1
|
Óvári J, Kovács ZM, Dienes C, Magyar J, Bányász T, Nánási PP, Horváth B, Domingos GJ, Feher A, Varga Z, Szentandrássy N. Delavirdine modifies action potential configuration via inhibition of I Kr and I to in canine ventricular myocytes. Biomed Pharmacother 2025; 186:117994. [PMID: 40120556 DOI: 10.1016/j.biopha.2025.117994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Delavirdine is an anti-HIV agent with structural similarity (presence of methanesulfonamide group) to well-known rapid delayed rectifier potassium current (IKr) inhibitors like dofetilide and E-4031. In spite of this and the fact, that IKr blockade can lead to long QT syndrome with a risk of early afterdepolarization, cardiac arrhythmia and even sudden cardiac death development, there is no information on the cardiac electrophysiological effects of the delavirdine. Therefore, we examined the concentration-dependent acute effects of delavirdine on action potential morphology and on the underlying ion currents in enzymatically dispersed canine ventricular cardiomyocytes, as well as in a hERG-channel- and NaV1.5-expressing cell lines. Delavirdine application in the µM concentration range resulted in rapid, potent and reversible blockade of expressed hERG channels. NaV1.5 channels were slightly reduced by delavirdine. Moreover, APD increase, reduction of maximal rates of phases 1 and 3 and a small increase of Plateau20 amplitude were detected in canine left ventricular isolated cardiomyocytes in a concentration-dependent manner. Accordingly, the delavirdine-sensitive current contained an early and transient, as well as a late outward component corresponding to phases 1 and 3 of the AP. These results support the delavirdine-evoked inhibition of IKr, transient outward potassium current as well as sodium current. As these concentrations are similar to therapeutic plasma values (7-30 µM), delavirdine application might carry some risk of cardiac side effects especially in HIV-infected patients with altered cardiac function or patients with co-administration of drugs metabolized by certain cytochrome P450 isoforms.
Collapse
Affiliation(s)
- József Óvári
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Dental Sciences, University of Debrecen, Debrecen, Hungary
| | - Zsigmond Máté Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Geraldo Jorge Domingos
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Adam Feher
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
2
|
Fiore CM, Quigley K, Vorobyov I, Clancy CE, Harvey RD. Effect of the Membrane Environment on Pharmacologic Inhibition of hERG K + Channel Activity. JACC Clin Electrophysiol 2025:S2405-500X(24)01022-3. [PMID: 39895450 DOI: 10.1016/j.jacep.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND hERG encodes KV11.1 voltage-gated K+ channels, which generate the rapidly activating delayed rectifier K+ current that contributes to repolarization of the cardiac action potential. In addition to being targeted by many class III antiarrhythmic agents, these channels are also inhibited by a multitude of other pharmacological compounds, which can produce acquired long QT syndrome, leading to polymorphic ventricular tachycardia. While most drugs are thought to interact with a hydrophilic binding site in the channel pore, it has been postulated that some compounds act by perturbing the membrane environment or acting at hydrophobic sites accessed through the plasma membrane. OBJECTIVES Because hERG channels reside in cholesterol rich lipid raft domains, we hypothesized that disrupting the membrane environment by depleting cholesterol might alter inhibition of channel activity by certain drugs. METHODS We tested our hypothesis by examining the effect that depleting membrane cholesterol with methyl-β-cyclodextrin has on the ability of several compounds to inhibit hERG channels expressed in HEK293 cells. RESULTS We found that cholesterol depletion significantly increased the sensitivity of the whole cell current to inhibition by ibutilide, while decreasing the currents sensitivity to dofetilide and amiodarone at negative membrane potentials. CONCLUSIONS These results support the idea that the lipid environment of the plasma membrane plays a role in the ability of certain drugs to inhibit hERG channel activity. Differences in membrane cholesterol content may affect the ability of some hERG channel blockers to produce arrhythmogenic behavior.
Collapse
Affiliation(s)
- Chase M Fiore
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA
| | - Kate Quigley
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California- Davis, Davis, California, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California- Davis, Davis, California, USA
| | - Robert D Harvey
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA.
| |
Collapse
|
3
|
Davis J, Cornwell JD, Campagna N, Guo J, Li W, Yang T, Wang T, Zhang S. Rescue of expression and function of long QT syndrome-causing mutant hERG channels by enhancing channel stability in the plasma membrane. J Biol Chem 2024; 300:107526. [PMID: 38960041 PMCID: PMC11325228 DOI: 10.1016/j.jbc.2024.107526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes the Kv11.1 (or hERG) channel that conducts the rapidly activating delayed rectifier potassium current (IKr). Naturally occurring mutations in hERG impair the channel function and cause long QT syndrome type 2. Many missense hERG mutations lead to a lack of channel expression on the cell surface, representing a major mechanism for the loss-of-function of mutant channels. While it is generally thought that a trafficking defect underlies the lack of channel expression on the cell surface, in the present study, we demonstrate that the trafficking defective mutant hERG G601S can reach the plasma membrane but is unstable and quickly degrades, which is akin to WT hERG channels under low K+ conditions. We previously showed that serine (S) residue at 624 in the innermost position of the selectivity filter of hERG is involved in hERG membrane stability such that substitution of serine 624 with threonine (S624T) enhances hERG stability and renders hERG insensitive to low K+ culture. Here, we report that the intragenic addition of S624T substitution to trafficking defective hERG mutants G601S, N470D, and P596R led to a complete rescue of the function of these otherwise loss-of-function mutant channels to a level similar to the WT channel, representing the most effective rescue means for the function of mutant hERG channels. These findings not only provide novel insights into hERG mutation-mediated channel dysfunction but also point to the critical role of S624 in hERG stability on the plasma membrane.
Collapse
Affiliation(s)
- Jordan Davis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - James D Cornwell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Noah Campagna
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tingzhong Wang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
4
|
Osorio LA, Lozano M, Soto P, Moreno-Hidalgo V, Arévalo-Gil A, Ramírez-Balaguera A, Hevia D, Cifuentes J, Hidalgo Y, Alcayaga-Miranda F, Pasten C, Morales D, Varela D, Urquidi C, Iturriaga A, Rivera-Palma A, Larrea-Gómez R, Irarrázabal CE. Levels of Small Extracellular Vesicles Containing hERG-1 and Hsp47 as Potential Biomarkers for Cardiovascular Diseases. Int J Mol Sci 2024; 25:4913. [PMID: 38732154 PMCID: PMC11084293 DOI: 10.3390/ijms25094913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 05/13/2024] Open
Abstract
The diagnosis of cardiovascular disease (CVD) is still limited. Therefore, this study demonstrates the presence of human ether-a-go-go-related gene 1 (hERG1) and heat shock protein 47 (Hsp47) on the surface of small extracellular vesicles (sEVs) in human peripheral blood and their association with CVD. In this research, 20 individuals with heart failure and 26 participants subjected to cardiac stress tests were enrolled. The associations between hERG1 and/or Hsp47 in sEVs and CVD were established using Western blot, flow cytometry, electron microscopy, ELISA, and nanoparticle tracking analysis. The results show that hERG1 and Hsp47 were present in sEV membranes, extravesicularly exposing the sequences 430AFLLKETEEGPPATE445 for hERG1 and 169ALQSINEWAAQTT- DGKLPEVTKDVERTD196 for Hsp47. In addition, upon exposure to hypoxia, rat primary cardiomyocytes released sEVs into the media, and human cardiomyocytes in culture also released sEVs containing hERG1 (EV-hERG1) and/or Hsp47 (EV-Hsp47). Moreover, the levels of sEVs increased in the blood when cardiac ischemia was induced during the stress test, as well as the concentrations of EV-hERG1 and EV-Hsp47. Additionally, the plasma levels of EV-hERG1 and EV-Hsp47 decreased in patients with decompensated heart failure (DHF). Our data provide the first evidence that hERG1 and Hsp47 are present in the membranes of sEVs derived from the human cardiomyocyte cell line, and also in those isolated from human peripheral blood. Total sEVs, EV-hERG1, and EV-Hsp47 may be explored as biomarkers for heart diseases such as heart failure and cardiac ischemia.
Collapse
Affiliation(s)
- Luis A. Osorio
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Mauricio Lozano
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Paola Soto
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Viviana Moreno-Hidalgo
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angely Arévalo-Gil
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angie Ramírez-Balaguera
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Daniel Hevia
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Jorge Cifuentes
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Yessia Hidalgo
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Consuelo Pasten
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| | - Danna Morales
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Varela
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Cinthya Urquidi
- Department of Epidemiology and Health Studies, Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Andrés Iturriaga
- Departamento de Matemática y Ciencia de la Computación, Facultad de Ciencia, Universidad de Santiago de Chile, Santiago 9170020, Chile
| | | | | | - Carlos E. Irarrázabal
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| |
Collapse
|
5
|
Kovács ZM, Óvári J, Dienes C, Magyar J, Bányász T, Nánási PP, Horváth B, Feher A, Varga Z, Szentandrássy N. ABT-333 (Dasabuvir) Increases Action Potential Duration and Provokes Early Afterdepolarizations in Canine Left Ventricular Cells via Inhibition of I Kr. Pharmaceuticals (Basel) 2023; 16:488. [PMID: 37111245 PMCID: PMC10143825 DOI: 10.3390/ph16040488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
ABT-333 (dasabuvir) is an antiviral agent used in hepatitis C treatment. The molecule, similarly to some inhibitors of hERG channels, responsible for the delayed rectifier potassium current (IKr), contains the methanesulfonamide group. Reduced IKr current leads to long QT syndrome and early afterdepolarizations (EADs), therefore potentially causing life-threatening arrhythmias and sudden cardiac death. Our goal was to investigate the acute effects of ABT-333 in enzymatically isolated canine left ventricular myocardial cells. Action potentials (APs) and ion currents were recorded with a sharp microelectrode technique and whole-cell patch clamp, respectively. Application of 1 μM ABT-333 prolonged the AP in a reversible manner. The maximal rates of phases 0 and 1 were irreversibly decreased. Higher ABT-333 concentrations caused larger AP prolongation, elevation of the early plateau potential, and reduction of maximal rates of phases 0, 1, and 3. EADs occurred in some cells in 3-30 μM ABT-333 concentrations. The 10 μM ABT-333-sensitive current, recorded with AP voltage clamp, contained a late outward component corresponding to IKr and an early outward one corresponding to transient outward potassium current (Ito). ABT-333 reduced hERG-channel-mediated ion current in a concentration-dependent, partially reversible manner with a half-inhibitory concentration of 3.2 μM. As the therapeutic plasma concentration of ABT-333 can reach the low μM range, ABT-333 application carries a risk of cardiac side effects especially in case of coadministration with strong inhibitors of CYP2C8.
Collapse
Affiliation(s)
- Zsigmond Máté Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - József Óvári
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Doctoral School of Dental Sciences, University of Debrecen, H-4032 Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, H-4032 Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Adam Feher
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
6
|
Li XT. Beneficial effects of carvedilol modulating potassium channels on the control of glucose. Biomed Pharmacother 2022; 150:113057. [PMID: 35658228 DOI: 10.1016/j.biopha.2022.113057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/25/2022] Open
Abstract
The increased prevalence of hypertensive patients with type 2 diabetes mellitus (T2DM) is evident worldwide, leading to a higher risk of cardiovascular disease onset, which is substantially associated with disabilities and mortality in the clinic. In order to achieve the satisfyingly clinical outcomes and prognosis, the comprehensive therapies have been conducted with a beneficial effect on both blood pressure and glucose homeostasis, and clinical trials reveal that some kind of antihypertensive drugs such as angiotensin converting enzyme inhibitors (ACE-I) may, at least in part, meet the dual requirement during the disease management. As a nonselective β-blocker, carvedilol is employed for treating many cardiovascular diseases in clinical practice, including hypertension, angina pectoris and heart failure, and also exhibit the effectiveness for glycemic control and insulin resistance. Apart from alleviating sympathetic nervous system activity, several causes, such as lowering oxygen reactive species, may contribute to the effects of carvedilol on controlling plasma glucose levels, suggesting a feature of this drug having multiple targets. Interestingly, numerous distinct K+ channels expressed in pancreatic β-cells and peripheral insulin-sensitive tissues, which play a sentential role in glucose metabolism, are subjected to extensive modulation of carvdilol, establishing a linkage between K+ channels and drug's effects on the control of glucose. A variety of evidence shows that the impact of carvedilol on different K+ channels, including Kv, KAch, KATP and K2 P, can lead to positive influences for glucose homeostasis, contributing to its clinical beneficial effectiveness in treatment of hypertensive patients with T2DM. This review focus on the control of plasma glucose conferred by carvedilol modulation on K+ channels, providing the novel mechanistic explanation for drug's actions.
Collapse
Affiliation(s)
- Xian-Tao Li
- Department of Neuroscience, South-Central University for Nationalities, Wuhan 430074, China; School of Medicine, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
7
|
Abramochkin DV, Filatova TS, Pustovit KB, Voronina YA, Kuzmin VS, Vornanen M. Ionic currents underlying different patterns of electrical activity in working cardiac myocytes of mammals and non-mammalian vertebrates. Comp Biochem Physiol A Mol Integr Physiol 2022; 268:111204. [PMID: 35346823 DOI: 10.1016/j.cbpa.2022.111204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022]
Abstract
The orderly contraction of the vertebrate heart is determined by generation and propagation of cardiac action potentials (APs). APs are generated by the integrated activity of time- and voltage-dependent ionic channels which carry inward Na+ and Ca2+ currents, and outward K+ currents. This review compares atrial and ventricular APs and underlying ion currents between different taxa of vertebrates. We have collected literature data and attempted to find common electrophysiological features for two or more vertebrate groups, show differences between taxa and cardiac chambers, and indicate gaps in the existing data. Although electrical excitability of the heart in all vertebrates is based on the same superfamily of channels, there is a vast variability of AP waveforms between atrial and ventricular myocytes, between different species of the same vertebrate class and between endothermic and ectothermic animals. The wide variability of AP shapes is related to species-specific differences in animal size, heart rate, stage of ontogenetic development, excitation-contraction coupling, temperature and oxygen availability. Some of the differences between taxa are related to evolutionary development of genomes, which appear e.g. in the expression of different Na+ and K+ channel orthologues in cardiomyocytes of vertebrates. There is a wonderful variability of AP shapes and underlying ion currents with which electrical excitability of vertebrate heart can be generated depending on the intrinsic and extrinsic conditions of animal body. This multitude of ionic mechanisms provides excellent material for studying how the function of the vertebrate heart can adapt or acclimate to prevailing physiological and environmental conditions.
Collapse
Affiliation(s)
- Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia.
| | - Tatiana S Filatova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia
| | - Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia
| | - Yana A Voronina
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia; Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, 3(rd) Cherepkovskaya str., 15A, Moscow, Russia
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia; Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
8
|
Abstract
Human ether-a-go-go-related gene (hERG) channel plays an essential role in the repolarization of the cardiac action potential. Genetic mutations and some chemicals/drugs interfere with hERG channel activity, which may prolong the QT interval and potentially cause long QT syndrome. The FluxOR™ thallium flux assay performed in two cell lines, U2OS and HEK293, with stable hERG expression can be used to identify compounds that inhibit hERG channel activity. This chapter describes a cell-based hERG channel inhibition assay that has been optimized and performed in a 1536-well plate format. The homogeneous and robust assay can be used to identify compounds that inhibit hERG channel activity.
Collapse
Affiliation(s)
- Jinghua Zhao
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Ozturk N, Uslu S, Ozdemir S. Diabetes-induced changes in cardiac voltage-gated ion channels. World J Diabetes 2021; 12:1-18. [PMID: 33520105 PMCID: PMC7807254 DOI: 10.4239/wjd.v12.i1.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus affects the heart through various mechanisms such as microvascular defects, metabolic abnormalities, autonomic dysfunction and incompatible immune response. Furthermore, it can also cause functional and structural changes in the myocardium by a disease known as diabetic cardiomyopathy (DCM) in the absence of coronary artery disease. As DCM progresses it causes electrical remodeling of the heart, left ventricular dysfunction and heart failure. Electrophysiological changes in the diabetic heart contribute significantly to the incidence of arrhythmias and sudden cardiac death in diabetes mellitus patients. In recent studies, significant changes in repolarizing K+ currents, Na+ currents and L-type Ca2+ currents along with impaired Ca2+ homeostasis and defective contractile function have been identified in the diabetic heart. In addition, insulin levels and other trophic factors change significantly to maintain the ionic channel expression in diabetic patients. There are many diagnostic tools and management options for DCM, but it is difficult to detect its development and to effectively prevent its progress. In this review, diabetes-associated alterations in voltage-sensitive cardiac ion channels are comprehensively assessed to understand their potential role in the pathophysiology and pathogenesis of DCM.
Collapse
Affiliation(s)
- Nihal Ozturk
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Serkan Uslu
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Semir Ozdemir
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| |
Collapse
|
10
|
Hegyi B, Chen-Izu Y, Izu LT, Rajamani S, Belardinelli L, Bers DM, Bányász T. Balance Between Rapid Delayed Rectifier K + Current and Late Na + Current on Ventricular Repolarization: An Effective Antiarrhythmic Target? Circ Arrhythm Electrophysiol 2020; 13:e008130. [PMID: 32202931 PMCID: PMC7331791 DOI: 10.1161/circep.119.008130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Rapid delayed rectifier K+ current (IKr) and late Na+ current (INaL) significantly shape the cardiac action potential (AP). Changes in their magnitudes can cause either long or short QT syndromes associated with malignant ventricular arrhythmias and sudden cardiac death. METHODS Physiological self AP-clamp was used to measure INaL and IKr during the AP in rabbit and porcine ventricular cardiomyocytes to test our hypothesis that the balance between IKr and INaL affects repolarization stability in health and disease conditions. RESULTS We found comparable amount of net charge carried by IKr and INaL during the physiological AP, suggesting that outward K+ current via IKr and inward Na+ current via INaL are in balance during physiological repolarization. Remarkably, IKr and INaL integrals in each control myocyte were highly correlated in both healthy rabbit and pig myocytes, despite high overall cell-to-cell variability. This close correlation was lost in heart failure myocytes from both species. Pretreatment with E-4031 to block IKr (mimicking long QT syndrome 2) or with sea anemone toxin II to impair Na+ channel inactivation (mimicking long QT syndrome 3) prolonged AP duration (APD); however, using GS-967 to inhibit INaL sufficiently restored APD to control in both cases. Importantly, INaL inhibition significantly reduced the beat-to-beat and short-term variabilities of APD. Moreover, INaL inhibition also restored APD and repolarization stability in heart failure. Conversely, pretreatment with GS-967 shortened APD (mimicking short QT syndrome), and E-4031 reverted APD shortening. Furthermore, the amplitude of AP alternans occurring at high pacing frequency was decreased by INaL inhibition, increased by IKr inhibition, and restored by combined INaL and IKr inhibitions. CONCLUSIONS Our data demonstrate that IKr and INaL are counterbalancing currents during the physiological ventricular AP and their integrals covary in individual myocytes. Targeting these ionic currents to normalize their balance may have significant therapeutic potential in heart diseases with repolarization abnormalities. Visual Overview: A visual overview is available for this article.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis
- Department of Biomedical Engineering, University of California, Davis
- Department of Internal Medicine/Cardiology, University of California, Davis
| | | | - Sridharan Rajamani
- Amgen, Inc., South San Francisco, University of Debrecen, Debrecen, Hungary
| | - Luiz Belardinelli
- InCarda Therapeutics, Inc., Newark, CA, University of Debrecen, Debrecen, Hungary
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis
| | - Tamás Bányász
- Department of Pharmacology, University of California, Davis
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
11
|
Jeevaratnam K, Chadda KR, Huang CLH, Camm AJ. Cardiac Potassium Channels: Physiological Insights for Targeted Therapy. J Cardiovasc Pharmacol Ther 2017; 23:119-129. [PMID: 28946759 PMCID: PMC5808825 DOI: 10.1177/1074248417729880] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of novel drugs specifically directed at the ion channels underlying particular features of cardiac action potential (AP) initiation, recovery, and refractoriness would contribute to an optimized approach to antiarrhythmic therapy that minimizes potential cardiac and extracardiac toxicity. Of these, K+ channels contribute numerous and diverse currents with specific actions on different phases in the time course of AP repolarization. These features and their site-specific distribution make particular K+ channel types attractive therapeutic targets for the development of pharmacological agents attempting antiarrhythmic therapy in conditions such as atrial fibrillation. However, progress in the development of such temporally and spatially selective antiarrhythmic drugs against particular ion channels has been relatively limited, particularly in view of our incomplete understanding of the complex physiological roles and interactions of the various ionic currents. This review summarizes the physiological properties of the main cardiac potassium channels and the way in which they modulate cardiac electrical activity and then critiques a number of available potential antiarrhythmic drugs directed at them.
Collapse
Affiliation(s)
- Kamalan Jeevaratnam
- 1 Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,2 School of Medicine, Perdana University-Royal College of Surgeons Ireland, Serdang, Selangor Darul Ehsan, Malaysia
| | - Karan R Chadda
- 1 Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,3 Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L-H Huang
- 3 Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom.,4 Division of Cardiovascular Biology, Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - A John Camm
- 5 Cardiac Clinical Academic Group, St George's Hospital Medical School, University of London, Cranmer Terrace, London, United Kingdom
| |
Collapse
|
12
|
Kan L, Zhao W, Pan L, Xu J, Chen Q, Xu K, Xiao L, Chen Y. Peimine inhibits hERG potassium channels through the channel inactivation states. Biomed Pharmacother 2017; 89:838-844. [DOI: 10.1016/j.biopha.2017.02.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 11/29/2022] Open
|
13
|
Abstract
Cardiac delayed rectifier potassium channels conduct outward potassium currents during the plateau phase of action potentials and play pivotal roles in cardiac repolarization. These include IKs, IKr and the atrial specific IKur channels. In this article, we will review their molecular identities and biophysical properties. Mutations in the genes encoding delayed rectifiers lead to loss- or gain-of-function phenotypes, disrupt normal cardiac repolarization and result in various cardiac rhythm disorders, including congenital Long QT Syndrome, Short QT Syndrome and familial atrial fibrillation. We will also discuss the prospect of using delayed rectifier channels as therapeutic targets to manage cardiac arrhythmia.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Kevin J Sampson
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Robert S Kass
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
14
|
Liu J, Laksman Z, Backx PH. The electrophysiological development of cardiomyocytes. Adv Drug Deliv Rev 2016; 96:253-73. [PMID: 26788696 DOI: 10.1016/j.addr.2015.12.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023]
Abstract
The generation of human cardiomyocytes (CMs) from human pluripotent stem cells (hPSCs) has become an important resource for modeling human cardiac disease and for drug screening, and also holds significant potential for cardiac regeneration. Many challenges remain to be overcome however, before innovation in this field can translate into a change in the morbidity and mortality associated with heart disease. Of particular importance for the future application of this technology is an improved understanding of the electrophysiologic characteristics of CMs, so that better protocols can be developed and optimized for generating hPSC-CMs. Many different cell culture protocols are currently utilized to generate CMs from hPSCs and all appear to yield relatively “developmentally” immature CMs with highly heterogeneous electrical properties. These hPSC-CMs are characterized by spontaneous beating at highly variable rates with a broad range of depolarization-repolarization patterns, suggestive of mixed populations containing atrial, ventricular and nodal cells. Many recent studies have attempted to introduce approaches to promote maturation and to create cells with specific functional properties. In this review, we summarize the studies in which the electrical properties of CMs derived from stem cells have been examined. In order to place this information in a useful context, we also review the electrical properties of CMs as they transition from the developing embryo to the adult human heart. The signal pathways involved in the regulation of ion channel expression during development are also briefly considered.
Collapse
|
15
|
Haraguchi Y, Ohtsuki A, Oka T, Shimizu T. Electrophysiological analysis of mammalian cells expressing hERG using automated 384-well-patch-clamp. BMC Pharmacol Toxicol 2015; 16:39. [PMID: 26671227 PMCID: PMC4681162 DOI: 10.1186/s40360-015-0042-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An in vitro electrophysiological assay system, which can assess compound effects and thus show cardiotoxicity including arrhythmia risks of test drugs, is an essential method in the field of drug development and toxicology. METHODS In this study, high-throughput electrophysiological recordings of human embryonic kidney (HEK 293) cells and Chinese hamster ovary (CHO) cells stably expressing human ether-a-go-go related gene (hERG) were performed utilizing an automated 384-well-patch-clamp system, which records up to 384 cells simultaneously. hERG channel inhibition, which is closely related to a drug-induced QT prolongation and is increasing the risk of sudden cardiac death, was investigated in the high-throughput screening patch-clamp system. RESULTS In the automated patch-clamp measurements performed here, Kv currents were investigated with high efficiency. Various hERG channel blockers showed concentration-dependent inhibition, the 50 % inhibitory concentrations (IC50) of those blockers were in good agreement with previous reports. CONCLUSIONS The high-throughput patch-clamp system has a high potential in the field of pharmacology, toxicology, and cardiac physiology, and will contribute to the acceleration of pharmaceutical drug development and drug safety testing.
Collapse
Affiliation(s)
- Yuji Haraguchi
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Atsushi Ohtsuki
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.,Nanion Technologies GmbH, Gabrielenstr.9, 80636, Munich, Germany
| | - Takayuki Oka
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.,Nanion Technologies GmbH, Gabrielenstr.9, 80636, Munich, Germany
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| |
Collapse
|
16
|
Long VP, Bonilla IM, Vargas-Pinto P, Nishijima Y, Sridhar A, Li C, Mowrey K, Wright P, Velayutham M, Kumar S, Lee NY, Zweier JL, Mohler PJ, Györke S, Carnes CA. Heart failure duration progressively modulates the arrhythmia substrate through structural and electrical remodeling. Life Sci 2015; 123:61-71. [PMID: 25596015 PMCID: PMC4763601 DOI: 10.1016/j.lfs.2014.12.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 01/10/2023]
Abstract
AIMS Ventricular arrhythmias are a common cause of death in patients with heart failure (HF). Structural and electrical abnormalities in the heart provide a substrate for such arrhythmias. Canine tachypacing-induced HF models of 4-6 weeks duration are often used to study pathophysiology and therapies for HF. We hypothesized that a chronic canine model of HF would result in greater electrical and structural remodeling than a short term model, leading to a more arrhythmogenic substrate. MAIN METHODS HF was induced by ventricular tachypacing for one (short-term) or four (chronic) months to study remodeling. KEY FINDINGS Left ventricular contractility was progressively reduced, while ventricular hypertrophy and interstitial fibrosis were evident at 4 month but not 1 month of HF. Left ventricular myocyte action potentials were prolonged after 4 (p<0.05) but not 1 month of HF. Repolarization instability and early afterdepolarizations were evident only after 4 months of HF (p<0.05), coinciding with a prolonged QTc interval (p<0.05). The transient outward potassium current was reduced in both HF groups (p<0.05). The outward component of the inward rectifier potassium current was reduced only in the 4 month HF group (p<0.05). The delayed rectifier potassium currents were reduced in 4 (p<0.05) but not 1 month of HF. Reactive oxygen species were increased at both 1 and 4 months of HF (p<0.05). SIGNIFICANCE Reduced Ito, outward IK1, IKs, and IKr in HF contribute to EAD formation. Chronic, but not short term canine HF, results in the altered electrophysiology and repolarization instability characteristic of end-stage human HF.
Collapse
Affiliation(s)
- Victor P Long
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Ingrid M Bonilla
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Pedro Vargas-Pinto
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Arun Sridhar
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Chun Li
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | | | - Patrick Wright
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Murugesan Velayutham
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sanjay Kumar
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Nam Y Lee
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Jay L Zweier
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Sandor Györke
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Cynthia A Carnes
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
17
|
Gasparoli L, D'Amico M, Masselli M, Pillozzi S, Caves R, Khuwaileh R, Tiedke W, Mugridge K, Pratesi A, Mitcheson JS, Basso G, Becchetti A, Arcangeli A. New pyrimido-indole compound CD-160130 preferentially inhibits the KV11.1B isoform and produces antileukemic effects without cardiotoxicity. Mol Pharmacol 2015; 87:183-96. [PMID: 25411366 DOI: 10.1124/mol.114.094920] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
KV11.1 (hERG1) channels are often overexpressed in human cancers. In leukemias, KV11.1 regulates pro-survival signals that promote resistance to chemotherapy, raising the possibility that inhibitors of KV11.1 could be therapeutically beneficial. However, because of the role of KV11.1 in cardiac repolarization, blocking these channels may cause cardiac arrhythmias. We show that CD-160130, a novel pyrimido-indole compound, blocks KV11.1 channels with a higher efficacy for the KV11.1 isoform B, in which the IC50 (1.8 μM) was approximately 10-fold lower than observed in KV11.1 isoform A. At this concentration, CD-160130 also had minor effects on Kir2.1, KV 1.3, Kv1.5, and KCa3.1. In vitro, CD-160130 induced leukemia cell apoptosis, and could overcome bone marrow mesenchymal stromal cell (MSC)-induced chemoresistance. This effect was caused by interference with the survival signaling pathways triggered by MSCs. In vivo, CD-160130 produced an antileukemic activity, stronger than that caused by cytarabine. Consistent with its atypical target specificity, CD-160130 did not bind to the main binding site of the arrhythmogenic KV11.1 blockers (the Phe656 pore residue). Importantly, in guinea pigs CD-160130 produced neither alteration of the cardiac action potential shape in dissociated cardiomyocytes nor any lengthening of the QT interval in vivo. Moreover, CD-160130 had no myelotoxicity on human bone marrow-derived cells. Therefore, CD-160130 is a promising first-in-class compound to attempt oncologic therapy without cardiotoxicity, based on targeting KV11.1. Because leukemia and cardiac cells tend to express different ratios of the A and B KV11.1 isoforms, the pharmacological properties of CD-160130 may depend, at least in part, on isoform specificity.
Collapse
Affiliation(s)
- Luca Gasparoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Massimo D'Amico
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Marika Masselli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Rachel Caves
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Rawan Khuwaileh
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Wolfgang Tiedke
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Kenneth Mugridge
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Alessandro Pratesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - John S Mitcheson
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Giuseppe Basso
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Andrea Becchetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| |
Collapse
|
18
|
Wang S, Min XY, Pang SS, Qian J, Xu D, Guo Y. Adrenergic regulation of the rapid component of delayed rectifier K+ currents in guinea pig cardiomyocytes. J Thorac Dis 2015; 6:1778-84. [PMID: 25589973 DOI: 10.3978/j.issn.2072-1439.2014.12.27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/13/2014] [Indexed: 11/14/2022]
Abstract
BACKGROUND Guinea pig ventricular cardiomyocytes display the rapid component of the delayed rectifier potassium current (Ikr) that contributes to ventricular repolarization and promotes stress-induced arrhythmias. Adrenergic stimulation favors ventricular arrhythmogenesis but its effects on Ikr are poorly understood. METHODS Adrenergic modulation of Ikr was studied in isolated guinea pig ventricular cardiomyocytes using whole-cell patch clamping. RESULTS We found that the Ikr amplitude was reduced to 0.66±0.02 and 0.62±0.03 in response to 0.1 µM phenylephrine (PE), an α1AR agonist, and 10 µM isoproterenol (ISO), a βAR agonist, respectively. The effect of PE can be blocked by the selective α1A-adrenoceptor antagonist 5-methylurapidil, but not by the α1B-adrenoceptor antagonist chloroethylclonidine or α1D-adrenoceptor antagonist BMY7378. Additionally, the effect of ISO can be blocked by the β1-selective AR antagonist CGP-20712A, but not by the β2-selective AR antagonist ICI-118551. Although PE and ISO was continuously added to cells, ISO did not decrease the current to a greater extent when cells were first given PE. In addition, PE's effect on Ikr was suppressed by β1AR stimulation. CONCLUSIONS Ikr can by regulated by both the α1 and β ARs system, and that in addition to direct regulation by each receptor system, crosstalk may exist between the two systems.
Collapse
Affiliation(s)
- Sen Wang
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiao-Yan Min
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Si-Si Pang
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jin Qian
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Di Xu
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yan Guo
- Department of Geriatric Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
19
|
Cai Y, Wang Y, Xu J, Zuo X, Xu Y. Down-regulation of ether-a-go-go-related gene potassium channel protein through sustained stimulation of AT1 receptor by angiotensin II. Biochem Biophys Res Commun 2014; 452:852-7. [PMID: 25218469 DOI: 10.1016/j.bbrc.2014.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 01/26/2023]
Abstract
We investigated the effects of AT1 receptor stimulation by angiotensin II (Ang II) on human ether-a-go-go-related gene (hERG) potassium channel protein in a heterogeneous expression system with the human embryonic kidney (HEK) 293 cells which stably expressed hERG channel protein and were transiently transfected with the human AT1 receptors (HEK293/hERG). Western-blot analysis showed that Ang II significantly decreased the expression of mature hERG channel protein (155-kDa band) in a time- and dose-dependent manner without affecting the level of immature hERG channel protein (135-kDa band). The relative intensity of 155-kDa band was 64.7±6.8% of control (P<0.01) after treatment of Ang II at 100nM for 24h. To investigate the effect of Ang II on the degradation of mature hERG channel protein, we blocked forward trafficking from ER to Golgi with a Golgi transit inhibitor brefeldin A (10μM). Ang II significantly enhanced the time-dependent reduction of mature hERG channel protein. In addition, the proteasomal inhibitor lactacystin (5μM) inhibited Ang II-mediated the reduction of mature hERG channel protein, but the lysosomal inhibitor bafilomycin A1 (1μM) had no effect on the protein. The protein kinase C (PKC) inhibitor bisindolylmaleimide 1 (1μM) antagonized the reduction of mature hERG channel protein induced by Ang II. The results indicate that sustained stimulation of AT1 receptors by Ang II reduces the mature hERG channel protein via accelerating channel proteasomal degradation involving the PKC pathway.
Collapse
Affiliation(s)
- Yue Cai
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuhong Wang
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jia Xu
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xu Zuo
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yanfang Xu
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
20
|
Functional cross-talk between the α1- and β1-adrenergic receptors modulates the rapidly activating delayed rectifier potassium current in guinea pig ventricular myocytes. Int J Mol Sci 2014; 15:14220-33. [PMID: 25196520 PMCID: PMC4159847 DOI: 10.3390/ijms150814220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 12/02/2022] Open
Abstract
The rapidly activating delayed rectifier potassium current (IKr) plays a critical role in cardiac repolarization. Although IKr is known to be regulated by both α1- and β1-adrenergic receptors (ARs), the cross-talk and feedback mechanisms that dictate its response to α1- and β1-AR activation are not known. In the present study, IKr was recorded using the whole-cell patch-clamp technique. IKr amplitude was measured before and after the sequential application of selective adrenergic agonists targeting α1- and β1-ARs. Stimulation of either receptor alone (α1-ARs using 1 μM phenylephrine (PE) or β1-ARs using 10 μM xamoterol (Xamo)) reduced IKr by 0.22 ± 0.03 and 0.28 ± 0.01, respectively. The voltage-dependent activation curve of IKr shifted in the negative direction. The half-maximal activation voltage (V0.5) was altered by −6.35 ± 1.53 and −1.95 ± 2.22 mV, respectively, with no major change in the slope factor (k). When myocytes were pretreated with Xamo, PE-induced reduction in IKr was markedly blunted and the corresponding change in V0.5 was significantly altered. Similarly, when cells were pretreated with PE, Xamo-induced reduction of IKr was significantly attenuated. The present results demonstrate that functional cross-talk between α1- and β1-AR signaling regulates IKr. Such non-linear regulation may form a protective mechanism under excessive adrenergic stimulation.
Collapse
|
21
|
Affiliation(s)
- Enid Gilbert-Barness
- Laboratory Medicine, Pediatric, Obstetrics and Gynecology, Department of Pathology, College of Medicine, Tampa General Hospital, University of South Florida Morsani, 1 Tampa General Circle, Tampa, FL 33606, USA.
| |
Collapse
|
22
|
HAN SHENGNA, YANG SONGHUA, ZHANG YU, SUN XIAOYAN, DUAN YANYAN, HU XIANGJIE, FAN TIANLI, HUANG CHENZHENG, YANG GE, ZHANG ZHAO, ZHANG LIRONG. Identification and functional characterization of the human ether-a-go-go-related gene Q738X mutant associated with hereditary long QT syndrome type 2. Int J Mol Med 2014; 34:810-5. [DOI: 10.3892/ijmm.2014.1827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/23/2014] [Indexed: 11/06/2022] Open
|
23
|
Thurner P, Stary-Weinzinger A, Gafar H, Gawali VS, Kudlacek O, Zezula J, Hilber K, Boehm S, Sandtner W, Koenig X. Mechanism of hERG channel block by the psychoactive indole alkaloid ibogaine. J Pharmacol Exp Ther 2014; 348:346-58. [PMID: 24307198 DOI: 10.1124/jpet.113.209643] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ibogaine is a psychoactive indole alkaloid. Its use as an antiaddictive agent has been accompanied by QT prolongation and cardiac arrhythmias, which are most likely caused by human ether a go-go-related gene (hERG) potassium channel inhibition. Therefore, we studied in detail the interaction of ibogaine with hERG channels heterologously expressed in mammalian kidney tsA-201 cells. Currents through hERG channels were blocked regardless of whether ibogaine was applied via the extracellular or intracellular solution. The extent of inhibition was determined by the relative pH values. Block occurred during activation of the channels and was not observed for resting channels. With increasing depolarizations, ibogaine block grew and developed faster. Steady-state activation and inactivation of the channel were shifted to more negative potentials. Deactivation was slowed, whereas inactivation was accelerated. Mutations in the binding site reported for other hERG channel blockers (Y652A and F656A) reduced the potency of ibogaine, whereas an inactivation-deficient double mutant (G628C/S631C) was as sensitive as wild-type channels. Molecular drug docking indicated binding within the inner cavity of the channel independently of the protonation of ibogaine. Experimental current traces were fit to a kinetic model of hERG channel gating, revealing preferential binding of ibogaine to the open and inactivated state. Taken together, these findings show that ibogaine blocks hERG channels from the cytosolic side either in its charged form alone or in company with its uncharged form and alters the currents by changing the relative contribution of channel states over time.
Collapse
Affiliation(s)
- Patrick Thurner
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria (H.G., V.S.G., K.H., S.B., X.K.), Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Vienna, Austria (P.T., O.K., J.Z., W.S.), Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (A.S.-W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Konstantopoulou A, Tsikrikas S, Asvestas D, Korantzopoulos P, Letsas KP. Mechanisms of drug-induced proarrhythmia in clinical practice. World J Cardiol 2013; 5:175-185. [PMID: 23847724 PMCID: PMC3708047 DOI: 10.4330/wjc.v5.i6.175] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/05/2013] [Accepted: 05/19/2013] [Indexed: 02/06/2023] Open
Abstract
Drug-induced proarrhythmia represents a great challenge for those involved in the development of novel pharmaceuticals and in the regulatory bodies for drug approval as well as for the prescribing clinicians. Our understanding of the mechanisms that underlie drug-induced proarrhythmia has grown dramatically over the last two decades. A growing number of cardiac and non-cardiac agents have been shown to alter cardiac repolarization predisposing to fatal cardiac arrhythmias such as ventricular tachycardia or ventricular fibrillation and sudden cardiac death. These agents may induce the phenotype of long QT syndrome and less commonly of short QT syndrome and Brugada syndrome (BS). Although, genetic susceptibility underlie drug-induced proarrhythmia in certain cases, current data are limited regarding this topic. The present review surveys the current published literature on the mechanisms and the offending medical agents that predispose to drug-induced long QT syndrome, short QT syndrome and BS. Drug-induced proarrhythmia should be considered as a predictor of sudden cardiac death and should prompt critical re-evaluation of the risks and benefits of the suspicious medication. Survivors of drug-induced proarrhythmia and family members require careful examination and possibly genetic testing for the presence of a channelopathy. Treating physicians are advised to follow the lists of agents implicated in drug-induced proarrhythmia in order to minimize the risk of arrhythmia and sudden cardiac death.
Collapse
|
25
|
Lariccia V, Moraca A, Marini M, Nasti AA, Battistoni I, Amoroso S, Perna GP. Unusual case of severe arrhythmia developed after acute intoxication with tosylchloramide. BMC Pharmacol Toxicol 2013; 14:8. [PMID: 23347670 PMCID: PMC3566980 DOI: 10.1186/2050-6511-14-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 01/21/2013] [Indexed: 01/09/2023] Open
Abstract
Background Drugs not commonly considered to be cardioactive agents may cause prolongation of the QT interval with resultant torsades de pointes and ventricular fibrillation. This form of drug toxicity often causes cardiac arrest or sudden death. Case presentation After accidental ingestion of tosylchloramide a caucasian 77-year-old woman, with a family history of cardiovascular disease and hypertension, was admitted to the intensive care unit following episodes of torsades de pointes with a prolonged QT/QTc interval (640/542 ms). The patient received an implantable cardioverter-defibrillator, was discharged from the hospital with normal QT/QTc interval and did not experience additional ventricular arrhythmias during one year of follow-up. Conclusion This is the first report concerning an unusual case of torsades de pointes after accidental intoxication by ingestion of tosylchloramide. The pronounced impact of the oxidyzing agent tosylchloramide on the activity of some of the ion channels regulating the QT interval was identified as a probable cause of the arrhythmia.
Collapse
Affiliation(s)
- Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, University Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
26
|
Di Martino GP, Masetti M, Ceccarini L, Cavalli A, Recanatini M. An Automated Docking Protocol for hERG Channel Blockers. J Chem Inf Model 2013; 53:159-75. [DOI: 10.1021/ci300326d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Giovanni Paolo Di Martino
- Department of Pharmacy and Biotechnology,
Alma Mater Studiorum, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology,
Alma Mater Studiorum, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Luisa Ceccarini
- Department of Pharmacy and Biotechnology,
Alma Mater Studiorum, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology,
Alma Mater Studiorum, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
- Department of Drug Discovery
and Development, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Maurizio Recanatini
- Department of Pharmacy and Biotechnology,
Alma Mater Studiorum, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
27
|
Hausammann GJ, Heitkamp T, Matile H, Gsell B, Thoma R, Schmid G, Frasson D, Sievers M, Hennig M, Grütter MG. Generation of an antibody toolbox to characterize hERG. Biochem Biophys Res Commun 2012; 431:70-5. [PMID: 23277102 DOI: 10.1016/j.bbrc.2012.12.089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 11/28/2022]
Abstract
The human ether-a-go-go related gene (hERG) potassium channel plays a major role in the repolarization of the cardiac action potential. Inhibition of the hERG function by mutations or a wide variety of pharmaceutical compounds cause long QT syndrome and lead to potentially lethal arrhythmias. For detailed insights into the structural and biochemical background of hERG function and drug binding, the purification of recombinant protein is essential. Because the hERG channel is a challenging protein to purify, fast and easy techniques to evaluate different expression, solubilization and purification conditions are of primary importance. Here, we describe the generation of a set of 12 monoclonal antibodies against hERG. Beside their suitability in western blot, immunoprecipitation and immunostaining, these antibodies were used to establish a sandwich ELISA for the detection and relative quantification of hERG in different expression systems. Furthermore, a Fab fragment was used in fluorescence size exclusion chromatography to determine the oligomeric state of hERG after solubilization. These new tools can be used for a fast and efficient screening of expression, solubilization and purification conditions.
Collapse
Affiliation(s)
- Georg J Hausammann
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lee HA, Kim KS, Hyun SA, Park SG, Kim SJ. Wide spectrum of inhibitory effects of sertraline on cardiac ion channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:327-32. [PMID: 23118556 PMCID: PMC3484517 DOI: 10.4196/kjpp.2012.16.5.327] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 09/11/2012] [Accepted: 09/11/2012] [Indexed: 01/21/2023]
Abstract
Sertraline is a commonly used antidepressant of the selective serotonin reuptake inhibitors (SSRIs) class. In these experiments, we have used the whole cell patch clamp technique to examine the effects of sertraline on the major cardiac ion channels expressed in HEK293 cells and the native voltage-gated Ca(2+) channels in rat ventricular myocytes. According to the results, sertraline is a potent blocker of cardiac K(+) channels, such as hERG, I(Ks) and I(K1). The rank order of inhibitory potency was hERG >I(K1)> I(Ks) with IC(50) values of 0.7, 10.5, and 15.2 µM, respectively. In addition to K(+) channels, sertraline also inhibited I(Na) and I(Ca), and the IC(50) values are 6.1 and 2.6 µM, respectively. Modification of these ion channels by sertraline could induce changes of the cardiac action potential duration and QT interval, and might result in cardiac arrhythmia.
Collapse
Affiliation(s)
- Hyang-Ae Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejeon 305-600, Korea. ; Department of Physiology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | |
Collapse
|
29
|
Liu Q, Mao X, Zeng F, Jin S, Yang X. Effect of daurisoline on HERG channel electrophysiological function and protein expression. JOURNAL OF NATURAL PRODUCTS 2012; 75:1539-1545. [PMID: 22974355 DOI: 10.1021/np300232b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Daurisoline (1) is a bis-benzylisoquinoline alkaloid isolated from the rhizomes of Menispermum dauricum. The antiarrhythmic effect of 1 has been demonstrated in different experimental animals. In previous studies, daurisoline (1) prolonged action potential duration (APD) in a normal use-dependent manner. However, the electrophysiological mechanisms for 1-induced prolongation of APD have not been documented. In the present study, the direct effect of 1 was investigated on the hERG current and the expression of mRNA and protein in human embryonic kidney 293 (HEK293) cells stably expressing the hERG channel. It was shown that 1 inhibits hERG current in a concentration- and voltage-dependent manner. In the presence of 10 μM 1, steady-state inactivation of V(1/2) was shifted negatively by 15.9 mV, and 1 accelerated the onset of inactivation. Blockade of hERG channels was dependent on channel opening. The expression and function of hERG were unchanged by 1 at 1 and 10 μM, while hERG expression and the hERG current were decreased significantly by 1 at 30 μM. These results indicate that 1, at concentrations below 30 μM, exerts a blocking effect on hERG, but does not affect the expression and function of the hERG channel. This may explain the relatively lower risk of long QT syndrome after long-term usage.
Collapse
Affiliation(s)
- Qiangni Liu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology , The Key Laboratory of Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, People's Republic of China
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Jonsson MKB, van der Heyden MAG, van Veen TAB. Deciphering hERG channels: molecular basis of the rapid component of the delayed rectifier potassium current. J Mol Cell Cardiol 2012; 53:369-74. [PMID: 22742967 DOI: 10.1016/j.yjmcc.2012.06.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 06/07/2012] [Accepted: 06/19/2012] [Indexed: 12/23/2022]
Abstract
The rapid component of the delayed rectifier potassium current (I(Kr)), encoded by the ether-a-go-go-related gene (ERG1, officially denominated as KCNH2), is a major contributor to repolarization in the mammalian heart. Acute (e.g. drug-induced) and chronic (e.g. inherited genetic disorder) disruptions of this current can lead to prolongation of the action potential and potentiate occurrence of lethal arrhythmias. Many cardiac and non-cardiac drugs show high affinity for the I(Kr) channel and it is therefore extensively studied during safety pharmacology. The unique biophysical and pharmacological properties of the I(Kr) channel are largely recapitulated by expressing the human variant (hERG1a) in overexpressing systems. hERG1a channels are tetramers consisting of four 1159 amino acid long proteins and have electrophysiological properties similar, but not identical, to native I(Kr). In the search for an explanation to the discrepancies between I(Kr) and hERG1a channels, two alternative hERG1 proteins have been found. Alternative transcription of hERG1 leads to a protein with a 56 amino acid shorter N-terminus, known as hERG1b. hERG1b can form channels alone or coassemble with hERG1a. Alternative splicing leads to an alternate C-terminus and a protein known as hERGuso. hERGuso and hERG1b regulate hERG1a channel trafficking, functional expression and channel kinetics. Expression of hERGuso leads to a reduced number of channels at the plasma membrane and thereby reduces current density. On the contrary, co-assembly with hERG1b alters channel kinetics resulting in more available channels and a larger current. These findings have implication for understanding mechanisms of disease, acute and chronic drug effects, and potential gender differences.
Collapse
Affiliation(s)
- Malin K B Jonsson
- Department of Medical Physiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|
32
|
Zhao J, Lian Y, Lu C, Jing L, Yuan H, Peng S. Inhibitory effects of a bisbenzylisoquinline alkaloid dauricine on HERG potassium channels. JOURNAL OF ETHNOPHARMACOLOGY 2012; 141:685-691. [PMID: 21920426 DOI: 10.1016/j.jep.2011.08.054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 08/14/2011] [Accepted: 08/25/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The roots of Menispermum dauricum have been widely used for the treatment of inflammation, allergy and arrhythmia in China for a long time. Dauricine (Dau), a bisbenzylisoquinline alkaloid from Menispermum dauricum, mainly contributes to the anti-arrhythmic effect and has received pharmacological attention. Dau can prolong the action potential duration (APD), which has been attributed to its ability to modulate Ca(2+) and several K(+) channels. However, its effects on human-ether-a-go-go-related gene (HERG) channels are unknown. AIM OF THE STUDY The effects of Dau on HERG channels were investigated. MATERIALS AND METHODS Whole-cell patch-clamp technique was used to record HERG current (I(HERG)) carried by recombinant HERG channels expressed in HEK293 cells. RESULTS Dau inhibited I(HERG) in a concentration-dependent manner with an IC(50) of 3.5 μM. Development of block and washout were fast. The inhibitory action of Dau was contingent on channel gating, showing significant voltage and time dependence. Dau inhibited I(HERG) in the open and inactivated states, but not in the closed states. The activation curve was shifted in a negative direction. CONCLUSIONS Dau inhibits HERG encoded potassium channels and this action might be a molecular mechanism for the previously reported APD prolongation with this drug.
Collapse
Affiliation(s)
- Jing Zhao
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Fengtai District, Beijing, PR China.
| | | | | | | | | | | |
Collapse
|
33
|
Guo J, Zhang X, Hu Z, Zhuang Z, Zhu Z, Chen Z, Chen W, Zhao Z, Zhang C, Zhang Z. A422T mutation in HERG potassium channel retained in ER is rescurable by pharmacologic or molecular chaperones. Biochem Biophys Res Commun 2012; 422:305-10. [PMID: 22580281 DOI: 10.1016/j.bbrc.2012.04.153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 04/27/2012] [Indexed: 10/28/2022]
Abstract
In the present study, we characterized biologic and electrophysiologic consequences of A422T mutation in HERG K(+) channel and the role of pharmacologic or molecular chaperons by employing a heterogeneous expression system in HEK 293 cells. It was found that A422T mutation led to a marked decrease in whole-cell recording currents, and that a complexly glycosylated form protein band at 155 kDa was missing by Western blotting analysis compared to wild type (WT). And the mutant protein was mainly located in the cytoplasm as illustrated in immunocytochemical assay, indicating that the mutation underwent a trafficking defect. In addition, A422T mutation exerted remarkable dominant-negative suppression on WT, resulting in the alteration in the kinetic processes. Strikingly, trafficking-deficient A422T mutation was partially rescued by incubating the cells at a lower temperature, administration of pharmacologic chaperon, E4031 or overexpression of a chaperon molecule, Hsp90, but not Hsp70. In conclusion, missense A422T mutation in HERG K(+) channel results in its trafficking defect, which is rescurable by pharmacologic or molecular chaperones.
Collapse
Affiliation(s)
- Jia Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Interacting sites of scorpion toxin ErgTx1 with hERG1 K+ channels. Toxicon 2012; 59:633-41. [PMID: 22366117 DOI: 10.1016/j.toxicon.2012.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/12/2012] [Accepted: 02/02/2012] [Indexed: 11/23/2022]
Abstract
Peptides purified from scorpion venoms were shown to interact with specific amino acid residues present in the outer vestibule of various sub-types of potassium channels, occluding the pore and causing a decrement of K(+) permeability through the membrane of excitable and non excitable cells. This communication describes the identification of several interacting sites of toxin ErgTx1, a toxin purified from the venom of the scorpion Centruroides noxius, with the human ERG1 K(+) channels, by means of site-directed mutagenesis of specific residues of the toxin. Recombinant mutants of the gene coding for ErgTx1 were expressed heterologously in Escherichia coli, properly folded and their affinities and interactions with hERG1 channels were determined by patch-clamp techniques. Residues in position Y14, Y17 and F37 of the solvent exposed hydrophobic surface, and charged residues at the position K13 and K38 of ErgTx1 were shown to cause a decrement of the affinity from 20 folds to 3 orders of magnitude, thus suggesting that they are certainly participating on the binding surface of this toxin towards the hERG1 channels. Double mutants at positions K13 and F37, Y14 and F37, Y17 and F37 and K13 and K38 were also prepared and assayed, but the results obtained are not much different from the single point mutants of ErgTx1. The results of the present work indicate the most probable surface area of ErgTx1 that makes contact with the hERG channels.
Collapse
|
35
|
Jeong I, Choi BH, Yoon SH, Hahn SJ. Carvedilol blocks the cloned cardiac Kv1.5 channels in a β-adrenergic receptor-independent manner. Biochem Pharmacol 2012; 83:497-505. [PMID: 22146582 DOI: 10.1016/j.bcp.2011.11.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/21/2011] [Accepted: 11/21/2011] [Indexed: 10/14/2022]
Abstract
Carvedilol, a non-selective β-adrenergic blocker, is widely used for the treatment of angina pectoris and hypertension. We examined the action of carvedilol on cloned Kv1.5 expressed in CHO cells, using the whole-cell patch clamp technique. Carvedilol reduced the peak amplitude of Kv1.5 and accelerated the inactivation rate in a concentration-dependent manner with an IC50 of 2.56 μM. Using a first-order kinetics analysis, we calculated k(+1) = 19.68 μM(-1)s(-1) for the association rate constant, and k(-1) = 44.89 s(-1) for the dissociation rate constant. The apparent K(D) (k(-1)/k(+1)) was 2.28 μM, which is similar to the IC50 value. Other β-adrenergic blockers (alprenolol, oxprenolol and carteolol) had little or no effect on Kv1.5 currents. Carvedilol slowed the deactivation time course, resulting in a tail crossover phenomenon. Carvedilol-induced block was voltage-dependent in the voltage range for channel activation, but voltage-independent in the voltage range for full activation. The voltage dependences for both steady-state activation and inactivation were unchanged by carvedilol. Carvedilol affected Kv1.5 in a use-dependent manner. When stimulation frequencies were increased to quantify a use-dependent block, however, the block by carvedilol was slightly increased with IC50 values of 2.56 μM at 0.1 Hz, 2.38 μM at 1 Hz and 2.03 μM at 2 Hz. Carvedilol also slowed the time course of recovery from inactivation of Kv1.5. These results indicate that carvedilol blocks Kv1.5 in a reversible, concentration-, voltage-, time-, and use-dependent manner, but only at concentrations slightly higher than therapeutic plasma concentrations in humans. These effects are probably relevant to an understanding of the ionic mechanism underlying the antiarrhythmic property of carvedilol.
Collapse
Affiliation(s)
- Imju Jeong
- Department of Physiology, Medical Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | | | | | | |
Collapse
|
36
|
In silico analysis of conformational changes induced by mutation of aromatic binding residues: consequences for drug binding in the hERG K+ channel. PLoS One 2011; 6:e28778. [PMID: 22194911 PMCID: PMC3240635 DOI: 10.1371/journal.pone.0028778] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 11/15/2011] [Indexed: 01/24/2023] Open
Abstract
Pharmacological inhibition of cardiac hERG K+ channels is associated with increased risk of lethal arrhythmias. Many drugs reduce hERG current by directly binding to the channel, thereby blocking ion conduction. Mutation of two aromatic residues (F656 and Y652) substantially decreases the potency of numerous structurally diverse compounds. Nevertheless, some drugs are only weakly affected by mutation Y652A. In this study we utilize molecular dynamics simulations and docking studies to analyze the different effects of mutation Y652A on a selected number of hERG blockers. MD simulations reveal conformational changes in the binding site induced by mutation Y652A. Loss of π-π-stacking between the two aromatic residues induces a conformational change of the F656 side chain from a cavity facing to cavity lining orientation. Docking studies and MD simulations qualitatively reproduce the diverse experimentally observed modulatory effects of mutation Y652A and provide a new structural interpretation for the sensitivity differences.
Collapse
|
37
|
Es-Salah-Lamoureux Z, Xiong PY, Goodchild SJ, Ahern CA, Fedida D. Blockade of permeation by potassium but normal gating of the G628S nonconducting hERG channel mutant. Biophys J 2011; 101:662-70. [PMID: 21806934 DOI: 10.1016/j.bpj.2011.06.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 06/10/2011] [Accepted: 06/17/2011] [Indexed: 12/21/2022] Open
Abstract
G628S is a mutation in the signature sequence that forms the selectivity filter of the human ether-a-go-go-related gene (hERG) channel (GFG) and is associated with long-QT2 syndrome. G628S channels are known to have a dominant-negative effect on hERG currents, and the mutant is therefore thought to be nonfunctional. This study aims to assess the physiological mechanism that prevents the surface-expressing G628S channels from conducting ions. We used voltage-clamp fluorimetry along with two-microelectrode voltage clamping in Xenopus oocytes to confirm that the channels express well at the surface, and to show that they are actually functional, with activation kinetics comparable to that of wild-type, and that the mutation leads to a reduced selectivity to potassium. Although ionic currents are not detected in physiological solutions, removing extracellular K(+) results in the appearance of an inward Na(+)-dependent current. Using whole-cell patch clamp in mammalian transfected cells, we demonstrate that the G628S channels conduct Na(+), but that this can be blocked by both intracellular and higher-than-physiological extracellular K(+). Using solutions devoid of K(+) allows the appearance of nA-sized Na(+) currents with activation and inactivation gating analogous to wild-type channels. The G628S channels are functionally conducting but are normally blocked by intracellular K(+).
Collapse
Affiliation(s)
- Zeineb Es-Salah-Lamoureux
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
38
|
Models of HERG gating. Biophys J 2011; 101:631-42. [PMID: 21806931 DOI: 10.1016/j.bpj.2011.06.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 06/22/2011] [Accepted: 06/27/2011] [Indexed: 11/21/2022] Open
Abstract
HERG (Kv11.1, KCNH2) is a voltage-gated potassium channel with unique gating characteristics. HERG has fast voltage-dependent inactivation, relatively slow deactivation, and fast recovery from inactivation. This combination of gating kinetics makes study of HERG difficult without using mathematical models. Several HERG models have been developed, with fundamentally different organization. HERG is the molecular basis of I(Kr), which plays a critical role in repolarization. We programmed and compared five distinct HERG models. HERG gating cannot be adequately replicated using Hodgkin-Huxley type formulation. Using Markov models, a five-state model is required with three closed, one open, and one inactivated state, and a voltage-independent step between some of the closed states. A fundamental difference between models is the presence/absence of a transition directly from the proximal closed state to the inactivated state. The only models that effectively reproduce HERG data have no direct closed-inactivated transition, or have a closed-inactivated transition that is effectively zero compared to the closed-open transition, rendering the closed-inactivation transition superfluous. Our single-channel model demonstrates that channels can inactivate without conducting with a flickering or bursting open-state. The various models have qualitative and quantitative differences that are critical to accurate predictions of HERG behavior during repolarization, tachycardia, and premature depolarizations.
Collapse
|
39
|
Windisch A, Timin E, Schwarz T, Stork-Riedler D, Erker T, Ecker G, Hering S. Trapping and dissociation of propafenone derivatives in HERG channels. Br J Pharmacol 2011; 162:1542-52. [PMID: 21175572 DOI: 10.1111/j.1476-5381.2010.01159.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Human ether-a-go-go related gene (HERG) channel inhibitors may be subdivided into compounds that are trapped in the closed channel conformation and others that dissociate at rest. The structural peculiarities promoting resting state dissociation from HERG channels are currently unknown. A small molecule-like propafenone is efficiently trapped in the closed HERG channel conformation. The aim of this study was to identify structural moieties that would promote dissociation of propafenone derivatives. EXPERIMENTAL APPROACH Human ether-a-go-go related gene channels were heterologously expressed in Xenopus oocytes and potassium currents were recorded using the two-microelectrode voltage clamp technique. Recovery from block by 10 propafenone derivatives with variable side chains, but a conserved putative pharmacophore, was analysed. KEY RESULTS We have identified structural determinants of propafenone derivatives that enable drug dissociation from the closed channel state. Propafenone and four derivatives with 'short' side chains were trapped in the closed channel. Five out of six bulky derivatives efficiently dissociated from the channel at rest. One propafenone derivative with a similar bulk but lacking an H-bond acceptor in this region was trapped. Correlations were observed between molecular weight and onset of channel block as well as between pK(a) and recovery at rest. CONCLUSION AND IMPLICATIONS The data show that extending the size of a trapped HERG blocker-like propafenone by adding a bulky side chain may impede channel closure and thereby facilitate drug dissociation at rest. The presence of an H-bond acceptor in the bulky side chain is, however, essential.
Collapse
Affiliation(s)
- A Windisch
- Department of Pharmacology and Toxicology University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
40
|
Zehnder L, Bennett M, Meng J, Huang B, Ninkovic S, Wang F, Braganza J, Tatlock J, Jewell T, Zhou JZ, Burke B, Wang J, Maegley K, Mehta PP, Yin MJ, Gajiwala KS, Hickey MJ, Yamazaki S, Smith E, Kang P, Sistla A, Dovalsantos E, Gehring MR, Kania R, Wythes M, Kung PP. Optimization of Potent, Selective, and Orally Bioavailable Pyrrolodinopyrimidine-Containing Inhibitors of Heat Shock Protein 90. Identification of Development Candidate 2-Amino-4-{4-chloro-2-[2-(4-fluoro-1H-pyrazol-1-yl)ethoxy]-6-methylphenyl}-N-(2,2-difluoropropyl)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidine-6-carboxamide. J Med Chem 2011; 54:3368-85. [DOI: 10.1021/jm200128m] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Luke Zehnder
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michael Bennett
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jerry Meng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Buwen Huang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Sacha Ninkovic
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Fen Wang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - John Braganza
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - John Tatlock
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Tanya Jewell
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Joe Zhongxiang Zhou
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ben Burke
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jeff Wang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Karen Maegley
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Pramod P. Mehta
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Min-Jean Yin
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ketan S. Gajiwala
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michael J. Hickey
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Shinji Yamazaki
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Evan Smith
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ping Kang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Anand Sistla
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Elena Dovalsantos
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michael R. Gehring
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert Kania
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Martin Wythes
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Pei-Pei Kung
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
41
|
Xia M, Shahane SA, Huang R, Titus SA, Shum E, Zhao Y, Southall N, Zheng W, Witt KL, Tice RR, Austin CP. Identification of quaternary ammonium compounds as potent inhibitors of hERG potassium channels. Toxicol Appl Pharmacol 2011; 252:250-8. [PMID: 21362439 DOI: 10.1016/j.taap.2011.02.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 02/17/2011] [Accepted: 02/21/2011] [Indexed: 02/03/2023]
Abstract
The human ether-a-go-go-related gene (hERG) channel, a member of a family of voltage-gated potassium (K(+)) channels, plays a critical role in the repolarization of the cardiac action potential. The reduction of hERG channel activity as a result of adverse drug effects or genetic mutations may cause QT interval prolongation and potentially leads to acquired long QT syndrome. Thus, screening for hERG channel activity is important in drug development. Cardiotoxicity associated with the inhibition of hERG channels by environmental chemicals is also a public health concern. To assess the inhibitory effects of environmental chemicals on hERG channel function, we screened the National Toxicology Program (NTP) collection of 1408 compounds by measuring thallium influx into cells through hERG channels. Seventeen compounds with hERG channel inhibition were identified with IC(50) potencies ranging from 0.26 to 22μM. Twelve of these compounds were confirmed as hERG channel blockers in an automated whole cell patch clamp experiment. In addition, we investigated the structure-activity relationship of seven compounds belonging to the quaternary ammonium compound (QAC) series on hERG channel inhibition. Among four active QAC compounds, tetra-n-octylammonium bromide was the most potent with an IC(50) value of 260nM in the thallium influx assay and 80nM in the patch clamp assay. The potency of this class of hERG channel inhibitors appears to depend on the number and length of their aliphatic side-chains surrounding the charged nitrogen. Profiling environmental compound libraries for hERG channel inhibition provides information useful in prioritizing these compounds for cardiotoxicity assessment in vivo.
Collapse
Affiliation(s)
- Menghang Xia
- NIH Chemical Genomics Center, National Institutes of Health, Bethesda, MD 20892-3370, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Benson AP, Al-Owais M, Holden AV. Quantitative prediction of the arrhythmogenic effects of de novo hERG mutations in computational models of human ventricular tissues. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2011; 40:627-39. [PMID: 21234558 DOI: 10.1007/s00249-010-0663-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/13/2010] [Accepted: 12/20/2010] [Indexed: 11/24/2022]
Abstract
Mutations to hERG which result in changes to the rapid delayed rectifier current I(Kr) can cause long and short QT syndromes and are associated with an increased risk of cardiac arrhythmias. Experimental recordings of I(Kr) reveal the effects of mutations at the channel level, but how these changes translate to the cell and tissue levels remains unclear. We used computational models of human ventricular myocytes and tissues to predict and quantify the effects that de novo hERG mutations would have on cell and tissue electrophysiology. Mutations that decreased I(Kr) maximum conductance resulted in an increased cell and tissue action potential duration (APD) and a long QT interval on the electrocardiogram (ECG), whereas those that caused a positive shift in the inactivation curve resulted in a decreased APD and a short QT. Tissue vulnerability to re-entrant arrhythmias was correlated with transmural dispersion of repolarisation, and any change to this vulnerability could be inferred from the ECG QT interval or T wave peak-to-end time. Faster I(Kr) activation kinetics caused cell APD alternans to appear over a wider range of pacing rates and with a larger magnitude, and spatial heterogeneity in these cellular alternans resulted in discordant alternans at the tissue level. Thus, from channel kinetic data, we can predict the tissue-level electrophysiological effects of any hERG mutations and identify how the mutation would manifest clinically, as either a long or short QT syndrome with or without an increased risk of alternans and re-entrant arrhythmias.
Collapse
Affiliation(s)
- Alan P Benson
- Institute of Membrane & Systems Biology, and Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK.
| | | | | |
Collapse
|
43
|
Durdagi S, Duff HJ, Noskov SY. Combined Receptor and Ligand-Based Approach to the Universal Pharmacophore Model Development for Studies of Drug Blockade to the hERG1 Pore Domain. J Chem Inf Model 2011; 51:463-74. [DOI: 10.1021/ci100409y] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Serdar Durdagi
- Department of Biological Sciences, University of Calgary, Institute for Biocomplexity and Informatics Calgary, Alberta, Canada
| | - Henry J. Duff
- Faculty of Medicine, University of Calgary, Libin Cardiovascular Institute of Alberta, Calgary, Alberta, Canada
| | - Sergei Yu. Noskov
- Department of Biological Sciences, University of Calgary, Institute for Biocomplexity and Informatics Calgary, Alberta, Canada
| |
Collapse
|
44
|
Abstract
Inherited arrhythmia syndromes comprise an increasingly complex group of diseases involving mutations in multiple genes encoding ion channels, ion channel accessory subunits and channel interacting proteins, and various regulatory elements. These mutations serve to disrupt normal electrophysiology in the heart, leading to increased arrhythmogenic risk and death. These diseases have added impact as they often affect young people, sometimes without warning. Although originally thought to alter ion channel function, it is now increasingly recognized that mutations may alter ion channel protein and messenger RNA processing, to reduce the number of channels reaching the surface membrane. For many of these mutations, it is also known that several interventions may restore protein processing of mutant channels to increase their surface membrane expression toward normal. In this article, we reviewed inherited arrhythmia syndromes, focusing on long QT syndrome type 2, and discuss the complex biology of ion channel trafficking and pharmacological rescue of disease-causing mutant channels. Pharmacological rescue of misprocessed mutant channel proteins, or their transcripts providing appropriate small molecule drugs can be developed, has the potential for novel clinical therapies in some patients with inherited arrhythmia syndromes.
Collapse
|
45
|
Zhang XH, Jin MW, Sun HY, Zhang S, Li GR. The calmodulin inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalene sulphonamide directly blocks human ether à-go-go-related gene potassium channels stably expressed in human embryonic kidney 293 cells. Br J Pharmacol 2010; 161:872-84. [PMID: 20860665 DOI: 10.1111/j.1476-5381.2010.00916.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE N-(6-aminohexyl)-5-chloro-1-naphthalene sulphonamide (W-7) is a well-known calmodulin inhibitor used to study calmodulin regulation of intracellular Ca(2+) signalling-related process. Here, we have determined whether W-7 would inhibit human ether gene (hERG or K(v) 11.1) potassium channels, hK(v) 1.5 channels or hK(IR) 2.1 channels expressed in human embryonic kidney (HEK) 293 cells. EXPERIMENTAL APPROACH The hERG channel current, hK(v) 1.5 channel current or hK(IR) 2.1 channel current was recorded with a whole-cell patch clamp technique. KEY RESULTS It was found that the calmodulin inhibitor W-7 blocked hERG, hK(v) 1.5 and hK(IR) 2.1 channels. W-7 decreased the hERG current (I(hERG) ) in a concentration-dependent manner (IC(50) : 3.5 µM), and the inhibition was more significant at depolarization potentials between +10 and +60 mV. The hERG mutations in the S6 region Y652A and F656V, and in the pore helix S631A, had the IC(50) s of 5.5, 9.8 and 25.4 µM respectively. In addition, the compound inhibited hK(v) 1.5 and hK(IR) 2.1 channels with IC(50) s of 6.5 and 13.4 µM respectively. CONCLUSION AND IMPLICATIONS These results indicate that the calmodulin inhibitor W-7 exerts a direct channel-blocking effect on hERG, hK(v) 1.5 and hK(IR) 2.1 channels stably expressed in HEK 293 cells. Caution should be taken in the interpretation of calmodulin regulation of ion channels with W-7.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | |
Collapse
|
46
|
Subbotina J, Yarov-Yarovoy V, Lees-Miller J, Durdagi S, Guo J, Duff HJ, Noskov SY. Structural refinement of the hERG1 pore and voltage-sensing domains with ROSETTA-membrane and molecular dynamics simulations. Proteins 2010; 78:2922-34. [PMID: 20740484 PMCID: PMC2939218 DOI: 10.1002/prot.22815] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The hERG1 gene (Kv11.1) encodes a voltage-gated potassium channel. Mutations in this gene lead to one form of the Long QT Syndrome (LQTS) in humans. Promiscuous binding of drugs to hERG1 is known to alter the structure/function of the channel leading to an acquired form of the LQTS. Expectably, creation and validation of reliable 3D model of the channel have been a key target in molecular cardiology and pharmacology for the last decade. Although many models were built, they all were limited to pore domain. In this work, a full model of the hERG1 channel is developed which includes all transmembrane segments. We tested a template-driven de-novo design with ROSETTA-membrane modeling using side-chain placements optimized by subsequent molecular dynamics (MD) simulations. Although backbone templates for the homology modeled parts of the pore and voltage sensors were based on the available structures of KvAP, Kv1.2 and Kv1.2-Kv2.1 chimera channels, the missing parts are modeled de-novo. The impact of several alignments on the structure of the S4 helix in the voltage-sensing domain was also tested. Herein, final models are evaluated for consistency to the reported structural elements discovered mainly on the basis of mutagenesis and electrophysiology. These structural elements include salt bridges and close contacts in the voltage-sensor domain; and the topology of the extracellular S5-pore linker compared with that established by toxin foot-printing and nuclear magnetic resonance studies. Implications of the refined hERG1 model to binding of blockers and channels activators (potent new ligands for channel activations) are discussed.
Collapse
Affiliation(s)
- Julia Subbotina
- Institute for Biocomplexity and Informatics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | | | - James Lees-Miller
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Serdar Durdagi
- Institute for Biocomplexity and Informatics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Jiqing Guo
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Henry J. Duff
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sergei Yu. Noskov
- Institute for Biocomplexity and Informatics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
47
|
Hou L, Deo M, Furspan P, Pandit SV, Mironov S, Auerbach DS, Gong Q, Zhou Z, Berenfeld O, Jalife J. A major role for HERG in determining frequency of reentry in neonatal rat ventricular myocyte monolayer. Circ Res 2010; 107:1503-11. [PMID: 20947828 DOI: 10.1161/circresaha.110.232470] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE the rapid delayed rectifier potassium current, I(Kr), which flows through the human ether-a-go-go-related (hERG) channel, is a major determinant of the shape and duration of the human cardiac action potential (APD). However, it is unknown whether the time dependency of I(Kr) enables it to control APD, conduction velocity (CV), and wavelength (WL) at the exceedingly high activation frequencies that are relevant to cardiac reentry and fibrillation. OBJECTIVE to test the hypothesis that upregulation of hERG increases functional reentry frequency and contributes to its stability. METHODS AND RESULTS using optical mapping, we investigated the effects of I(Kr) upregulation on reentry frequency, APD, CV, and WL in neonatal rat ventricular myocyte (NRVM) monolayers infected with GFP (control), hERG (I(Kr)), or dominant negative mutant hERG G628S. Reentry frequency was higher in the I(Kr)-infected monolayers (21.12 ± 0.8 Hz; n=43 versus 9.21 ± 0.58 Hz; n=16; P<0.001) but slightly reduced in G628S-infected monolayers. APD(80) in the I(Kr)-infected monolayers was shorter (>50%) than control during pacing at 1 to 5 Hz. CV was similar in both groups at low frequency pacing. In contrast, during high-frequency reentry, the CV measured at varying distances from the center of rotation was significantly faster in I(Kr)-infected monolayers than controls. Simulations using a modified NRVM model predicted that rotor acceleration was attributable, in part, to a transient hyperpolarization immediately following the AP. The transient hyperpolarization was confirmed experimentally. CONCLUSIONS hERG overexpression dramatically accelerates reentry frequency in NRVM monolayers. Both APD and WL shortening, together with transient hyperpolarization, underlies the increased rotor frequency and stability.
Collapse
Affiliation(s)
- Luqia Hou
- Center for Arrhythmia Research, University of Michigan, 5025 Venture Dr, Ann Arbor, MI 48108, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gong Q, Stump MR, Dunn AR, Deng V, Zhou Z. Alternative splicing and polyadenylation contribute to the generation of hERG1 C-terminal isoforms. J Biol Chem 2010; 285:32233-41. [PMID: 20693282 DOI: 10.1074/jbc.m109.095695] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The human ether-a-go-go-related gene 1 (hERG1) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel. Several hERG1 isoforms with different N- and C-terminal ends have been identified. The hERG1a, hERG1b, and hERG1-3.1 isoforms contain the full-length C terminus, whereas the hERG1(USO) isoforms, hERG1a(USO) and hERG1b(USO), lack most of the C-terminal domain and contain a unique C-terminal end. The mechanisms underlying the generation of hERG1(USO) isoforms are not understood. We show that hERG1 isoforms with different C-terminal ends are generated by alternative splicing and polyadenylation of hERG1 pre-mRNA. We identified an intrinsically weak, noncanonical poly(A) signal, AGUAAA, within intron 9 of hERG1 that modulates the expression of hERG1a and hERG1a(USO). Replacing AGUAAA with the strong, canonical poly(A) signal AAUAAA resulted in the predominant production of hERG1a(USO) and a marked decrease in hERG1 current. In contrast, eliminating the intron 9 poly(A) signal or increasing the strength of 5' splice site led to the predominant production of hERG1a and a significant increase in hERG1 current. We found significant variation in the relative abundance of hERG1 C-terminal isoforms in different human tissues. Taken together, these findings suggest that post-transcriptional regulation of hERG1 pre-mRNA may represent a novel mechanism to modulate the expression and function of hERG1 channels.
Collapse
Affiliation(s)
- Qiuming Gong
- Division of Cardiovascular Medicine, Department of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | | | | | | | |
Collapse
|
49
|
Unger A, Jung E, Winklbaur B, Fischer G. Gender issues in the pharmacotherapy of opioid-addicted women: buprenorphine. J Addict Dis 2010; 29:217-30. [PMID: 20407978 DOI: 10.1080/10550881003684814] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gender, a biological determinant of mental health and illness, plays a critical role in determining patients' susceptibility, exposure to mental health risks, and related outcomes. Regarding sex differences in the epidemiology of opioid dependence, one third of the patients are women of childbearing age. Women have an earlier age of initiation of substance use and a more rapid progression to drug involvement and dependence than men. Generally few studies exist which focus on the special needs of women in opioid maintenance therapy. The aim of this paper is to provide an overview of treatment options for opioid-dependent women, with a special focus on buprenorphine, and to look at recent findings related to other factors that should be taken into consideration in optimizing the treatment of opioid-dependent women. Issues addressed include the role of gender in the choice of medication assisted treatment, sex differences in pharmacodynamics and pharmacokinetics of buprenorphine drug interactions, cardiac interactions, induction of buprenorphine in pregnant patients, the neonatal abstinence syndrome and breastfeeding. This paper aims to heighten the awareness for the need to take gender into consideration when making treatment decisions in an effort to optimize services and enhance the quality of life of women suffering from substance abuse.
Collapse
Affiliation(s)
- Annemarie Unger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
50
|
Stary A, Wacker SJ, Boukharta L, Zachariae U, Karimi-Nejad Y, Aqvist J, Vriend G, de Groot BL. Toward a consensus model of the HERG potassium channel. ChemMedChem 2010; 5:455-67. [PMID: 20104563 DOI: 10.1002/cmdc.200900461] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malfunction of hERG potassium channels, due to inherited mutations or inhibition by drugs, can cause long QT syndrome, which can lead to life-threatening arrhythmias. A three-dimensional structure of hERG is a prerequisite to understand the molecular basis of hERG malfunction. To achieve a consensus model, we carried out an extensive analysis of hERG models based on various alignments of helix S5. We analyzed seven models using a combination of conventional geometry/packing/normality validation methods as well as molecular dynamics simulations and molecular docking. A synthetic test set with the X-ray crystal structure of K(v)1.2 with artificially shifted S5 sequences modeled into the structure served as a reference case. We docked the known hERG inhibitors (+)-cisapride, (S)-terfenadine, and MK-499 into the hERG models and simulation snapshots. None of the single analyses unambiguously identified a preferred model, but the combination of all three revealed that there is only one model that fulfils all quality criteria. This model is confirmed by a recent mutation scanning experiment (P. Ju, G. Pages, R. P. Riek, P. C. Chen, A. M. Torres, P. S. Bansal, S. Kuyucak, P. W. Kuchel, J. I. Vandenberg, J. Biol. Chem. 2009, 284, 1000-1008). We expect the modeled structure to be useful as a basis both for computational studies of channel function and kinetics as well as the design of experiments.
Collapse
Affiliation(s)
- Anna Stary
- Computational Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|