new player in the field of 'channelopathies'.
Collapse
Affiliation(s)
- Crystal F Kline
- Vanderbilt University School of Medicine, Graduate Program in Pathology, Nashville, TN 37232, USA.
| | | |
Collapse
|
73
|
Du C, El Harchi A, Zhang H, Hancox JC. Modification by KCNE1 variants of the hERG potassium channel response to premature stimulation and to pharmacological inhibition. Physiol Rep 2013; 1:e00175. [PMID: 24400172 PMCID: PMC3871485 DOI: 10.1002/phy2.175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/02/2013] [Accepted: 11/04/2013] [Indexed: 01/08/2023] Open
Abstract
human Ether-à-go-go-Related Gene (hERG) encodes the pore-forming subunit of cardiac rapid delayed rectifier K(+) current (I Kr) channels, which play important roles in ventricular repolarization, in protecting the myocardium from unwanted premature stimuli, and in drug-induced Long QT Syndrome (LQTS). KCNE1, a small transmembrane protein, can coassemble with hERG. However, it is not known how KCNE1 variants influence the channel's response to premature stimuli or if they influence the sensitivity of hERG to pharmacological inhibition. Accordingly, whole-cell patch-clamp measurements of hERG current (I hERG) were made at 37°C from hERG channels coexpressed with either wild-type (WT) KCNE1 or with one of three KCNE1 variants (A8V, D76N, and D85N). Under both conventional voltage clamp and ventricular action potential (AP) clamp, the amplitude of I hERG was smaller for A8V, D76N, and D85N KCNE1 + hERG than for WT KCNE1 + hERG. Using paired AP commands, with the second AP waveform applied at varying time intervals following the first to mimic premature ventricular excitation, the response of I hERG carried by each KCNE1 variant was reduced compared to that with WT KCNE1 + hERG. The I hERG blocking potency of the antiarrhythmic drug quinidine was similar between WT KCNE1 and the three KCNE1 variants. However, the I hERG inhibitory potency of the antibiotic clarithromycin and of the prokinetic drug cisapride was altered by KCNE1 variants. These results demonstrate that naturally occurring KCNE1 variants can reduce the response of hERG channels to premature excitation and also alter the sensitivity of hERG channels to inhibition by some drugs linked to acquired LQTS.
Collapse
Affiliation(s)
- Chunyun Du
- School of Physiology and Pharmacology and Cardiovascular Research Laboratories, Medical Sciences Building, University of Bristol Bristol, BS8 1TD, U.K
| | - Aziza El Harchi
- School of Physiology and Pharmacology and Cardiovascular Research Laboratories, Medical Sciences Building, University of Bristol Bristol, BS8 1TD, U.K
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, University of Manchester Manchester, M13 9PL, U.K
| | - Jules C Hancox
- School of Physiology and Pharmacology and Cardiovascular Research Laboratories, Medical Sciences Building, University of Bristol Bristol, BS8 1TD, U.K
| |
Collapse
|
74
|
Bhuiyan ZA, Al-Shahrani S, Al-Aama J, Wilde AAM, Momenah TS. Congenital Long QT Syndrome: An Update and Present Perspective in Saudi Arabia. Front Pediatr 2013; 1:39. [PMID: 24400285 PMCID: PMC3864249 DOI: 10.3389/fped.2013.00039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 11/05/2013] [Indexed: 12/14/2022] Open
Abstract
Primary cardiac arrhythmias are often caused by defects, predominantly in the genes responsible for generation of cardiac electrical potential, i.e., cardiac rhythm generation. Due to the variability in underlying genetic defects, type, and location of the mutations and putative modifiers, clinical phenotypes could be moderate to severe, even absent in many individuals. Clinical presentation and severity could be quite variable, syncope, or sudden cardiac death could also be the first and the only manifestation in a patient who had previously no symptoms at all. Despite usual familial occurrence of such cardiac arrhythmias, disease causal genetic defects could also be de novo in significant number of patients. Long QT syndrome (LQTS) is the most eloquently investigated primary cardiac rhythm disorder. A genetic defect can be identified in ∼70% of definitive LQTS patients, followed by Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) and Brugada syndrome (BrS), where a genetic defect is found in <40% cases. In addition to these widely investigated hereditary arrhythmia syndromes, there remain many other relatively less common arrhythmia syndromes, where researchers also have unraveled the genetic etiology, e.g., short QT syndrome (SQTS), sick sinus syndrome (SSS), cardiac conduction defect (CCD), idiopathic ventricular fibrillation (IVF), early repolarization syndrome (ERS). There exist also various other ill-defined primary cardiac rhythm disorders with strong genetic and familial predisposition. In the present review we will focus on the genetic basis of LQTS and its clinical management. We will also discuss the presently available genetic insight in this context from Saudi Arabia.
Collapse
Affiliation(s)
- Zahurul A. Bhuiyan
- Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Safar Al-Shahrani
- Department of Pediatrics, Faculty of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Jumana Al-Aama
- Princess Al Jawhara Albrahim Center of Excellence in Research of Hereditary Disorders, Jeddah, Saudi Arabia
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arthur A. M. Wilde
- Princess Al Jawhara Albrahim Center of Excellence in Research of Hereditary Disorders, Jeddah, Saudi Arabia
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tarek S. Momenah
- Department of Pediatric Cardiology, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
75
|
Trolle C, Mortensen KH, Pedersen LN, Berglund A, Jensen HK, Andersen NH, Gravholt CH. Long QT interval in Turner syndrome--a high prevalence of LQTS gene mutations. PLoS One 2013; 8:e69614. [PMID: 23936059 PMCID: PMC3723856 DOI: 10.1371/journal.pone.0069614] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/07/2013] [Indexed: 01/15/2023] Open
Abstract
Objectives QT-interval prolongation of unknown aetiology is common in Turner syndrome. This study set out to explore the presence of known long QT mutations in Turner syndrome and to examine the corrected QT-interval (QTc) over time and relate the findings to the Turner syndrome phenotype. Methods Adult women with Turner syndrome (n = 88) were examined thrice and 68 age-matched healthy controls were examined once. QTc was measured by one blinded reader (intra-reader variability: 0.7%), and adjusted for influence of heart rate by Bazett’s (bQTc) and Hodges’s formula (hQTc). The prevalence of mutations in genes related to Long QT syndrome was determined in women with Turner syndrome and a QTc >432.0 milliseconds (ms). Echocardiographic assessment of aortic valve morphology, 24-hour blood pressures and blood samples were done. Results The mean hQTc in women with Turner syndrome (414.0±25.5 ms) compared to controls (390.4±17.8 ms) was prolonged (p<0.001) and did not change over time (416.9±22.6 vs. 415.6±25.5 ms; p = 0.4). 45,X karyotype was associated with increased hQTc prolongation compared to other Turner syndrome karyotypes (418.2±24.8 vs. 407.6±25.5 ms; p = 0.055). In women with Turner syndrome and a bQTc >432 ms, 7 had mutations in major Long QT syndrome genes (SCN5A and KCNH2) and one in a minor Long QT syndrome gene (KCNE2). Conclusion There is a high prevalence of mutations in the major LQTS genes in women with TS and prolonged QTc. It remains to be settled, whether these findings are related to the unexplained excess mortality in Turner women. Clinical Trial Registration NCT00624949. https://register.clinicaltrials.gov/prs/app/action/SelectProtocol/sid/S0001FLI/selectaction/View/ts/3/uid/U000099E.
Collapse
Affiliation(s)
- Christian Trolle
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian H. Mortensen
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark
- Department of Radiology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Lisbeth N. Pedersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Agnethe Berglund
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik K. Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels H. Andersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Claus H. Gravholt
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
76
|
Sroubek J, Krishnan Y, McDonald TV. Sequence and structure-specific elements of HERG mRNA determine channel synthesis and trafficking efficiency. FASEB J 2013; 27:3039-53. [PMID: 23608144 DOI: 10.1096/fj.12-227009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human ether-á-gogo-related gene (HERG) encodes a potassium channel that is highly susceptible to deleterious mutations resulting in susceptibility to fatal cardiac arrhythmias. Most mutations adversely affect HERG channel assembly and trafficking. Why the channel is so vulnerable to missense mutations is not well understood. Since nothing is known of how mRNA structural elements factor in channel processing, we synthesized a codon-modified HERG cDNA (HERG-CM) where the codons were synonymously changed to reduce GC content, secondary structure, and rare codon usage. HERG-CM produced typical IKr-like currents; however, channel synthesis and processing were markedly different. Translation efficiency was reduced for HERG-CM, as determined by heterologous expression, in vitro translation, and polysomal profiling. Trafficking efficiency to the cell surface was greatly enhanced, as assayed by immunofluorescence, subcellular fractionation, and surface labeling. Chimeras of HERG-NT/CM indicated that trafficking efficiency was largely dependent on 5' sequences, while translation efficiency involved multiple areas. These results suggest that HERG translation and trafficking rates are independently governed by noncoding information in various regions of the mRNA molecule. Noncoding information embedded within the mRNA may play a role in the pathogenesis of hereditary arrhythmia syndromes and could provide an avenue for targeted therapeutics.
Collapse
Affiliation(s)
- Jakub Sroubek
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | |
Collapse
|
77
|
Mahida S, Hogarth AJ, Cowan C, Tayebjee MH, Graham LN, Pepper CB. Genetics of congenital and drug-induced long QT syndromes: current evidence and future research perspectives. J Interv Card Electrophysiol 2013; 37:9-19. [PMID: 23515882 DOI: 10.1007/s10840-013-9779-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/07/2013] [Indexed: 12/17/2022]
Abstract
The long QT syndrome (LQTS) is a condition characterized by abnormal prolongation of the QT interval with an associated risk of ventricular arrhythmias and sudden cardiac death. Congenital forms of LQTS arise due to rare and highly penetrant mutations that segregate in a Mendelian fashion. Over the years, multiple mutations in genes encoding ion channels and ion channel binding proteins have been reported to underlie congenital LQTS. Drugs are by far the most common cause of acquired forms of LQTS. Emerging evidence suggests that drug-induced LQTS also has a significant heritable component. However, the genetic substrate underlying drug-induced LQTS is presently largely unknown. In recent years, advances in next-generation sequencing technology and molecular biology techniques have significantly enhanced our ability to identify genetic variants underlying both monogenic diseases and more complex traits. In this review, we discuss the genetic basis of congenital and drug-induced LQTS and focus on future avenues of research in the field. Ultimately, a detailed characterization of the genetic substrate underlying congenital and drug-induced LQTS will enhance risk stratification and potentially result in the development of tailored genotype-based therapies.
Collapse
Affiliation(s)
- Saagar Mahida
- Leeds General Infirmary, Great George Street, Leeds, LS1 3EX, UK.
| | | | | | | | | | | |
Collapse
|
78
|
Swiatkowski P, Sesti F. Delayed pharyngeal repolarization promotes abnormal calcium buildup in aging muscle. Biochem Biophys Res Commun 2013; 433:354-7. [PMID: 23510998 DOI: 10.1016/j.bbrc.2013.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 03/08/2013] [Indexed: 11/28/2022]
Abstract
In the pharynx of Caenorhabditis elegans, the accessory subunit MPS-4, homolog to human KCNE1, forms a complex with K(+) channel EXP-2 that terminates the action potential. An aspartate residue critical for KCNE1 function, asp76, is conserved in MPS-4 (asp74). Here, we studied the effects of D74N-MPS-4 on the aging pharynx. Electrophysiological studies showed that D74N delays pharyngeal repolarization. Pharynxes of transgenic worms expressing D74N exhibited higher levels of intracellular calcium compared to normal pharynxes. Accordingly, loss of pharyngeal function was accelerated in aging D74N worms. The pharyngeal action potential resembles the action potential that controls the mechanical activity of human left ventricle. Hence, these findings argue that the hearts of patients affected by delayed repolarization, a condition known as long QT syndrome, may experience dysregulated calcium homeostasis.
Collapse
Affiliation(s)
- Przemyslaw Swiatkowski
- University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, Piscataway, NJ 08854, USA
| | | |
Collapse
|
79
|
Köhncke C, Lisewski U, Schleußner L, Gaertner C, Reichert S, Roepke TK. Isolation and Kv channel recordings in murine atrial and ventricular cardiomyocytes. J Vis Exp 2013:e50145. [PMID: 23524949 DOI: 10.3791/50145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
KCNE genes encode for a small family of Kv channel ancillary subunits that form heteromeric complexes with Kv channel alpha subunits to modify their functional properties. Mutations in KCNE genes have been found in patients with cardiac arrhythmias such as the long QT syndrome and/or atrial fibrillation. However, the precise molecular pathophysiology that leads to these diseases remains elusive. In previous studies the electrophysiological properties of the disease causing mutations in these genes have mostly been studied in heterologous expression systems and we cannot be sure if the reported effects can directly be translated into native cardiomyocytes. In our laboratory we therefore use a different approach. We directly study the effects of KCNE gene deletion in isolated cardiomyocytes from knockout mice by cellular electrophysiology - a unique technique that we describe in this issue of the Journal of Visualized Experiments. The hearts from genetically engineered KCNE mice are rapidly excised and mounted onto a Langendorff apparatus by aortic cannulation. Free Ca(2+) in the myocardium is bound by EGTA, and dissociation of cardiac myocytes is then achieved by retrograde perfusion of the coronary arteries with a specialized low Ca(2+) buffer containing collagenase. Atria, free right ventricular wall and the left ventricle can then be separated by microsurgical techniques. Calcium is then slowly added back to isolated cardiomyocytes in a multiple step comprising washing procedure. Atrial and ventricular cardiomyocytes of healthy appearance with no spontaneous contractions are then immediately subjected to electrophysiological analyses by patch clamp technique or other biochemical analyses within the first 6 hours following isolation.
Collapse
Affiliation(s)
- Clemens Köhncke
- Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbrück Center for Molecular Medicine
| | | | | | | | | | | |
Collapse
|
80
|
Abstract
The abrupt cessation of effective cardiac function due to an aberrant heart rhythm can cause sudden and unexpected death at any age, a syndrome called sudden cardiac death (SCD). Annually, more than 300,000 cases of SCD occur in the United States alone, making this a major public health concern. Our current understanding of the mechanisms responsible for SCD has emerged from decades of basic science investigation into the normal electrophysiology of the heart, the molecular physiology of cardiac ion channels, fundamental cellular and tissue events associated with cardiac arrhythmias, and the molecular genetics of monogenic disorders of heart rhythm. This knowledge has helped shape the current diagnosis and treatment of inherited arrhythmia susceptibility syndromes associated with SCD and has provided a pathophysiological framework for understanding more complex conditions predisposing to this tragic event. This Review presents an overview of the molecular basis of SCD, with a focus on monogenic arrhythmia syndromes.
Collapse
Affiliation(s)
- Alfred L George
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA.
| |
Collapse
|
81
|
Abbott GW. KCNE genetics and pharmacogenomics in cardiac arrhythmias: much ado about nothing? Expert Rev Clin Pharmacol 2013; 6:49-60. [PMID: 23272793 PMCID: PMC4917007 DOI: 10.1586/ecp.12.76] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Voltage-gated ion channels respond to changes in membrane potential with conformational shifts that either facilitate or stem the movement of charged ions across the cell membrane. This controlled movement of ions is particularly important for the action potentials of excitable cells such as cardiac myocytes and therefore essential for timely beating of the heart. Inherited mutations in ion channel genes and in the genes encoding proteins that regulate them can cause lethal cardiac arrhythmias either by direct channel disruption or by altering interactions with therapeutic drugs, the best-understood example of both these scenarios being long QT syndrome (LQTS). Unsurprisingly, mutations in the genes encoding ion channel pore-forming α subunits underlie the large majority (~90%) of identified cases of inherited LQTS. Given that inherited LQTS is comparatively rare in itself (~0.04% of the US population), is pursuing study of the remaining known and unknown LQTS-associated genes subject to the law of diminishing returns? Here, with a particular focus on the KCNE family of single transmembrane domain K(+) channel ancillary subunits, the significance to cardiac pharmacogenetics of ion channel regulatory subunits is discussed.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Department of Pharmacology, Department of Physiology & Biophysics, University of California, Irvine, CA, USA.
| |
Collapse
|
82
|
Roden DM. Cardiovascular pharmacogenomics: the future of cardiovascular therapeutics? Can J Cardiol 2013; 29:58-66. [PMID: 23200096 PMCID: PMC3529768 DOI: 10.1016/j.cjca.2012.07.845] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/17/2012] [Accepted: 07/31/2012] [Indexed: 01/08/2023] Open
Abstract
Responses to drug therapy vary from benefit to no effect to adverse effects which can be serious or occasionally fatal. Increasing evidence supports the idea that genetic variants can play a major role in this spectrum of responses. Well-studied examples in cardiovascular therapeutics include predictors of steady-state warfarin dosage, predictors of reduced efficacy among patients receiving clopidogrel for drug eluting stents, and predictors of some serious adverse drug effects. This review summarizes contemporary approaches to identifying and validating genetic predictors of variability in response to drug treatment. Approaches to incorporating this new knowledge into clinical care, and the barriers to this concept, are addressed.
Collapse
Affiliation(s)
- Dan M Roden
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
83
|
Pharmacogenomics. Mol Pharmacol 2012. [DOI: 10.1002/9781118451908.ch7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
84
|
Abstract
Drug-induced Torsades de Pointes is a rare, unpredictable, and life-threatening serious adverse event. It can be caused by both cardiac and non-cardiac drugs and has become a major issue in novel drug development and for the regulatory authorities. This review describes the problem, predisposing factors, and the underlying genetic predisposition as it is understood currently. The future potential for pharmacogenomic-guided and personalized prescription to prevent drug-induced Torsades de Pointes is discussed. Database searches utilized reports from www.qtdrugs.org up to January 2012, case reports and articles from www.pubmed.com up to January 2012, and the British National Formulary edition at www.bnf.org.
Collapse
Affiliation(s)
- Elijah R Behr
- Cardiovascular Sciences Research Centre, St George's University of London, London SW17 0RE, UK.
| | | |
Collapse
|
85
|
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
|
86
|
Abstract
The Long QT syndrome (LQTS) is a rare disorder in which patients are prone to life threatening ventricular arrhythmia and is a leading cause of sudden death in childhood. Asthma is common and its management in those with LQTS presents a number of potential difficulties. The mainstay of therapy in LQTS is beta-blockade, which may worsen symptoms of asthma. Conversely, beta-agonist therapy is the mainstay of asthma management; which, in those with LQTS, may provoke ventricular arrhythmias. We review available data regarding the management of coexistent LQTS and asthma, and provide a summary of the necessary considerations in managing these patients.
Collapse
Affiliation(s)
- Samuel Collins
- Department Respiratory Paediatrics, Royal Children's Hospital, 50 Flemington Road, Melbourne, Victoria, 3052, Australia.
| | | | | | | |
Collapse
|
87
|
Zaklyazminskaya EV, Abriel H. Prevalence of Significant Genetic Variants in Congenital Long QT Syndrome is Largely Underestimated. Front Pharmacol 2012; 3:72. [PMID: 22557970 PMCID: PMC3338122 DOI: 10.3389/fphar.2012.00072] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 04/06/2012] [Indexed: 11/17/2022] Open
|
88
|
Abstract
KCNE2, originally designated MinK-related peptide 1 (MiRP1), belongs to a five-strong family of potassium channel ancillary (β) subunits that, despite the diminutive size of the family and its members, has loomed large in the field of ion channel physiology. KCNE2 dictates K (+) channel gating, conductance, α subunit composition, trafficking and pharmacology, and also modifies functional properties of monovalent cation-nonselective HCN channels. The Kcne2 (-/-) mouse exhibits cardiac arrhythmia and hypertrophy, achlorhydria, gastric neoplasia, hypothyroidism, alopecia, stunted growth and choroid plexus epithelial dysfunction, illustrating the breadth and depth of the influence of KCNE2, mutations which are also associated with human cardiac arrhythmias. Here, the modus operandi and physiological roles of this potent regulator of membrane excitability and ion secretion are reviewed with particular emphasis on the ability of KCNE2 to shape the electrophysiological landscape of both excitable and non-excitable cells.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Departments of Pharmacology and Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
89
|
The voltage-gated channel accessory protein KCNE2: multiple ion channel partners, multiple ways to long QT syndrome. Expert Rev Mol Med 2011; 13:e38. [DOI: 10.1017/s1462399411002092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The single-pass transmembrane protein KCNE2 or MIRP1 was once thought to be the missing accessory protein that combined with hERG to fully recapitulate the cardiac repolarising current IKr. As a result of this role, it was an easy next step to associate mutations in KCNE2 to long QT syndrome, in which there is delayed repolarisation of the heart. Since that time however, KCNE2 has been shown to modify the behaviour of several other channels and currents, and its role in the heart and in the aetiology of long QT syndrome has become less clear. In this article, we review the known interactions of the KCNE2 protein and the resulting functional effects, and the effects of mutations in KCNE2 and their clinical role.
Collapse
|
90
|
Milan DJ, MacRae CA. Cardiotoxicity Studies in Zebrafish. Zebrafish 2011. [DOI: 10.1002/9781118102138.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
91
|
Varró A, Baczkó I. Cardiac ventricular repolarization reserve: a principle for understanding drug-related proarrhythmic risk. Br J Pharmacol 2011; 164:14-36. [PMID: 21545574 PMCID: PMC3171857 DOI: 10.1111/j.1476-5381.2011.01367.x] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/22/2011] [Accepted: 03/02/2011] [Indexed: 12/11/2022] Open
Abstract
Cardiac repolarization abnormalities can be caused by a wide range of cardiac and non-cardiac compounds and may lead to the development of life-threatening Torsades de Pointes (TdP) ventricular arrhythmias. Drug-induced torsades de pointes is associated with unexpected and unexplained sudden cardiac deaths resulting in the withdrawal of several compounds in the past. To better understand the mechanism of such unexpected sudden cardiac deaths, the concept of repolarization reserve has recently emerged. According to this concept, pharmacological, congenital or acquired impairment of one type of transmembrane ion channel does not necessarily result in excessive repolarization changes because other repolarizing currents can take over and compensate. In this review, the major factors contributing to repolarization reserve are discussed in the context of their clinical significance in physiological and pathophysiological conditions including drug administration, genetic defects, heart failure, diabetes mellitus, gender, renal failure, hypokalaemia, hypothyroidism and athletes' sudden deaths. In addition, pharmacological support of repolarization reserve as a possible therapeutic option is discussed. Some methods for the quantitative estimation of repolarization reserve are also recommended. It is concluded that repolarization reserve should be considered by safety pharmacologists to better understand, predict and prevent previously unexplained drug-induced sudden cardiac deaths.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary.
| | | |
Collapse
|
92
|
Roepke TK, Kanda VA, Purtell K, King EC, Lerner DJ, Abbott GW. KCNE2 forms potassium channels with KCNA3 and KCNQ1 in the choroid plexus epithelium. FASEB J 2011; 25:4264-73. [PMID: 21859894 DOI: 10.1096/fj.11-187609] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cerebrospinal fluid (CSF) is crucial for normal function and mechanical protection of the CNS. The choroid plexus epithelium (CPe) is primarily responsible for secreting CSF and regulating its composition by mechanisms currently not fully understood. Previously, the heteromeric KCNQ1-KCNE2 K(+) channel was functionally linked to epithelial processes including gastric acid secretion and thyroid hormone biosynthesis. Here, using Kcne2(-/-) tissue as a negative control, we found cerebral expression of KCNE2 to be markedly enriched in the CPe apical membrane, where we also discovered expression of KCNQ1. Targeted Kcne2 gene deletion in C57B6 mice increased CPe outward K(+) current 2-fold. The Kcne2 deletion-enhanced portion of the current was inhibited by XE991 (10 μM) and margatoxin (10 μM) but not by dendrotoxin (100 nM), indicating that it arose from augmentation of KCNQ subfamily and KCNA3 but not KCNA1 K(+) channel activity. Kcne2 deletion in C57B6 mice also altered the polarity of CPe KCNQ1 and KCNA3 trafficking, hyperpolarized the CPe membrane by 9 ± 2 mV, and increased CSF [Cl(-)] by 14% compared with wild-type mice. These findings constitute the first report of CPe dysfunction caused by cation channel gene disruption and suggest that KCNE2 influences blood-CSF anion flux by regulating KCNQ1 and KCNA3 in the CPe.
Collapse
Affiliation(s)
- Torsten K Roepke
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Ave., New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
93
|
Bas T, Gao GY, Lvov A, Chandrasekhar KD, Gilmore R, Kobertz WR. Post-translational N-glycosylation of type I transmembrane KCNE1 peptides: implications for membrane protein biogenesis and disease. J Biol Chem 2011; 286:28150-9. [PMID: 21676880 PMCID: PMC3151060 DOI: 10.1074/jbc.m111.235168] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 05/16/2011] [Indexed: 12/21/2022] Open
Abstract
N-Glycosylation of membrane proteins is critical for their proper folding, co-assembly and subsequent matriculation through the secretory pathway. Here, we examine the kinetics of N-glycan addition to type I transmembrane KCNE1 K(+) channel β-subunits, where point mutations that prevent N-glycosylation at one consensus site give rise to disorders of the cardiac rhythm and congenital deafness. We show that KCNE1 has two distinct N-glycosylation sites: a typical co-translational site and a consensus site ∼20 residues away that unexpectedly acquires N-glycans after protein synthesis (post-translational). Mutations that ablate the co-translational site concomitantly reduce glycosylation at the post-translational site, resulting in unglycosylated KCNE1 subunits that cannot reach the cell surface with their cognate K(+) channel. This long range inhibition is highly specific for post-translational N-glycosylation because mutagenic conversion of the KCNE1 post-translational site into a co-translational site restored both monoglycosylation and anterograde trafficking. These results directly explain how a single point mutation can prevent N-glycan attachment at multiple sites, providing a new biogenic mechanism for human disease.
Collapse
Affiliation(s)
- Tuba Bas
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| | - Grace Y. Gao
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| | - Anatoli Lvov
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| | - Kshama D. Chandrasekhar
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| | - Reid Gilmore
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| | - William R. Kobertz
- From the Department of Biochemistry and Molecular Pharmacology and Programs in Neuroscience and Chemical Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324
| |
Collapse
|
94
|
Lewis AS, Estep CM, Chetkovich DM. The fast and slow ups and downs of HCN channel regulation. Channels (Austin) 2011; 4:215-31. [PMID: 20305382 DOI: 10.4161/chan.4.3.11630] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (h channels) form the molecular basis for the hyperpolarization-activated current, I(h), and modulation of h channels contributes to changes in cellular properties critical for normal functions in the mammalian brain and heart. Numerous mechanisms underlie h channel modulation during both physiological and pathological conditions, leading to distinct changes in gating, kinetics, surface expression, channel conductance or subunit composition of h channels. Here we provide a focused review examining mechanisms of h channel regulation, with an emphasis on recent findings regarding interacting proteins such as TRIP8b. This review is intended to serve as a comprehensive resource for physiologists to provide potential molecular mechanisms underlying functionally important changes in I(h) in different biological models, as well as for molecular biologists to delineate the predicted h channel changes associated with complex regulatory mechanisms in both normal function and in disease states.
Collapse
Affiliation(s)
- Alan S Lewis
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | |
Collapse
|
95
|
Song MK, Bae EJ, Baek JS, Kwon BS, Kim GB, Noh CI, Choi JY, Park SS. QT Prolongation and Life Threatening Ventricular Tachycardia in a Patient Injected With Intravenous Meperidine (Demerol®). Korean Circ J 2011; 41:342-5. [PMID: 21779290 PMCID: PMC3132699 DOI: 10.4070/kcj.2011.41.6.342] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/01/2010] [Accepted: 10/12/2010] [Indexed: 12/24/2022] Open
Abstract
QT prolongation is a serious adverse drug effect, which is associated with an increased risk of Torsade de pointes and sudden death. Many drugs, including both cardiac and non-cardiac drugs, have been reported to cause prolongation of QT interval. Although meperidine has not been considered proarrhythmic, we present a unique case of a 16-year-old boy without an underlying cardiac disease, who developed polymorphic ventricular tachycardia, ventricular fibrillation and QT prolongation after an intravenous meperidine injection. He had no mutation in long QT syndrome genes (KCNQ1, KCNH2, and SCN5A), but single nucleotide polymorphisms were reported, including H558R in SCNA5A and K897T in KCNH2.
Collapse
Affiliation(s)
- Mi Kyoung Song
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Shimizu W, Horie M. Phenotypic Manifestations of Mutations in Genes Encoding Subunits of Cardiac Potassium Channels. Circ Res 2011; 109:97-109. [DOI: 10.1161/circresaha.110.224600] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Since 1995, when a potassium channel gene,
hERG
(human ether-à-go-go-related gene), now referred to as
KCNH2
, encoding the rapid component of cardiac delayed rectifier potassium channels was identified as being responsible for type 2 congenital long-QT syndrome, a number of potassium channel genes have been shown to cause different types of inherited cardiac arrhythmia syndromes. These include congenital long-QT syndrome, short-QT syndrome, Brugada syndrome, early repolarization syndrome, and familial atrial fibrillation. Genotype-phenotype correlations have been investigated in some inherited arrhythmia syndromes, and as a result, gene-specific risk stratification and gene-specific therapy and management have become available, particularly for patients with congenital long-QT syndrome. In this review article, the molecular structure and function of potassium channels, the clinical phenotype due to potassium channel gene mutations, including genotype-phenotype correlations, and the diverse mechanisms underlying the potassium channel gene–related diseases will be discussed.
Collapse
Affiliation(s)
- Wataru Shimizu
- From the Division of Arrhythmia and Electrophysiology, Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center (W.S.), Suita, Japan, and the Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science (M.H.), Otsu, Japan
| | - Minoru Horie
- From the Division of Arrhythmia and Electrophysiology, Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center (W.S.), Suita, Japan, and the Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science (M.H.), Otsu, Japan
| |
Collapse
|
97
|
Abstract
Individual variability in drug efficacy and drug safety is a major challenge in current clinical practice, drug development, and drug regulation. For more than 5 decades, studies of pharmacogenetics have provided ample examples of causal relations between genotypes and drug response to account for phenotypic variations of clinical importance in drug therapy. The convergence of pharmacogenetics and human genomics in recent years has dramatically accelerated the discovery of new genetic variations that potentially underlie variability in drug response, giving birth to pharmacogenomics. In addition to the rapid accumulation of knowledge on genome-disease and genome-drug interactions, there arises the hope of individualized medicine. Here we review recent progress in the understanding of genetic contributions to major individual variability in drug therapy with focus on genetic variations of drug target, drug metabolism, drug transport, disease susceptibility, and drug safety. Challenges to future pharmacogenomics and its translation into individualized medicine, drug development, and regulation are discussed. For example, knowledge on genetic determinants of disease pathogenesis and drug action, especially those of complex disease and drug response, is not always available. Relating the many gene variations from genomic sequencing to clinical phenotypes may not be straightforward. It is often very challenging to conduct large scale, prospective studies to establish causal associations between genetic variations and drug response or to evaluate the utility and cost-effectiveness of genomic medicine. Overcoming the obstacles holds promise for achieving the ultimate goal of effective and safe medication to targeted patients with appropriate genotypes.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, 1095 Willowdale Rd., Morgantown, WV 26505, USA.
| | | |
Collapse
|
98
|
Van Horn WD, Vanoye CG, Sanders CR. Working model for the structural basis for KCNE1 modulation of the KCNQ1 potassium channel. Curr Opin Struct Biol 2011; 21:283-91. [PMID: 21296569 PMCID: PMC3070781 DOI: 10.1016/j.sbi.2011.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/03/2011] [Accepted: 01/04/2011] [Indexed: 12/19/2022]
Abstract
The voltage-gated potassium channel KCNQ1 (Kv7.1) is modulated by KCNE1 (minK) to generate the I(Ks) current crucial to heartbeat. Defects in either protein result in serious cardiac arrhythmias. Recently developed structural models of the open and closed state KCNQ1/KCNE1 complexes offer a compelling explanation for how KCNE1 slows channel opening and provides a platform from which to refine and test hypotheses for other aspects of KCNE1 modulation. These working models were developed using an integrative approach based on results from nuclear magnetic resonance spectroscopy, electrophysiology, biochemistry, and computational methods-an approach that can be applied iteratively for model testing and revision. We present a critical review of these structural models, illustrating the strengths and challenges of the integrative approach.
Collapse
Affiliation(s)
- Wade D. Van Horn
- Department of Biochemistry Vanderbilt University School of Medicine, Nashville Tennessee, U.S.A. 37232-8725
| | - Carlos G. Vanoye
- Department of Medicine Vanderbilt University School of Medicine, Nashville Tennessee, U.S.A. 37232-8725
| | - Charles R. Sanders
- Department of Biochemistry Vanderbilt University School of Medicine, Nashville Tennessee, U.S.A. 37232-8725
| |
Collapse
|
99
|
Abstract
The drug-induced long QT syndrome is a distinct clinical entity that has evolved from an electrophysiologic curiosity to a centerpiece in drug regulation and development. This evolution reflects an increasing recognition that a rare adverse drug effect can profoundly upset the balance between benefit and risk that goes into the prescription of a drug by an individual practitioner as well as the approval of a new drug entity by a regulatory agency. This review will outline how defining the central mechanism, block of the cardiac delayed-rectifier potassium current I(Kr), has contributed to defining risk in patients and in populations. Models for studying risk, and understanding the way in which clinical risk factors modulate cardiac repolarization at the molecular level are discussed. Finally, the role of genetic variants in modulating risk is described.
Collapse
Affiliation(s)
- Prince Kannankeril
- Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, Nashville, TN 37232-0575, USA
| | | | | |
Collapse
|
100
|
Place et rôle de la pharmacologie de sécurité dans le développement des médicaments. ANNALES PHARMACEUTIQUES FRANÇAISES 2010; 68:291-300. [DOI: 10.1016/j.pharma.2010.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 05/24/2010] [Accepted: 05/25/2010] [Indexed: 11/19/2022]
|