1
|
Pinto-Anwandter BI, Bassetto CAZ, Latorre R, Bezanilla F. Energy landscape of a Kv channel revealed by temperature steps while perturbing its electromechanical coupling. Nat Commun 2025; 16:3379. [PMID: 40204695 PMCID: PMC11982254 DOI: 10.1038/s41467-025-58443-9] [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: 08/10/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Voltage-dependent potassium channels (Kv) play a crucial role in membrane repolarization during action potentials. They undergo voltage-dependent structural conformational transitions according to their distribution across their energy landscape. Understanding these transitions helps us comprehend their molecular function. Here, we used sudden and sustained temperature changes (Tstep) combined with different voltage protocols and mutations to dissect the energy landscape of the Shaker K+ channel. We used two mutations, ILT (V369I, I372L, and S376T) and I384N, that affect the coupling between the voltage sensor (VSD) and the pore domain (PD), to obtain the temperature dependence of VSD last transition and the intrinsic temperature dependence of the pore, respectively. Our findings support a loose or tight conformation of the electromechanical coupling. In the loose conformation, the movement of the VSD is necessary but not sufficient to efficiently propagate the electromechanical energy to open the pore. In contrast, this movement is effectively translated into pore opening in the tight conformation. Our results describe the energy landscape of the Shaker channel and how its temperature dependence can be modulated by affecting its electromechanical coupling.
Collapse
Affiliation(s)
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencias de Valparaiso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso, Chile.
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA.
- Centro Interdisciplinario de Neurociencias de Valparaiso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso, Chile.
| |
Collapse
|
2
|
Olov N, Nour S, Harris AR, Li D, Cook M, Williams RJ, Cheeseman S, Nisbet DR. Using Nanoscale Passports To Understand and Unlock Ion Channels as Gatekeepers of the Cell. ACS NANO 2024; 18:22709-22733. [PMID: 39136685 DOI: 10.1021/acsnano.4c05654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Natural ion channels are proteins embedded in the cell membrane that control many aspects of cell and human physiology by acting as gatekeepers, regulating the flow of ions in and out of cells. Advances in nanotechnology have influenced the methods for studying ion channels in vitro, as well as ways to unlock the delivery of therapeutics by modulating them in vivo. This review provides an overview of nanotechnology-enabled approaches for ion channel research with a focus on the synthesis and applications of synthetic ion channels. Further, the uses of nanotechnology for therapeutic applications are critically analyzed. Finally, we provide an outlook on the opportunities and challenges at the intersection of nanotechnology and ion channels. This work highlights the key role of nanoscale interactions in the operation and modulation of ion channels, which may prompt insights into nanotechnology-enabled mechanisms to study and exploit these systems in the near future.
Collapse
Affiliation(s)
- Nafiseh Olov
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Polymer Science Group, Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexander R Harris
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Dan Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark Cook
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Department of Medicine, St Vincent's Hospital, Melbourne, Fitzroy, VIC 3065, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Centre for Sustainable Bioproducts, Deakin University, Waurn Ponds, VIC 3217, Australia
- IMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3217, Australia
| | - Samuel Cheeseman
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - David R Nisbet
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| |
Collapse
|
3
|
Pinto-Anwandter BI, Bassetto CAZ, Latorre R, Bezanilla F. Turning a Kv channel into hot and cold receptor by perturbing its electromechanical coupling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607202. [PMID: 39149297 PMCID: PMC11326270 DOI: 10.1101/2024.08.08.607202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Voltage-dependent potassium channels (Kv) are extremely sensitive to membrane voltage and play a crucial role in membrane repolarization during action potentials. Kv channels undergo voltage-dependent transitions between closed states before opening. Despite all we have learned using electrophysiological methods and structural studies, we still lack a detailed picture of the energetics of the activation process. We show here that even a single mutation can drastically modify the temperature response of the Shaker Kv channel. Using rapid cell membrane temperature steps (Tsteps), we explored the effects of temperature on the ILT mutant (V369I, I372L, and S376T) and the I384N mutant. The ILT mutant produces a significant separation between the transitions of the voltage sensor domain (VSD) activation and the I384N uncouples its movement from the opening of the domain (PD). ILT and I384N respond to temperature in drastically different ways. In ILT, temperature facilitates the opening of the channel akin to a "hot" receptor, reflecting the temperature dependence of the voltage sensor's last transition and facilitating VSD to PD coupling (electromechanical coupling). In I384N, temperature stabilizes the channel closed configuration analogous to a "cold" receptor. Since I384N drastically uncouples the VSD from the pore opening, we reveal the intrinsic temperature dependence of the PD itself. Here, we propose that the electromechanical coupling has either a "loose" or "tight" conformation. In the loose conformation, the movement of the VSD is necessary but not sufficient to efficiently propagate the electromechanical energy to the S6 gate. In the tight conformation the VSD activation is more effectively translated into the opening of the PD. This conformational switch can be tuned by temperature and modifications of the S4 and S4-S5 linker. Our results show that we can modulate the temperature dependence of Kv channels by affecting its electromechanical coupling.
Collapse
Affiliation(s)
- Bernardo I Pinto-Anwandter
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- These authors contributed equally to this work
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- These authors contributed equally to this work
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencias de Valparaiso, Universidad de Valparaiso, Valparaiso, Chile
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- Centro Interdisciplinario de Neurociencias de Valparaiso, Universidad de Valparaiso, Valparaiso, Chile
| |
Collapse
|
4
|
Abramochkin DV, Pustovit OB, Mironov NY, Filatova TS, Nesterova T. Characterization of hERG K + channel inhibition by the new class III antiarrhythmic drug cavutilide. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5093-5104. [PMID: 38224347 DOI: 10.1007/s00210-023-02940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
Cavutilide (niferidil, refralon) is a new class III antiarrhythmic drug which effectively terminates persistent atrial fibrillation (AF; 84.6% of patients, mean AF duration 3 months) and demonstrates low risk of torsade de pointes (1.7%). ERG channels of rapid delayed rectifier current(IKr) are the primary target of cavutilide, but the particular reasons of higher effectiveness and lower proarrhythmic risk in comparison with other class III IKr blockers are unclear. The inhibition of hERG channels expressed in CHO-K1 cells by cavutilide was studied using whole-cell patch-clamp. The present study demonstrates high sensitivity of IhERG expressed in CHO-K1 cells to cavutilide (IC50 = 12.8 nM). Similarly to methanesulfonanilide class III agents, but unlike amiodarone and related drugs, cavutilide does not bind to hERG channels in their resting state. However, in contrast to dofetilide, cavutilide binds not only to opened, but also to inactivated channels. Moreover, at positive constantly set membrane potential (+ 60 mV) inhibition of IhERG by 100 nM cavutilide develops faster than at 0 mV and, especially, - 30 mV (τ of inhibition was 78.8, 103, and 153 ms, respectively). Thereby, cavutilide produces IhERG inhibition only when the cell is depolarized. During the same period of time, cavutilide produces greater block of IhERG when the cell is depolarized with 2 Hz frequency, if compared to 0.2 Hz. We suggest that, during the limited time after injection, cavutilide produces stronger inhibition of IKr in fibrillating atrium than in non-fibrillating ventricle. This leads to beneficial combination of antiarrhythmic effectiveness and low proarrhythmicity of cavutilide.
Collapse
Affiliation(s)
- Denis V Abramochkin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.
| | - Oksana B Pustovit
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| | | | - Tatiana S Filatova
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
- Chazov National Medical Research Center for Cardiology, Moscow, Russia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Tatiana Nesterova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049, Ekaterinburg, Russia
- Institute of Natural Sciences and Mathematics, Ural Federal University, 620075, Ekaterinburg, Russia
| |
Collapse
|
5
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Abramochkin D, Li B, Zhang H, Kravchuk E, Nesterova T, Glukhov G, Shestak A, Zaklyazminskaya E, Sokolova OS. Novel Gain-of-Function Mutation in the Kv11.1 Channel Found in the Patient with Brugada Syndrome and Mild QTc Shortening. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:543-552. [PMID: 38648771 DOI: 10.1134/s000629792403012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Brugada syndrome (BrS) is an inherited disease characterized by right precordial ST-segment elevation in the right precordial leads on electrocardiograms (ECG), and high risk of life-threatening ventricular arrhythmia and sudden cardiac death (SCD). Mutations in the responsible genes have not been fully characterized in the BrS patients, except for the SCN5A gene. We identified a new genetic variant, c.1189C>T (p.R397C), in the KCNH2 gene in the asymptomatic male proband diagnosed with BrS and mild QTc shortening. We hypothesize that this variant could alter IKr-current and may be causative for the rare non-SCN5A-related form of BrS. To assess its pathogenicity, we performed patch-clamp analysis on IKr reconstituted with this KCNH2 mutation in the Chinese hamster ovary cells and compared the phenotype with the wild type. It appeared that the R397C mutation does not affect the IKr density, but facilitates activation, hampers inactivation of the hERG channels, and increases magnitude of the window current suggesting that the p.R397C is a gain-of-function mutation. In silico modeling demonstrated that this missense mutation potentially leads to the shortening of action potential in the heart.
Collapse
Affiliation(s)
- Denis Abramochkin
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Bowen Li
- Shenzhen MSU-BIT University, Shenzhen, China.
| | - Han Zhang
- Shenzhen MSU-BIT University, Shenzhen, China.
| | | | - Tatiana Nesterova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, 620049, Russia.
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, 620075, Russia
| | - Grigory Glukhov
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Anna Shestak
- Petrovsky National Research Center of Surgery, Moscow, 119991, Russia.
| | | | - Olga S Sokolova
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| |
Collapse
|
7
|
Goineau S, Gallet L, Froget G. Whole-Cell Configuration of the Patch-Clamp Technique in the hERG Channel Assay. Curr Protoc 2024; 4:e959. [PMID: 38334240 DOI: 10.1002/cpz1.959] [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] [Indexed: 02/10/2024]
Abstract
In vitro electrophysiological safety studies have become an integral part of the drug development process because, in many instances, compound-induced QT prolongation has been associated with a direct block of human ether-a-go-go-related gene (hERG) potassium channels or their native current, the rapidly activating delayed rectifier potassium current (IKr ). Therefore, according to the ICH S7B guideline, the in vitro hERG channel patch-clamp assay is commonly used as an early screen to predict the ability of a compound to prolong the QT interval prior to first-in-human testing. The protocols described in this article are designed to assess the effects of acute or long-term exposure to new chemical entities on the amplitude of IKr in HEK293 cells stably transfected with the hERG channel (whole-cell configuration of the patch-clamp technique). Examples of results obtained with moxifloxacin, terfenadine, arsenic, pentamidine, erythromycin, and sotalol are provided for illustrative purposes. © 2024 Wiley Periodicals LLC. Basic Protocol: Measurement of the acute effects of test items in the hERG channel test Alternate Protocol: Measurement of the long-term effects of test items in the hERG channel test.
Collapse
Affiliation(s)
- Sonia Goineau
- Porsolt Research Center, Le Genest-Saint-Isle, France
| | - Lucie Gallet
- Porsolt Research Center, Le Genest-Saint-Isle, France
| | | |
Collapse
|
8
|
Ma JG, Vandenberg JI, Ng CA. Development of automated patch clamp assays to overcome the burden of variants of uncertain significance in inheritable arrhythmia syndromes. Front Physiol 2023; 14:1294741. [PMID: 38089476 PMCID: PMC10712320 DOI: 10.3389/fphys.2023.1294741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 10/16/2024] Open
Abstract
Advances in next-generation sequencing have been exceptionally valuable for identifying variants in medically actionable genes. However, for most missense variants there is insufficient evidence to permit definitive classification of variants as benign or pathogenic. To overcome the deluge of Variants of Uncertain Significance, there is an urgent need for high throughput functional assays to assist with the classification of variants. Advances in parallel planar patch clamp technologies has enabled the development of automated high throughput platforms capable of increasing throughput 10- to 100-fold compared to manual patch clamp methods. Automated patch clamp electrophysiology is poised to revolutionize the field of functional genomics for inheritable cardiac ion channelopathies. In this review, we outline i) the evolution of patch clamping, ii) the development of high-throughput automated patch clamp assays to assess cardiac ion channel variants, iii) clinical application of these assays and iv) where the field is heading.
Collapse
Affiliation(s)
- Joanne G. Ma
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I. Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Chai-Ann Ng
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
9
|
Meier S, Grundland A, Dobrev D, Volders PG, Heijman J. In silico analysis of the dynamic regulation of cardiac electrophysiology by K v 11.1 ion-channel trafficking. J Physiol 2023; 601:2711-2731. [PMID: 36752166 PMCID: PMC10313819 DOI: 10.1113/jp283976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiac electrophysiology is regulated by continuous trafficking and internalization of ion channels occurring over minutes to hours. Kv 11.1 (also known as hERG) underlies the rapidly activating delayed-rectifier K+ current (IKr ), which plays a major role in cardiac ventricular repolarization. Experimental characterization of the distinct temporal effects of genetic and acquired modulators on channel trafficking and gating is challenging. Computer models are instrumental in elucidating these effects, but no currently available model incorporates ion-channel trafficking. Here, we present a novel computational model that reproduces the experimentally observed production, forward trafficking, internalization, recycling and degradation of Kv 11.1 channels, as well as their modulation by temperature, pentamidine, dofetilide and extracellular K+ . The acute effects of these modulators on channel gating were also incorporated and integrated with the trafficking model in the O'Hara-Rudy human ventricular cardiomyocyte model. Supraphysiological dofetilide concentrations substantially increased Kv 11.1 membrane levels while also producing a significant channel block. However, clinically relevant concentrations did not affect trafficking. Similarly, severe hypokalaemia reduced Kv 11.1 membrane levels based on long-term culture data, but had limited effect based on short-term data. By contrast, clinically relevant elevations in temperature acutely increased IKr due to faster kinetics, while after 24 h, IKr was decreased due to reduced Kv 11.1 membrane levels. The opposite was true for lower temperatures. Taken together, our model reveals a complex temporal regulation of cardiac electrophysiology by temperature, hypokalaemia, and dofetilide through competing effects on channel gating and trafficking, and provides a framework for future studies assessing the role of impaired trafficking in cardiac arrhythmias. KEY POINTS: Kv 11.1 channels underlying the rapidly activating delayed-rectifier K+ current are important for ventricular repolarization and are continuously shuttled from the cytoplasm to the plasma membrane and back over minutes to hours. Kv 11.1 gating and trafficking are modulated by temperature, drugs and extracellular K+ concentration but experimental characterization of their combined effects is challenging. Computer models may facilitate these analyses, but no currently available model incorporates ion-channel trafficking. We introduce a new two-state ion-channel trafficking model able to reproduce a wide range of experimental data, along with the effects of modulators of Kv 11.1 channel functioning and trafficking. The model reveals complex dynamic regulation of ventricular repolarization by temperature, extracellular K+ concentration and dofetilide through opposing acute (millisecond) effects on Kv 11.1 gating and long-term (hours) modulation of Kv 11.1 trafficking. This in silico trafficking framework provides a tool to investigate the roles of acute and long-term processes on arrhythmia promotion and maintenance.
Collapse
Affiliation(s)
- Stefan Meier
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Adaïa Grundland
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Data Science and Knowledge Engineering, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Quebec, Canada
| | - Paul G.A. Volders
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
10
|
Usuda K, Hayashi K, Nakajima T, Kurata Y, Cui S, Kusayama T, Tsuda T, Tada H, Kato T, Sakata K, Usui S, Fujino N, Tanaka Y, Kaneko Y, Kurabayashi M, Tange S, Saito T, Ohta K, Yamagishi M, Takamura M. Mechanisms of fever-induced QT prolongation and torsades de pointes in patients with KCNH2 mutation. Europace 2023; 25:euad161. [PMID: 37386841 PMCID: PMC10310978 DOI: 10.1093/europace/euad161] [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: 11/24/2022] [Accepted: 05/13/2023] [Indexed: 07/01/2023] Open
Abstract
AIMS Patients with particular mutations of type-2 long QT syndrome (LQT2) are at an increased risk for malignant arrhythmia during fever. This study aimed to determine the mechanism by which KCNH2 mutations cause fever-induced QT prolongation and torsades de pointes (TdP). METHODS AND RESULTS We evaluated three KCNH2 mutations, G584S, D609G, and T613M, in the Kv11.1 S5-pore region, identified in patients with marked QT prolongation and TdP during fever. We also evaluated KCNH2 M124T and R269W, which are not associated with fever-induced QT prolongation. We characterized the temperature-dependent changes in the electrophysiological properties of the mutant Kv11.1 channels by patch-clamp recording and computer simulation. The average tail current densities (TCDs) at 35°C for G584S, WT+D609G, and WT+T613M were significantly smaller and less increased with rising temperature from 35°C to 40°C than those for WT, M124T, and R269W. The ratios of the TCDs at 40°C to 35°C for G584S, WT+D609G, and WT+T613M were significantly smaller than for WT, M124T, and R269W. The voltage dependence of the steady-state inactivation curve for WT, M124T, and R269W showed a significant positive shift with increasing temperature; however, that for G584S, WT+D609G, and WT+T613M showed no significant change. Computer simulation demonstrated that G584S, WT+D609G, and WT+T613M caused prolonged action potential durations and early afterdepolarization formation at 40°C. CONCLUSION These findings indicate that KCNH2 G584S, D609G, and T613M in the S5-pore region reduce the temperature-dependent increase in TCDs through an enhanced inactivation, resulting in QT prolongation and TdP at a febrile state in patients with LQT2.
Collapse
Affiliation(s)
- Keisuke Usuda
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Tadashi Nakajima
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasutaka Kurata
- Department of Physiology, Kanazawa Medical University, Uchinada, Japan
| | - Shihe Cui
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Takashi Kusayama
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Toyonobu Tsuda
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Takeshi Kato
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Soichiro Usui
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Noboru Fujino
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| | - Yoshihiro Tanaka
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
- Department of Preventive Medicine Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yoshiaki Kaneko
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shoichi Tange
- Department of Cardiovascular Medicine, Maebashi Red Cross Hospital, Maebashi, Japan
| | - Takekatsu Saito
- Department of Pediatrics, Kanazawa University, Kanazawa, Japan
| | - Kunio Ohta
- Department of Pediatrics, Kanazawa University, Kanazawa, Japan
| | | | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1, Takara-machi, Kanazawa Ishikawa 920-8641, Japan
| |
Collapse
|
11
|
Alameh M, Oliveira-Mendes BR, Kyndt F, Rivron J, Denjoy I, Lesage F, Schott JJ, De Waard M, Loussouarn G. A need for exhaustive and standardized characterization of ion channels activity. The case of K V11.1. Front Physiol 2023; 14:1132533. [PMID: 36860515 PMCID: PMC9968853 DOI: 10.3389/fphys.2023.1132533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
hERG, the pore-forming subunit of the rapid component of the delayed rectifier K+ current, plays a key role in ventricular repolarization. Mutations in the KCNH2 gene encoding hERG are associated with several cardiac rhythmic disorders, mainly the Long QT syndrome (LQTS) characterized by prolonged ventricular repolarization, leading to ventricular tachyarrhythmias, sometimes progressing to ventricular fibrillation and sudden death. Over the past few years, the emergence of next-generation sequencing has revealed an increasing number of genetic variants including KCNH2 variants. However, the potential pathogenicity of the majority of the variants remains unknown, thus classifying them as variants of uncertain significance or VUS. With diseases such as LQTS being associated with sudden death, identifying patients at risk by determining the variant pathogenicity, is crucial. The purpose of this review is to describe, on the basis of an exhaustive examination of the 1322 missense variants, the nature of the functional assays undertaken so far and their limitations. A detailed analysis of 38 hERG missense variants identified in Long QT French patients and studied in electrophysiology also underlies the incomplete characterization of the biophysical properties for each variant. These analyses lead to two conclusions: first, the function of many hERG variants has never been looked at and, second, the functional studies done so far are excessively heterogeneous regarding the stimulation protocols, cellular models, experimental temperatures, homozygous and/or the heterozygous condition under study, a context that may lead to conflicting conclusions. The state of the literature emphasizes how necessary and important it is to perform an exhaustive functional characterization of hERG variants and to standardize this effort for meaningful comparison among variants. The review ends with suggestions to create a unique homogeneous protocol that could be shared and adopted among scientists and that would facilitate cardiologists and geneticists in patient counseling and management.
Collapse
Affiliation(s)
- Malak Alameh
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Barbara Ribeiro Oliveira-Mendes
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,*Correspondence: Barbara Ribeiro Oliveira-Mendes,
| | - Florence Kyndt
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Jordan Rivron
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Isabelle Denjoy
- Service de Cardiologie et CNMR Maladies Cardiaques Héréditaires Rares, Hôpital Bichat, Paris, France
| | - Florian Lesage
- Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Jean-Jacques Schott
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Michel De Waard
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Gildas Loussouarn
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| |
Collapse
|
12
|
SGLT2 Inhibitor Empagliflozin Modulates Ion Channels in Adult Zebrafish Heart. Int J Mol Sci 2022; 23:ijms23179559. [PMID: 36076956 PMCID: PMC9455557 DOI: 10.3390/ijms23179559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Empagliflozin, an inhibitor of sodium-glucose co-transporter 2 (iSGLT2), improves cardiovascular outcomes in patients with and without diabetes and possesses an antiarrhythmic activity. However, the mechanisms of these protective effects have not been fully elucidated. This study aimed to explore the impact of empagliflozin on ion channel activity and electrophysiological characteristics in the ventricular myocardium. The main cardiac ionic currents (INa, ICaL, ICaT, IKr, IKs) and action potentials (APs) were studied in zebrafish. Whole-cell currents were measured using the patch clamp method in the isolated ventricular cardiomyocytes. The conventional sharp glass microelectrode technique was applied for the recording of APs from the ventricular myocardium of the excised heart. Empagliflozin pretreatment compared to the control group enhanced potassium IKr step current density in the range of testing potentials from 0 to +30 mV, IKr tail current density in the range of testing potentials from +10 to +70 mV, and IKs current density in the range of testing potentials from −10 to +20 mV. Moreover, in the ventricular myocardium, empagliflozin pretreatment shortened AP duration APD as shown by reduced APD50 and APD90. Empagliflozin had no influence on sodium (INa) and L- and T-type calcium currents (ICaL and ICaT) in zebrafish ventricular cardiomyocytes. Thus, we conclude that empagliflozin increases the rapid and slow components of delayed rectifier K+ current (IKr and IKs). This mechanism could be favorable for cardiac protection.
Collapse
|
13
|
Al Salmani MK, Tavakoli R, Zaman W, Al Harrasi A. Multiple mechanisms underlie reduced potassium conductance in the p.T1019PfsX38 variant of hERG. Physiol Rep 2022; 10:e15341. [PMID: 35854468 PMCID: PMC9296870 DOI: 10.14814/phy2.15341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
Long QT syndrome type II (LQT2) is caused by loss-of-function mutations in the hERG K+ channel, leading to increased incidence of cardiac arrest and sudden death. Many genetic variants have been reported in the hERG gene with various consequences on channel expression, permeation, and gating. Only a small number of LQT2 causing variants has been characterized to define the underlying pathophysiological causes of the disease. We sought to determine the characteristics of the frameshift variant p.Thr1019ProfsX38 (T1019PfsX38) which affects the C-terminus of the protein. This mutation was identified in an extended Omani family of LQT2. It replaces the last 140 amino acids of hERG with 37 unique amino acids. T1019 is positioned at a distinguished region of the C-terminal tail of hERG, as predicted from the deep learning system AlphaFold v2.0. We employed the whole-cell configuration of the patch-clamp technique to study wild-type and mutant channels that were transiently expressed in human embryonic kidney 293 (HEK293) cells. Depolarizing voltages elicited slowly deactivating tail currents that appeared upon repolarization of cells that express either wild-type- or T1019PfsX38-hERG. There were no differences in the voltage and time dependencies of activation between the two variants. However, the rates of hERG channel deactivation at hyperpolarizing potentials were accelerated by T1019PfsX38. In addition, the voltage dependence of inactivation of T1019PfsX38-hERG was shifted by 20 mV in the negative direction when compared with wild-type hERG. The rates of channel inactivation were increased in the mutant channel variant. Next, we employed a step-ramp protocol to mimic membrane repolarization by the cardiac action potential. The amplitudes of outward currents and their integrals were reduced in the mutant variant when compared with the wild-type variant during repolarization. Thus, changes in the gating dynamics of hERG by the T1019PfsX38 variant contribute to the pathology seen in affected LQT2 patients.
Collapse
Affiliation(s)
| | - Rezvan Tavakoli
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| | - Wajid Zaman
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| | - Ahmed Al Harrasi
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| |
Collapse
|
14
|
Translating the measurement of hERG kinetics and drug block for CiPA to a high throughput platform. J Pharmacol Toxicol Methods 2022; 117:107192. [PMID: 35750310 DOI: 10.1016/j.vascn.2022.107192] [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: 03/09/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/23/2022]
Abstract
The Comprehensive in vitro Proarrhythmic Assay (CiPA) has promoted use of in silico models of drug effects on cardiac repolarization to improve proarrhythmic risk prediction. These models contain a pharmacodynamic component describing drug binding to hERG channels that required in vitro data for kinetics of block, in addition to potency, to constrain them. To date, development and validation has been undertaken using data from manual patch-clamp. The application of this approach at scale requires the development of a high-throughput, automated patch-clamp (APC) implementation. Here, we present a comprehensive analysis of the implementation of the Milnes, or CiPA dynamic protocol, on an APC platform, including quality control and data analysis. Kinetics and potency of block were assessed for bepridil, cisapride, terfenadine and verapamil with data retention/QC pass rate of 21.8% overall, or as high as 50.4% when only appropriate sweep lengths were considered for drugs with faster kinetics. The variability in IC50 and kinetics between manual and APC was comparable to that seen between sites/platforms in previous APC studies of potency. Whilst the experimental success is less than observed in screens of potency alone, it is still significantly greater than manual patch. With the modifications to protocol design, including sweep length, number of repetitions, and leak correction recommended in this study, this protocol can be applied on APC to acquire data comparable to manual patch clamp.
Collapse
|
15
|
Valentin JP, Hoffmann P, Ortemann-Renon C, Koerner J, Pierson J, Gintant G, Willard J, Garnett C, Skinner M, Vargas HM, Wisialowski T, Pugsley MK. The Challenges of Predicting Drug-Induced QTc Prolongation in Humans. Toxicol Sci 2022; 187:3-24. [PMID: 35148401 PMCID: PMC9041548 DOI: 10.1093/toxsci/kfac013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The content of this article derives from a Health and Environmental Sciences Institute (HESI) consortium with a focus to improve cardiac safety during drug development. A detailed literature review was conducted to evaluate the concordance between nonclinical repolarization assays and the clinical thorough QT (TQT) study. Food and Drug Administration and HESI developed a joint database of nonclinical and clinical data, and a retrospective analysis of 150 anonymized drug candidates was reviewed to compare the performance of 3 standard nonclinical assays with clinical TQT study findings as well as investigate mechanism(s) potentially responsible for apparent discrepancies identified. The nonclinical assays were functional (IKr) current block (Human ether-a-go-go related gene), action potential duration, and corrected QT interval in animals (in vivo corrected QT). Although these nonclinical assays demonstrated good specificity for predicting negative clinical QT prolongation, they had relatively poor sensitivity for predicting positive clinical QT prolongation. After review, 28 discordant TQT-positive drugs were identified. This article provides an overview of direct and indirect mechanisms responsible for QT prolongation and theoretical reasons for lack of concordance between clinical TQT studies and nonclinical assays. We examine 6 specific and discordant TQT-positive drugs as case examples. These were derived from the unique HESI/Food and Drug Administration database. We would like to emphasize some reasons for discordant data including, insufficient or inadequate nonclinical data, effects of the drug on other cardiac ion channels, and indirect and/or nonelectrophysiological effects of drugs, including altered heart rate. We also outline best practices that were developed based upon our evaluation.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- Department of Investigative Toxicology, UCB Biopharma SRL, Braine-l’Alleud B-1420, Belgium
| | | | | | - John Koerner
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005, USA
| | | | - James Willard
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Christine Garnett
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | | | - Hugo M Vargas
- Department of Safety Pharmacology & Animal Research Center, Amgen, Thousand Oaks, California 91320, USA
| | - Todd Wisialowski
- Department of Safety Pharmacology, Pfizer, Groton, Connecticut 06340, USA
| | - Michael K Pugsley
- Department of Toxicology, Cytokinetics, South San Francisco, California 94080, USA
| |
Collapse
|
16
|
TeBay C, Hill AP, Windley MJ. Metabolic and electrolyte abnormalities as risk factors in drug-induced long QT syndrome. Biophys Rev 2022; 14:353-367. [PMID: 35103080 PMCID: PMC8792523 DOI: 10.1007/s12551-022-00929-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
Drug-induced long QT syndrome (diLQTS) is the phenomenon by which the administration of drugs causes prolongation of cardiac repolarisation and leads to an increased risk of the ventricular tachycardia known as torsades de pointes (TdP). In most cases of diLQTS, the primary molecular target is the human ether-à-go-go-related gene protein (hERG) potassium channel, which carries the rapid delayed rectifier current (IKr) in the heart. However, the proarrhythmic risk associated with drugs that block hERG can be modified in patients by a range of environmental- and disease-related factors, such as febrile temperatures, alterations in pH, dyselectrolytaemias such as hypokalaemia and hypomagnesemia and coadministration with other drugs. In this review, we will discuss the clinical occurrence of drug-induced LQTS in the context of these modifying factors as well as the mechanisms by which they contribute to altered hERG potency and proarrhythmic risk.
Collapse
Affiliation(s)
- Clifford TeBay
- Victor Chang Cardiac Research Institute, 405, Liverpool street, Darlinghurst, Sydney, NSW 2020 Australia
| | - Adam P. Hill
- Victor Chang Cardiac Research Institute, 405, Liverpool street, Darlinghurst, Sydney, NSW 2020 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Sydney, Australia
| | - Monique J. Windley
- Victor Chang Cardiac Research Institute, 405, Liverpool street, Darlinghurst, Sydney, NSW 2020 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Sydney, Australia
| |
Collapse
|
17
|
Oliveira‐Mendes B, Feliciangeli S, Ménard M, Chatelain F, Alameh M, Montnach J, Nicolas S, Ollivier B, Barc J, Baró I, Schott J, Probst V, Kyndt F, Denjoy I, Lesage F, Loussouarn G, De Waard M. A standardised hERG phenotyping pipeline to evaluate KCNH2 genetic variant pathogenicity. Clin Transl Med 2021; 11:e609. [PMID: 34841674 PMCID: PMC8609418 DOI: 10.1002/ctm2.609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIMS Mutations in KCNH2 cause long or short QT syndromes (LQTS or SQTS) predisposing to life-threatening arrhythmias. Over 1000 hERG variants have been described by clinicians, but most remain to be characterised. The objective is to standardise and accelerate the phenotyping process to contribute to clinician diagnosis and patient counselling. In silico evaluation was also included to characterise the structural impact of the variants. METHODS We selected 11 variants from known LQTS patients and two variants for which diagnosis was problematic. Using the Gibson assembly strategy, we efficiently introduced mutations in hERG cDNA despite GC-rich sequences. A pH-sensitive fluorescent tag was fused to hERG for efficient evaluation of channel trafficking. An optimised 35-s patch-clamp protocol was developed to evaluate hERG channel activity in transfected cells. R software was used to speed up analyses. RESULTS In the present work, we observed a good correlation between cell surface expression, assessed by the pH-sensitive tag, and current densities. Also, we showed that the new biophysical protocol allows a significant gain of time in recording ion channel properties and provides extensive information on WT and variant channel biophysical parameters, that can all be recapitulated in a single parameter defined herein as the repolarisation power. The impacts of the variants on channel structure were also reported where structural information was available. These three readouts (trafficking, repolarisation power and structural impact) define three pathogenicity indexes that may help clinical diagnosis. CONCLUSIONS Fast-track characterisation of KCNH2 genetic variants shows its relevance to discriminate mutants that affect hERG channel activity from variants with undetectable effects. It also helped the diagnosis of two new variants. This information is meant to fill a patient database, as a basis for personalised medicine. The next steps will be to further accelerate the process using an automated patch-clamp system.
Collapse
Affiliation(s)
| | - Sylvain Feliciangeli
- Labex ICST, Université Côte d'Azur, INSERMCentre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et CellulaireValbonneFrance
| | - Mélissa Ménard
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| | - Frank Chatelain
- Labex ICST, Université Côte d'Azur, INSERMCentre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et CellulaireValbonneFrance
| | - Malak Alameh
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| | - Jérôme Montnach
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| | | | | | - Julien Barc
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| | - Isabelle Baró
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| | | | - Vincent Probst
- CHU Nantes, l'Institut du Thorax, INSERM, CNRSUNIV NantesNantesFrance
| | - Florence Kyndt
- CHU Nantes, l'Institut du Thorax, INSERM, CNRSUNIV NantesNantesFrance
| | - Isabelle Denjoy
- Service de Cardiologie et CNMR Maladies Cardiaques Héréditaires RaresHôpital BichatParisFrance
| | - Florian Lesage
- Labex ICST, Université Côte d'Azur, INSERMCentre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et CellulaireValbonneFrance
| | | | - Michel De Waard
- l'Institut du ThoraxInserm UMR 1087/CNRS UMR 6291NantesFrance
| |
Collapse
|
18
|
Hu W, Clark RB, Giles WR, Shibata E, Zhang H. Physiological Roles of the Rapidly Activated Delayed Rectifier K + Current in Adult Mouse Heart Primary Pacemaker Activity. Int J Mol Sci 2021; 22:4761. [PMID: 33946248 PMCID: PMC8124469 DOI: 10.3390/ijms22094761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Robust, spontaneous pacemaker activity originating in the sinoatrial node (SAN) of the heart is essential for cardiovascular function. Anatomical, electrophysiological, and molecular methods as well as mathematical modeling approaches have quite thoroughly characterized the transmembrane fluxes of Na+, K+ and Ca2+ that produce SAN action potentials (AP) and 'pacemaker depolarizations' in a number of different in vitro adult mammalian heart preparations. Possible ionic mechanisms that are responsible for SAN primary pacemaker activity are described in terms of: (i) a Ca2+-regulated mechanism based on a requirement for phasic release of Ca2+ from intracellular stores and activation of an inward current-mediated by Na+/Ca2+ exchange; (ii) time- and voltage-dependent activation of Na+ or Ca2+ currents, as well as a cyclic nucleotide-activated current, If; and/or (iii) a combination of (i) and (ii). Electrophysiological studies of single spontaneously active SAN myocytes in both adult mouse and rabbit hearts consistently reveal significant expression of a rapidly activating time- and voltage-dependent K+ current, often denoted IKr, that is selectively expressed in the leading or primary pacemaker region of the adult mouse SAN. The main goal of the present study was to examine by combined experimental and simulation approaches the functional or physiological roles of this K+ current in the pacemaker activity. Our patch clamp data of mouse SAN myocytes on the effects of a pharmacological blocker, E4031, revealed that a rapidly activating K+ current is essential for action potential (AP) repolarization, and its deactivation during the pacemaker potential contributes a small but significant component to the pacemaker depolarization. Mathematical simulations using a murine SAN AP model confirm that well known biophysical properties of a delayed rectifier K+ current can contribute to its role in generating spontaneous myogenic activity.
Collapse
Affiliation(s)
- Wei Hu
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| | - Robert B. Clark
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Wayne R. Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Erwin Shibata
- Department of Physiology, Carver School of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
19
|
Ford JB, Ganguly M, Zhuo J, McPheeters MT, Jenkins MW, Chiel HJ, Jansen ED. Optimizing thermal block length during infrared neural inhibition to minimize temperature thresholds. J Neural Eng 2021; 18:10.1088/1741-2552/abf00d. [PMID: 33735846 PMCID: PMC11189657 DOI: 10.1088/1741-2552/abf00d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/18/2021] [Indexed: 11/11/2022]
Abstract
Objective. Infrared neural inhibition (INI) is a method of blocking the generation or propagation of neural action potentials through laser heating with wavelengths strongly absorbed by water. Recent work has identified that the distance heated along axons, the block length (BL), modulates the temperature needed for inhibition; however, this relationship has not been characterized. This study explores how BL during INI can be optimized towards minimizing its temperature threshold.Approach. To understand the relationship between BL and the temperature required for INI, excised nerves fromAplysia californicawere laser-heated over different lengths of axon during electrical stimulation of compound action potentials. INI was provided by irradiation (λ= 1470 nm) from a custom probe (n= 6 nerves), and subsequent validation was performed by providing heat block using perfused hot media over nerves (n= 5 nerves).Main Results. Two BL regimes were identified. Short BLs (thermal full width at half maximum (tFWHM) = 0.81-1.13 mm) demonstrated that increasing the tFWHM resulted in lower temperature thresholds for INI (p< 0.0125), while longer BLs (tFWHM = 1.13-3.03 mm) showed no significant change between the temperature threshold and tFWHM (p> 0.0125). Validation of this longer regime was performed using perfused hot media over different lengths of nerves. This secondary heating method similarly showed no significant change (p> 0.025) in the temperature threshold (tFWHM = 1.25-4.42 mm).Significance. This work characterized how the temperature threshold for neural heat block varies with BL and identified an optimal BL around tFWHM = 1.13 mm which minimizes both the maximum temperature applied to tissue and the volume of tissue heated during INI. Understanding how to optimally target lengths of nerve to minimize temperature during INI can help inform the design of devices for longitudinal animal studies and human implementation.
Collapse
Affiliation(s)
- Jeremy B Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Biophotonics Center, Vanderbilt University, Nashville, TN, United States of America
| | - Mohit Ganguly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Biophotonics Center, Vanderbilt University, Nashville, TN, United States of America
| | - Junqi Zhuo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Matthew T McPheeters
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Hillel J Chiel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, United States of America
| | - E Duco Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Biophotonics Center, Vanderbilt University, Nashville, TN, United States of America
- Department of Neurological Surgery, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
20
|
Bai J, Zhu Y, Lo A, Gao M, Lu Y, Zhao J, Zhang H. In Silico Assessment of Class I Antiarrhythmic Drug Effects on Pitx2-Induced Atrial Fibrillation: Insights from Populations of Electrophysiological Models of Human Atrial Cells and Tissues. Int J Mol Sci 2021; 22:1265. [PMID: 33514068 PMCID: PMC7866025 DOI: 10.3390/ijms22031265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Electrical remodelling as a result of homeodomain transcription factor 2 (Pitx2)-dependent gene regulation was linked to atrial fibrillation (AF) and AF patients with single nucleotide polymorphisms at chromosome 4q25 responded favorably to class I antiarrhythmic drugs (AADs). The possible reasons behind this remain elusive. The purpose of this study was to assess the efficacy of the AADs disopyramide, quinidine, and propafenone on human atrial arrhythmias mediated by Pitx2-induced remodelling, from a single cell to the tissue level, using drug binding models with multi-channel pharmacology. Experimentally calibrated populations of human atrial action po-tential (AP) models in both sinus rhythm (SR) and Pitx2-induced AF conditions were constructed by using two distinct models to represent morphological subtypes of AP. Multi-channel pharmaco-logical effects of disopyramide, quinidine, and propafenone on ionic currents were considered. Simulated results showed that Pitx2-induced remodelling increased maximum upstroke velocity (dVdtmax), and decreased AP duration (APD), conduction velocity (CV), and wavelength (WL). At the concentrations tested in this study, these AADs decreased dVdtmax and CV and prolonged APD in the setting of Pitx2-induced AF. Our findings of alterations in WL indicated that disopyramide may be more effective against Pitx2-induced AF than propafenone and quinidine by prolonging WL.
Collapse
Affiliation(s)
- Jieyun Bai
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Yijie Zhu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Andy Lo
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand; (A.L.); (J.Z.)
| | - Meng Gao
- Department of Computer Science and Technology, College of Electrical Engineering and Information, Northeast Agricultural University, Harbin 150030, China
| | - Yaosheng Lu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Jichao Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand; (A.L.); (J.Z.)
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| |
Collapse
|
21
|
DeMaegd ML, Stein W. Temperature-robust activity patterns arise from coordinated axonal Sodium channel properties. PLoS Comput Biol 2020; 16:e1008057. [PMID: 32716930 PMCID: PMC7410338 DOI: 10.1371/journal.pcbi.1008057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 08/06/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
Action potentials are a key component of neuronal communication and their precise timing is critical for processes like learning, memory, and complex behaviors. Action potentials propagate through long axons to their postsynaptic partners, which requires axons not only to faithfully transfer action potentials to distant synaptic regions but also to maintain their timing. This is particularly challenging when axons differ in their morphological and physiological properties, as timing is predicted to diverge between these axons when extrinsic conditions change. It is unknown if and how diverse axons maintain timing during temperature changes that animals and humans encounter. We studied whether ambient temperature changes cause different timing in the periphery of neurons that centrally produce temperature-robust activity. In an approach combining modeling, imaging, and electrophysiology, we explored mechanisms that support timing by exposing the axons of three different neuron types from the same crustacean (Cancer borealis) motor circuit and involved in the same functional task to a range of physiological temperatures. We show that despite substantial differences between axons, the effects of temperature on action potential propagation were moderate and supported temperature-robust timing over long-distances. Our modeling demonstrates that to maintain timing, the underlying channel properties of these axons do not need to be temperature-insensitive or highly restricted, but coordinating the temperature sensitivities of the Sodium activation gate time constant and the maximum Sodium conductance is required. Thus, even highly temperature-sensitive ion channel properties can support temperature-robust timing between distinct neuronal types and across long distances.
Collapse
Affiliation(s)
- Margaret L. DeMaegd
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Wolfgang Stein
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| |
Collapse
|
22
|
The environmental toxicant ziram enhances neurotransmitter release and increases neuronal excitability via the EAG family of potassium channels. Neurobiol Dis 2020; 143:104977. [PMID: 32553709 DOI: 10.1016/j.nbd.2020.104977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 12/21/2022] Open
Abstract
Environmental toxicants have the potential to contribute to the pathophysiology of multiple complex diseases, but the underlying mechanisms remain obscure. One such toxicant is the widely used fungicide ziram, a dithiocarbamate known to have neurotoxic effects and to increase the risk of Parkinson's disease. We have used Drosophila melanogaster as an unbiased discovery tool to identify novel molecular pathways by which ziram may disrupt neuronal function. Consistent with previous results in mammalian cells, we find that ziram increases the probability of synaptic vesicle release by dysregulation of the ubiquitin signaling system. In addition, we find that ziram increases neuronal excitability. Using a combination of live imaging and electrophysiology, we find that ziram increases excitability in both aminergic and glutamatergic neurons. This increased excitability is phenocopied and occluded by null mutant animals of the ether a-go-go (eag) potassium channel. A pharmacological inhibitor of the temperature sensitive hERG (human ether-a-go-go related gene) phenocopies the excitability effects of ziram but only at elevated temperatures. seizure (sei), a fly ortholog of hERG, is thus another candidate target of ziram. Taken together, the eag family of potassium channels emerges as a candidate for mediating some of the toxic effects of ziram. We propose that ziram may contribute to the risk of complex human diseases by blockade of human eag and sei orthologs, such as hERG.
Collapse
|
23
|
Lei CL, Clerx M, Whittaker DG, Gavaghan DJ, de Boer TP, Mirams GR. Accounting for variability in ion current recordings using a mathematical model of artefacts in voltage-clamp experiments. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2020; 378:20190348. [PMID: 32448060 PMCID: PMC7287334 DOI: 10.1098/rsta.2019.0348] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/08/2020] [Indexed: 05/21/2023]
Abstract
Mathematical models of ion channels, which constitute indispensable components of action potential models, are commonly constructed by fitting to whole-cell patch-clamp data. In a previous study, we fitted cell-specific models to hERG1a (Kv11.1) recordings simultaneously measured using an automated high-throughput system, and studied cell-cell variability by inspecting the resulting model parameters. However, the origin of the observed variability was not identified. Here, we study the source of variability by constructing a model that describes not just ion current dynamics, but the entire voltage-clamp experiment. The experimental artefact components of the model include: series resistance, membrane and pipette capacitance, voltage offsets, imperfect compensations made by the amplifier for these phenomena, and leak current. In this model, variability in the observations can be explained by either cell properties, measurement artefacts, or both. Remarkably, by assuming that variability arises exclusively from measurement artefacts, it is possible to explain a larger amount of the observed variability than when assuming cell-specific ion current kinetics. This assumption also leads to a smaller number of model parameters. This result suggests that most of the observed variability in patch-clamp data measured under the same conditions is caused by experimental artefacts, and hence can be compensated for in post-processing by using our model for the patch-clamp experiment. This study has implications for the question of the extent to which cell-cell variability in ion channel kinetics exists, and opens up routes for better correction of artefacts in patch-clamp data. This article is part of the theme issue 'Uncertainty quantification in cardiac and cardiovascular modelling and simulation'.
Collapse
Affiliation(s)
- Chon Lok Lei
- Computational Biology & Health Informatics, Department of Computer Science, University of Oxford, Oxford, UK
| | - Michael Clerx
- Computational Biology & Health Informatics, Department of Computer Science, University of Oxford, Oxford, UK
| | - Dominic G. Whittaker
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - David J. Gavaghan
- Computational Biology & Health Informatics, Department of Computer Science, University of Oxford, Oxford, UK
| | - Teun P. de Boer
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
- e-mail:
| |
Collapse
|
24
|
Brewer KR, Kuenze G, Vanoye CG, George AL, Meiler J, Sanders CR. Structures Illuminate Cardiac Ion Channel Functions in Health and in Long QT Syndrome. Front Pharmacol 2020; 11:550. [PMID: 32431610 PMCID: PMC7212895 DOI: 10.3389/fphar.2020.00550] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
The cardiac action potential is critical to the production of a synchronized heartbeat. This electrical impulse is governed by the intricate activity of cardiac ion channels, among them the cardiac voltage-gated potassium (Kv) channels KCNQ1 and hERG as well as the voltage-gated sodium (Nav) channel encoded by SCN5A. Each channel performs a highly distinct function, despite sharing a common topology and structural components. These three channels are also the primary proteins mutated in congenital long QT syndrome (LQTS), a genetic condition that predisposes to cardiac arrhythmia and sudden cardiac death due to impaired repolarization of the action potential and has a particular proclivity for reentrant ventricular arrhythmias. Recent cryo-electron microscopy structures of human KCNQ1 and hERG, along with the rat homolog of SCN5A and other mammalian sodium channels, provide atomic-level insight into the structure and function of these proteins that advance our understanding of their distinct functions in the cardiac action potential, as well as the molecular basis of LQTS. In this review, the gating, regulation, LQTS mechanisms, and pharmacological properties of KCNQ1, hERG, and SCN5A are discussed in light of these recent structural findings.
Collapse
Affiliation(s)
- Kathryn R. Brewer
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| | - Georg Kuenze
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Carlos G. Vanoye
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alfred L. George
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Charles R. Sanders
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
25
|
Sutanto H, Laudy L, Clerx M, Dobrev D, Crijns HJ, Heijman J. Maastricht antiarrhythmic drug evaluator (MANTA): A computational tool for better understanding of antiarrhythmic drugs. Pharmacol Res 2019; 148:104444. [DOI: 10.1016/j.phrs.2019.104444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/10/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
|
26
|
Lei CL, Clerx M, Beattie KA, Melgari D, Hancox JC, Gavaghan DJ, Polonchuk L, Wang K, Mirams GR. Rapid Characterization of hERG Channel Kinetics II: Temperature Dependence. Biophys J 2019; 117:2455-2470. [PMID: 31451180 PMCID: PMC6990152 DOI: 10.1016/j.bpj.2019.07.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/20/2019] [Accepted: 07/17/2019] [Indexed: 11/29/2022] Open
Abstract
Ion channel behavior can depend strongly on temperature, with faster kinetics at physiological temperatures leading to considerable changes in currents relative to room temperature. These temperature-dependent changes in voltage-dependent ion channel kinetics (rates of opening, closing, inactivating, and recovery) are commonly represented with Q10 coefficients or an Eyring relationship. In this article, we assess the validity of these representations by characterizing channel kinetics at multiple temperatures. We focus on the human Ether-à-go-go-Related Gene (hERG) channel, which is important in drug safety assessment and commonly screened at room temperature so that results require extrapolation to physiological temperature. In Part I of this study, we established a reliable method for high-throughput characterization of hERG1a (Kv11.1) kinetics, using a 15-second information-rich optimized protocol. In this Part II, we use this protocol to study the temperature dependence of hERG kinetics using Chinese hamster ovary cells overexpressing hERG1a on the Nanion SyncroPatch 384PE, a 384-well automated patch-clamp platform, with temperature control. We characterize the temperature dependence of hERG gating by fitting the parameters of a mathematical model of hERG kinetics to data obtained at five distinct temperatures between 25 and 37°C and validate the models using different protocols. Our models reveal that activation is far more temperature sensitive than inactivation, and we observe that the temperature dependency of the kinetic parameters is not represented well by Q10 coefficients; it broadly follows a generalized, but not the standardly-used, Eyring relationship. We also demonstrate that experimental estimations of Q10 coefficients are protocol dependent. Our results show that a direct fit using our 15-s protocol best represents hERG kinetics at any given temperature and suggests that using the Generalized Eyring theory is preferable if no experimental data are available to derive model parameters at a given temperature.
Collapse
Affiliation(s)
- Chon Lok Lei
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Michael Clerx
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Kylie A Beattie
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Dario Melgari
- School of Physiology, Pharmacology and Neuroscience, and Cardiovascular Research Laboratories, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, and Cardiovascular Research Laboratories, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | - David J Gavaghan
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Liudmila Polonchuk
- Pharma Research and Early Development, Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ken Wang
- Pharma Research and Early Development, Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
27
|
Lei CL, Clerx M, Gavaghan DJ, Polonchuk L, Mirams GR, Wang K. Rapid Characterization of hERG Channel Kinetics I: Using an Automated High-Throughput System. Biophys J 2019; 117:2438-2454. [PMID: 31447109 PMCID: PMC6990155 DOI: 10.1016/j.bpj.2019.07.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/20/2019] [Accepted: 07/17/2019] [Indexed: 11/27/2022] Open
Abstract
Predicting how pharmaceuticals may affect heart rhythm is a crucial step in drug development and requires a deep understanding of a compound’s action on ion channels. In vitro hERG channel current recordings are an important step in evaluating the proarrhythmic potential of small molecules and are now routinely performed using automated high-throughput patch-clamp platforms. These machines can execute traditional voltage-clamp protocols aimed at specific gating processes, but the array of protocols needed to fully characterize a current is typically too long to be applied in a single cell. Shorter high-information protocols have recently been introduced that have this capability, but they are not typically compatible with high-throughput platforms. We present a new 15 second protocol to characterize hERG (Kv11.1) kinetics, suitable for both manual and high-throughput systems. We demonstrate its use on the Nanion SyncroPatch 384PE, a 384-well automated patch-clamp platform, by applying it to Chinese hamster ovary cells stably expressing hERG1a. From these recordings, we construct 124 cell-specific variants/parameterizations of a hERG model at 25°C. A further eight independent protocols are run in each cell and are used to validate the model predictions. We then combine the experimental recordings using a hierarchical Bayesian model, which we use to quantify the uncertainty in the model parameters, and their variability from cell-to-cell; we use this model to suggest reasons for the variability. This study demonstrates a robust method to measure and quantify uncertainty and shows that it is possible and practical to use high-throughput systems to capture full hERG channel kinetics quantitatively and rapidly.
Collapse
Affiliation(s)
- Chon Lok Lei
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Michael Clerx
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - David J Gavaghan
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Liudmila Polonchuk
- Pharma Research and Early Development, Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom.
| | - Ken Wang
- Pharma Research and Early Development, Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
28
|
Lions S, Dragu R, Carsenty Y, Zukermann R, Aronson D. Determinants of cardiac repolarization and risk for ventricular arrhythmias during mild therapeutic hypothermia. J Crit Care 2019; 46:151-156. [PMID: 29929706 DOI: 10.1016/j.jcrc.2018.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 11/20/2022]
Abstract
PURPOSE We aimed to investigate the factors that modulate the extent of QTc prolongation and potential arrhythmogenic consequences during mild therapeutic hypothermia (MTH). METHODS We studied 205 patients after out-of-hospital cardiac arrest (131 underwent MTH). QTc was measured at baseline, 3h, 6h, 12h, 24h (end of hypothermia), 48h and 72h, and ventricular arrhythmias quantified. RESULTS During MTH, the QTc interval increased progressively peaking at 12h (mean increase 42ms, 95% CI 30-55). There was a strong gender effect (P<0.001) and a significant gender-by-MTH interaction (P=0.004). At 12h, the QTc interval was markedly longer in women as compared with men (mean difference 50ms [95% CI 27-73]. Anoxic brain injury (P=0.002) was also positively associated with QTc prolongation. The risk for ventricular arrhythmic events was not higher with MTH compared with no hypothermia (incidence rate ratio 0.57, 95% CI 0.32-1.02, P=0.06). However, typical cases of Torsade de pointes occurred in association with AV block and LQT2. CONCLUSION QTc prolongation during MTH is strongly affected by female gender and moderately by concomitant anoxic brain injury. Although the overall risk for ventricular arrhythmias is not greater with MTH, Torsade de pointes may develop when other contributing factors coexist.
Collapse
Affiliation(s)
- Svetlana Lions
- Department of Cardiology, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Israel
| | - Robert Dragu
- Department of Cardiology, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Israel
| | - Yoav Carsenty
- Department of Cardiology, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Israel
| | - Robert Zukermann
- Department of Cardiology, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Israel
| | - Doron Aronson
- Department of Cardiology, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Israel.
| |
Collapse
|
29
|
Lee W, Windley MJ, Perry MD, Vandenberg JI, Hill AP. Protocol-Dependent Differences in IC 50 Values Measured in Human Ether-Á-Go-Go-Related Gene Assays Occur in a Predictable Way and Can Be Used to Quantify State Preference of Drug Binding. Mol Pharmacol 2019; 95:537-550. [PMID: 30770456 DOI: 10.1124/mol.118.115220] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/10/2019] [Indexed: 12/22/2022] Open
Abstract
Current guidelines around preclinical screening for drug-induced arrhythmias require the measurement of the potency of block of voltage-gated potassium channel subtype 11.1 (Kv11.1) as a surrogate for risk. A shortcoming of this approach is that the measured IC50 of Kv11.1 block varies widely depending on the voltage protocol used in electrophysiological assays. In this study, we aimed to investigate the factors that contribute to these differences and to identify whether it is possible to make predictions about protocol-dependent block that might facilitate the comparison of potencies measured using different assays. Our data demonstrate that state preferential binding, together with drug-binding kinetics and trapping, is an important determinant of the protocol dependence of Kv11.1 block. We show for the first time that differences in IC50 measured between protocols occurs in a predictable way, such that machine-learning algorithms trained using a selection of simple voltage protocols can indeed predict protocol-dependent potency. Furthermore, we also show that the preference of a drug for binding to the open versus the inactivated state of Kv11.1 can also be inferred from differences in IC50 values measured between protocols. Our work therefore identifies how state preferential drug binding is a major determinant of the protocol dependence of IC50 values measured in preclinical Kv11.1 assays. It also provides a novel method for quantifying the state dependence of Kv11.1 drug binding that will facilitate the development of more complete models of drug binding to Kv11.1 and improve our understanding of proarrhythmic risk associated with compounds that block Kv11.1.
Collapse
Affiliation(s)
- William Lee
- Victor Chang Cardiac Research Institute (W.L., M.J.W., M.D.P., J.I.V., A.P.H.) and St Vincent's Clinical School (W.L., M.J.W., M.D.P., J.I.V., A.P.H.), University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Monique J Windley
- Victor Chang Cardiac Research Institute (W.L., M.J.W., M.D.P., J.I.V., A.P.H.) and St Vincent's Clinical School (W.L., M.J.W., M.D.P., J.I.V., A.P.H.), University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Matthew D Perry
- Victor Chang Cardiac Research Institute (W.L., M.J.W., M.D.P., J.I.V., A.P.H.) and St Vincent's Clinical School (W.L., M.J.W., M.D.P., J.I.V., A.P.H.), University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute (W.L., M.J.W., M.D.P., J.I.V., A.P.H.) and St Vincent's Clinical School (W.L., M.J.W., M.D.P., J.I.V., A.P.H.), University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute (W.L., M.J.W., M.D.P., J.I.V., A.P.H.) and St Vincent's Clinical School (W.L., M.J.W., M.D.P., J.I.V., A.P.H.), University of New South Wales, Darlinghurst, New South Wales, Australia
| |
Collapse
|
30
|
Rayani K, Lin E, Craig C, Lamothe M, Shafaattalab S, Gunawan M, Li AY, Hove-Madsen L, Tibbits GF. Zebrafish as a model of mammalian cardiac function: Optically mapping the interplay of temperature and rate on voltage and calcium dynamics. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:69-90. [DOI: 10.1016/j.pbiomolbio.2018.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022]
|
31
|
Computational modeling of the effect of temperature variations on human pancreatic β-cell activity. J Therm Biol 2018; 75:69-80. [PMID: 30017054 DOI: 10.1016/j.jtherbio.2018.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022]
Abstract
The effect of temperature variations on the pancreatic β-cell activity and the role of different model compartments in temperature sensing have been investigated using a computational modeling approach. The results of our study show that temperature variations by several degrees can change the dynamical states of the β-cell system. In addition, temperature variations can alter the characteristic features of the membrane voltage, which correlates with insulin secretion. Simulation results show that the ion channels such as the L-type calcium, the hERG potassium, sodium channels and the glycolysis pathway are the possible sites for sensing temperature variation. Results indicate that for a small temperature change, even though the frequency and amplitude of electrical activity are altered, the area under the membrane potential curve remains almost unchanged, which implies the existence of a thermoregulatory mechanism for preserving the amount of insulin secretion. Furthermore, the computational analysis shows that the β-cell electrical activity exhibits a bursting pattern in physiological temperature (37 °C) while in vitro studies reported almost the spiking activity at lower temperatures. Since hormone-secreting systems work more efficient in bursting mode, we propose that the pancreatic β-cell works better in the physiological temperature compared with the reference temperature (33 °C).
Collapse
|
32
|
Windley MJ, Lee W, Vandenberg JI, Hill AP. The Temperature Dependence of Kinetics Associated with Drug Block of hERG Channels Is Compound-Specific and an Important Factor for Proarrhythmic Risk Prediction. Mol Pharmacol 2018; 94:760-769. [PMID: 29728448 DOI: 10.1124/mol.117.111534] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/02/2018] [Indexed: 02/14/2025] Open
Abstract
Current mandated preclinical tests for drug-induced proarrhythmia are very sensitive, but not sufficiently specific. This has led to concern that there is a high attrition rate of potentially safe drugs that could have been beneficial to patients. The comprehensive in vitro proarrhythmia initiative has proposed new metrics based around in silico risk predictions, which are informed, among other things, by measures of human ether-à-go-go-related gene channel (hERG) block kinetics. However, high-throughput patch-clamp systems set to collect these data largely operate at ambient temperature, whereas the simulations for risk prediction are carried out at physiologic temperature. The aims of this study were to: 1) determine to what degree kinetics of drug block of hERG are temperature-dependent, 2) assess the impact of any temperature dependence of drug binding kinetics on repolarization in silico, and 3) identify whether a common set of Q10 scalars can be used to extrapolate kinetic data gathered at ambient to physiologic temperatures for use in in silico proarrhythmic risk prediction. We show that, for a range of drugs, kinetics of block are temperature-dependent and, furthermore, that the degree of temperature dependence is different for each drug. As a result, no common set of Q10 scalars could describe the observed range of temperature dependencies. These results suggest that if accurate physiologic temperature models of the kinetics of drug binding are important for in silico risk prediction, the in vitro data should be acquired at physiologic temperature.
Collapse
Affiliation(s)
- Monique J Windley
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - William Lee
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| |
Collapse
|
33
|
Beattie KA, Hill AP, Bardenet R, Cui Y, Vandenberg JI, Gavaghan DJ, de Boer TP, Mirams GR. Sinusoidal voltage protocols for rapid characterisation of ion channel kinetics. J Physiol 2018; 596:1813-1828. [PMID: 29573276 PMCID: PMC5978315 DOI: 10.1113/jp275733] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/19/2018] [Indexed: 12/21/2022] Open
Abstract
Key points Ion current kinetics are commonly represented by current–voltage relationships, time constant–voltage relationships and subsequently mathematical models fitted to these. These experiments take substantial time, which means they are rarely performed in the same cell. Rather than traditional square‐wave voltage clamps, we fitted a model to the current evoked by a novel sum‐of‐sinusoids voltage clamp that was only 8 s long. Short protocols that can be performed multiple times within a single cell will offer many new opportunities to measure how ion current kinetics are affected by changing conditions. The new model predicts the current under traditional square‐wave protocols well, with better predictions of underlying currents than literature models. The current under a novel physiologically relevant series of action potential clamps is predicted extremely well. The short sinusoidal protocols allow a model to be fully fitted to individual cells, allowing us to examine cell–cell variability in current kinetics for the first time.
Abstract Understanding the roles of ion currents is crucial to predict the action of pharmaceuticals and mutations in different scenarios, and thereby to guide clinical interventions in the heart, brain and other electrophysiological systems. Our ability to predict how ion currents contribute to cellular electrophysiology is in turn critically dependent on our characterisation of ion channel kinetics – the voltage‐dependent rates of transition between open, closed and inactivated channel states. We present a new method for rapidly exploring and characterising ion channel kinetics, applying it to the hERG potassium channel as an example, with the aim of generating a quantitatively predictive representation of the ion current. We fitted a mathematical model to currents evoked by a novel 8 second sinusoidal voltage clamp in CHO cells overexpressing hERG1a. The model was then used to predict over 5 minutes of recordings in the same cell in response to further protocols: a series of traditional square step voltage clamps, and also a novel voltage clamp comprising a collection of physiologically relevant action potentials. We demonstrate that we can make predictive cell‐specific models that outperform the use of averaged data from a number of different cells, and thereby examine which changes in gating are responsible for cell–cell variability in current kinetics. Our technique allows rapid collection of consistent and high quality data, from single cells, and produces more predictive mathematical ion channel models than traditional approaches. Ion current kinetics are commonly represented by current–voltage relationships, time constant–voltage relationships and subsequently mathematical models fitted to these. These experiments take substantial time, which means they are rarely performed in the same cell. Rather than traditional square‐wave voltage clamps, we fitted a model to the current evoked by a novel sum‐of‐sinusoids voltage clamp that was only 8 s long. Short protocols that can be performed multiple times within a single cell will offer many new opportunities to measure how ion current kinetics are affected by changing conditions. The new model predicts the current under traditional square‐wave protocols well, with better predictions of underlying currents than literature models. The current under a novel physiologically relevant series of action potential clamps is predicted extremely well. The short sinusoidal protocols allow a model to be fully fitted to individual cells, allowing us to examine cell–cell variability in current kinetics for the first time.
Collapse
Affiliation(s)
- Kylie A Beattie
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, OX1 3QD, UK.,Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Adam P Hill
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Rémi Bardenet
- CNRS & CRIStAL, Université de Lille, 59651 Villeneuve d'Ascq, Lille, France
| | - Yi Cui
- Safety Evaluation and Risk Management, Global Clinical Safety and Pharmacovigilance, GlaxoSmithKline, Uxbridge, UB11 1BS, UK
| | - Jamie I Vandenberg
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - David J Gavaghan
- Computational Biology, Department of Computer Science, University of Oxford, Oxford, OX1 3QD, UK
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
34
|
Ferrante M, Tahvildari B, Duque A, Hadzipasic M, Salkoff D, Zagha EW, Hasselmo ME, McCormick DA. Distinct Functional Groups Emerge from the Intrinsic Properties of Molecularly Identified Entorhinal Interneurons and Principal Cells. Cereb Cortex 2018; 27:3186-3207. [PMID: 27269961 DOI: 10.1093/cercor/bhw143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Inhibitory interneurons are an important source of synaptic inputs that may contribute to network mechanisms for coding of spatial location by entorhinal cortex (EC). The intrinsic properties of inhibitory interneurons in the EC of the mouse are mostly undescribed. Intrinsic properties were recorded from known cell types, such as, stellate and pyramidal cells and 6 classes of molecularly identified interneurons (regulator of calcineurin 2, somatostatin, serotonin receptor 3a, neuropeptide Y neurogliaform (NGF), neuropeptide Y non-NGF, and vasoactive intestinal protein) in acute brain slices. We report a broad physiological diversity between and within cell classes. We also found differences in the ability to produce postinhibitory rebound spikes and in the frequency and amplitude of incoming EPSPs. To understand the source of this intrinsic variability we applied hierarchical cluster analysis to functionally classify neurons. These analyses revealed physiologically derived cell types in EC that mostly corresponded to the lines identified by biomarkers with a few unexpected and important differences. Finally, we reduced the complex multidimensional space of intrinsic properties to the most salient five that predicted the cellular biomolecular identity with 81.4% accuracy. These results provide a framework for the classification of functional subtypes of cortical neurons by their intrinsic membrane properties.
Collapse
Affiliation(s)
- Michele Ferrante
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Babak Tahvildari
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Alvaro Duque
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Muhamed Hadzipasic
- Interdepartmental Program in Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - David Salkoff
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Edward William Zagha
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Michael E Hasselmo
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - David A McCormick
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| |
Collapse
|
35
|
Lee W, Windley MJ, Vandenberg JI, Hill AP. In Vitro and In Silico Risk Assessment in Acquired Long QT Syndrome: The Devil Is in the Details. Front Physiol 2017; 8:934. [PMID: 29201009 PMCID: PMC5696636 DOI: 10.3389/fphys.2017.00934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022] Open
Abstract
Acquired long QT syndrome, mostly as a result of drug block of the Kv11. 1 potassium channel in the heart, is characterized by delayed cardiac myocyte repolarization, prolongation of the T interval on the ECG, syncope and sudden cardiac death due to the polymorphic ventricular arrhythmia Torsade de Pointes (TdP). In recent years, efforts are underway through the Comprehensive in vitro proarrhythmic assay (CiPA) initiative, to develop better tests for this drug induced arrhythmia based in part on in silico simulations of pharmacological disruption of repolarization. However, drug binding to Kv11.1 is more complex than a simple binary molecular reaction, meaning simple steady state measures of potency are poor surrogates for risk. As a result, there is a plethora of mechanistic detail describing the drug/Kv11.1 interaction—such as drug binding kinetics, state preference, temperature dependence and trapping—that needs to be considered when developing in silico models for risk prediction. In addition to this, other factors, such as multichannel pharmacological profile and the nature of the ventricular cell models used in simulations also need to be considered in the search for the optimum in silico approach. Here we consider how much of mechanistic detail needs to be included for in silico models to accurately predict risk and further, how much of this detail can be retrieved from protocols that are practical to implement in high throughout screens as part of next generation of preclinical in silico drug screening approaches?
Collapse
Affiliation(s)
- William Lee
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Monique J Windley
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I Vandenberg
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Adam P Hill
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
36
|
Drew D, Baranchuk A, Hopman W, Brison RJ. The impact of fever on corrected QT interval. J Electrocardiol 2017; 50:570-575. [DOI: 10.1016/j.jelectrocard.2017.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Indexed: 12/27/2022]
|
37
|
Oliver KL, Franceschetti S, Milligan CJ, Muona M, Mandelstam SA, Canafoglia L, Boguszewska-Chachulska AM, Korczyn AD, Bisulli F, Di Bonaventura C, Ragona F, Michelucci R, Ben-Zeev B, Straussberg R, Panzica F, Massano J, Friedman D, Crespel A, Engelsen BA, Andermann F, Andermann E, Spodar K, Lasek-Bal A, Riguzzi P, Pasini E, Tinuper P, Licchetta L, Gardella E, Lindenau M, Wulf A, Møller RS, Benninger F, Afawi Z, Rubboli G, Reid CA, Maljevic S, Lerche H, Lehesjoki AE, Petrou S, Berkovic SF. Myoclonus epilepsy and ataxia due to KCNC1 mutation: Analysis of 20 cases and K + channel properties. Ann Neurol 2017; 81:677-689. [PMID: 28380698 DOI: 10.1002/ana.24929] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To comprehensively describe the new syndrome of myoclonus epilepsy and ataxia due to potassium channel mutation (MEAK), including cellular electrophysiological characterization of observed clinical improvement with fever. METHODS We analyzed clinical, electroclinical, and neuroimaging data for 20 patients with MEAK due to recurrent KCNC1 p.R320H mutation. In vitro electrophysiological studies were conducted using whole cell patch-clamp to explore biophysical properties of wild-type and mutant KV 3.1 channels. RESULTS Symptoms began at between 3 and 15 years of age (median = 9.5), with progressively severe myoclonus and rare tonic-clonic seizures. Ataxia was present early, but quickly became overshadowed by myoclonus; 10 patients were wheelchair-bound by their late teenage years. Mild cognitive decline occurred in half. Early death was not observed. Electroencephalogram (EEG) showed generalized spike and polyspike wave discharges, with documented photosensitivity in most. Polygraphic EEG-electromyographic studies demonstrated a cortical origin for myoclonus and striking coactivation of agonist and antagonist muscles. Magnetic resonance imaging revealed symmetrical cerebellar atrophy, which appeared progressive, and a prominent corpus callosum. Unexpectedly, transient clinical improvement with fever was noted in 6 patients. To explore this, we performed high-temperature in vitro recordings. At elevated temperatures, there was a robust leftward shift in activation of wild-type KV 3.1, increasing channel availability. INTERPRETATION MEAK has a relatively homogeneous presentation, resembling Unverricht-Lundborg disease, despite the genetic and biological basis being quite different. A remarkable improvement with fever may be explained by the temperature-dependent leftward shift in activation of wild-type KV 3.1 subunit-containing channels, which would counter the loss of function observed for mutant channels, highlighting KCNC1 as a potential target for precision therapeutics. Ann Neurol 2017;81:677-689.
Collapse
Affiliation(s)
- Karen L Oliver
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Silvana Franceschetti
- Department of Neurophysiology, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Carol J Milligan
- Ion Channels and Disease Group, Epilepsy Division, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Mikko Muona
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Simone A Mandelstam
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Departments of Paediatrics and Radiology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Medical Imaging, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Laura Canafoglia
- Department of Neurophysiology, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | | | - Amos D Korczyn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Francesca Bisulli
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carlo Di Bonaventura
- Department of Neurological Sciences, University of Rome, La Sapienza, Rome, Italy
| | - Francesca Ragona
- Department of Pediatric Neuroscience, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Roberto Michelucci
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Unit of Neurology, Bellaria Hospital, Bologna, Italy
| | - Bruria Ben-Zeev
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Rachel Straussberg
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Epilepsy Unit, Schneider Children's Medical Center of Israel, Petah Tikvah, Israel
| | - Ferruccio Panzica
- Department of Neurophysiology, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - João Massano
- Department of Neurology, Hospital Pedro Hispano/ULS Matosinhos, Senhora da Hora, Portugal.,Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Langone Medical Center, New York, NY
| | - Arielle Crespel
- Epilepsy Unit, Gui de Chauliac Hospital, Montpellier, France
| | - Bernt A Engelsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Frederick Andermann
- Epilepsy Research Group, Montreal Neurological Hospital and Institute, Montreal, Quebec, Canada.,Departments of Neurology & Neurosurgery and Paediatrics, McGill University, Montreal, Quebec, Canada
| | - Eva Andermann
- Neurogenetics Unit and Epilepsy Research Group, Montreal Neurological Hospital and Institute, Montreal, Quebec, Canada.,Departments of Neurology & Neurosurgery and Human Genetics, McGill University, Montreal, Quebec, Canada
| | | | - Anetta Lasek-Bal
- High School of Science, Medical University of Silesia, Department of Neurology, Upper Silesian Medical Center, Katowice, Poland
| | - Patrizia Riguzzi
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Unit of Neurology, Bellaria Hospital, Bologna, Italy
| | - Elena Pasini
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Unit of Neurology, Bellaria Hospital, Bologna, Italy
| | - Paolo Tinuper
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Laura Licchetta
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elena Gardella
- Danish Epilepsy Center, Dianalund, Denmark.,Institute for Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Matthias Lindenau
- Department of Neurology and Epileptology, Epilepsy Center Hamburg-Alsterdorf, Hamburg, Germany
| | - Annette Wulf
- Department of Neurology and Epileptology, Epilepsy Center Hamburg-Alsterdorf, Hamburg, Germany
| | - Rikke S Møller
- Danish Epilepsy Center, Dianalund, Denmark.,Institute for Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Felix Benninger
- Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petah Tikvah, Israel
| | - Zaid Afawi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guido Rubboli
- IRCCS-Institute of Neurological Sciences of Bologna, Bologna, Italy.,Danish Epilepsy Center, Filadelfia/University of Copenhagen, Dianalund, Denmark
| | - Christopher A Reid
- Ion Channels and Disease Group, Epilepsy Division, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Snezana Maljevic
- Ion Channels and Disease Group, Epilepsy Division, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,University of Tübingen, Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Holger Lerche
- University of Tübingen, Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Anna-Elina Lehesjoki
- Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Steven Petrou
- Ion Channels and Disease Group, Epilepsy Division, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Centre for Neural Engineering, Department of Electrical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
38
|
Vandenberg JI, Perozo E, Allen TW. Towards a Structural View of Drug Binding to hERG K + Channels. Trends Pharmacol Sci 2017; 38:899-907. [PMID: 28711156 DOI: 10.1016/j.tips.2017.06.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/09/2017] [Accepted: 06/14/2017] [Indexed: 01/10/2023]
Abstract
The human ether-a-go-go-related gene (hERG) K+ channel is of great medical and pharmaceutical relevance. Inherited mutations in hERG result in congenital long-QT syndrome which is associated with a markedly increased risk of cardiac arrhythmia and sudden death. hERG K+ channels are also remarkably susceptible to block by a wide range of drugs, which in turn can cause drug-induced long-QT syndrome and an increased risk of sudden death. The recent determination of the near-atomic resolution structure of the hERG K+ channel, using single-particle cryo-electron microscopy (cryo-EM), provides tremendous insights into how these channels work. It also suggests a way forward in our quest to understand why these channels are so promiscuous with respect to drug binding.
Collapse
Affiliation(s)
- Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Eduardo Perozo
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Toby W Allen
- School of Science, RMIT University, Melbourne, VIC 3001, Australia
| |
Collapse
|
39
|
Mauerhöfer M, Bauer CK. Effects of Temperature on Heteromeric Kv11.1a/1b and Kv11.3 Channels. Biophys J 2017; 111:504-523. [PMID: 27508435 DOI: 10.1016/j.bpj.2016.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 01/17/2023] Open
Abstract
Kv11.1 channels are crucial in cardiac physiology, and there is increasing evidence of physiological roles of different Kv11 channels outside the heart. The HERG (human Kv11.1a) channel has previously been shown to carry substantially more current at elevated temperatures, and we have now comparably investigated the temperature dependence of neuronal Kv11.3 channels and the more ubiquitous heteromeric Kv11.1a/1b channels. Transiently expressed rat Kv11 channels were studied at 21°C, 30°C, and 35°C. At near-physiological temperature, the maximal sustained outward current density was almost three times the mean value obtained at room temperature for Kv11.1a/1b, and increased by ∼150% for Kv11.3. For both channels, reduced inactivation contributed to the current increase at higher temperature. Elevated temperature moved Kv11.1a/1b isochronal activation curves to more negative potentials, but shifted the potential of half-maximal Kv11.3 channel activation to more depolarized values and reduced its voltage sensitivity. Thus, increased temperature stabilized the open state over the closed state of Kv11.1a/1b channels and exerted the opposite effect on Kv11.3 channel activation. Both Kv11 channels exhibited an overall high temperature sensitivity of most gating parameters, with remarkably high Q10 factors of ∼5 for the rate of Kv11.1a/1b activation. The Q10 factors for Kv11.3 gating were more uniform, but still higher for activation than for inactivation kinetics. The results demonstrate that characteristic differences between Kv11.1a/1b and Kv11.3 determined at room temperature do not necessarily apply to physiological conditions. The data provided here can aid in the design of models that will enhance our understanding of the role of Kv11 currents in excitable cells.
Collapse
Affiliation(s)
- Maike Mauerhöfer
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane K Bauer
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
40
|
Phan K, Ng CA, David E, Shishmarev D, Kuchel PW, Vandenberg JI, Perry MD. The S1 helix critically regulates the finely tuned gating of Kv11.1 channels. J Biol Chem 2017; 292:7688-7705. [PMID: 28280240 DOI: 10.1074/jbc.m117.779298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/26/2017] [Indexed: 11/06/2022] Open
Abstract
Congenital mutations in the cardiac Kv11.1 channel can cause long QT syndrome type 2 (LQTS2), a heart rhythm disorder associated with sudden cardiac death. Mutations act either by reducing protein expression at the membrane and/or by perturbing the intricate gating properties of Kv11.1 channels. A number of clinical LQTS2-associated mutations have been reported in the first transmembrane segment (S1) of Kv11.1 channels, but the role of this region of the channel is largely unexplored. In part, this is due to problems defining the extent of the S1 helix, as a consequence of its low sequence homology with other Kv family members. Here, we used NMR spectroscopy and electrophysiological characterization to show that the S1 of Kv11.1 channels extends seven helical turns, from Pro-405 to Phe-431, and is flanked by unstructured loops. Functional analysis suggests that pre-S1 loop residues His-402 and Tyr-403 play an important role in regulating the kinetics and voltage dependence of channel activation and deactivation. Multiple residues within the S1 helix also play an important role in fine-tuning the voltage dependence of activation, regulating slow deactivation, and modulating C-type inactivation of Kv11.1 channels. Analyses of LQTS2-associated mutations in the pre-S1 loop or S1 helix of Kv11.1 channels demonstrate perturbations to both protein expression and most gating transitions. Thus, S1 region mutations would reduce both the action potential repolarizing current passed by Kv11.1 channels in cardiac myocytes, as well as the current passed in response to premature depolarizations that normally helps protect against the formation of ectopic beats.
Collapse
Affiliation(s)
- Kevin Phan
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Chai Ann Ng
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Erikka David
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010
| | - Dmitry Shishmarev
- the School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Philip W Kuchel
- the School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jamie I Vandenberg
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Matthew D Perry
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010, .,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| |
Collapse
|
41
|
Badr A, Hassinen M, El-Sayed MF, Vornanen M. Effects of seasonal acclimatization on action potentials and sarcolemmal K+ currents in roach (Rutilus rutilus) cardiac myocytes. Comp Biochem Physiol A Mol Integr Physiol 2017; 205:15-27. [DOI: 10.1016/j.cbpa.2016.12.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 01/10/2023]
|
42
|
Windley MJ, Abi-Gerges N, Fermini B, Hancox JC, Vandenberg JI, Hill AP. Measuring kinetics and potency of hERG block for CiPA. J Pharmacol Toxicol Methods 2017; 87:99-107. [PMID: 28192183 DOI: 10.1016/j.vascn.2017.02.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/25/2017] [Accepted: 02/07/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The Comprehensive in vitro Proarrhythmic Assay (CiPA) aims to update current cardiac safety testing to better evaluate arrhythmic risk. A central theme of CiPA is the use of in silico approaches to risk prediction incorporating models of drug binding to hERG. To parameterize these models, accurate in vitro measurement of potency and kinetics of block is required. The Ion Channel Working Group was tasked with: i) selecting a protocol that could measure kinetics of block and was easily implementable on automated platforms for future rollout in industry and ii) acquiring a reference dataset using the standardized protocol. METHODS Data were acquired using a 'step depolarisation' protocol using manual patch-clamp at ambient temperature. RESULTS Potency, kinetics and trapping characteristics of hERG block for the CiPA training panel of twelve drugs were measured. Timecourse of block and trapping characteristics could be reliably measured if the time constant for onset of block was between ~500ms and ~15s. Seven drugs, however had time courses of block faster than this cut-off. DISCUSSION Here we describe the implementation of the standardized protocol for measurement of kinetics and potency of hERG block for CiPA. The results highlight the challenges in identifying a single protocol to measure hERG block over a range of kinetics. The dataset from this study is being used by the In Silico Working Group to develop models of drug binding for risk prediction and is freely available as a 'gold standard' ambient temperature dataset to evaluate variability across high throughput platforms.
Collapse
Affiliation(s)
- Monique J Windley
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | | | - Bernard Fermini
- Coyne Scientific, LLC, 58 Edgewood Ave NE Atlanta, GA 30303, USA
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, UK
| | - Jamie I Vandenberg
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Adam P Hill
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
43
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
44
|
Zhao Y, Wang T, Guo J, Yang T, Li W, Koichopolos J, Lamothe SM, Kang Y, Ma A, Zhang S. Febrile temperature facilitates hERG/IKr degradation through an altered K(+) dependence. Heart Rhythm 2016; 13:2004-11. [PMID: 27321242 DOI: 10.1016/j.hrthm.2016.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Dysfunction of the rapidly activating delayed rectifier K(+) channel (IKr) encoded by the human ether-à-go-go-related gene (hERG) is the primary cause of acquired long QT syndrome (LQTS). Fever has been reported to trigger LQTS in various conditions. OBJECTIVE We aim to clarify the effect and underlying mechanisms of febrile temperature on hERG expressed in HEK cells, IKr in neonatal rat ventricular myocytes, and the QT interval in rabbits. METHODS Western blot analysis was used to determine the expression of hERG channel protein in stably transfected HEK 293 cells. Immunocytochemistry was used to visualize the localization of hERG channels. The whole-cell patch clamp technique was used to record hERG K(+) current (IhERG) in hERG expressing HEK 293 cells, as well as IKr, transient outward K(+) current (Ito), and L-type Ca(2+) current (ICa) in neonatal rat ventricular myocytes. Electrocardiographic recordings were performed in an in vivo rabbit model. RESULTS Compared with culture at 37°C, culture at 40°C reduced the mature hERG expression and IhERG in an extracellular K(+) concentration-dependent manner. Point mutations that remove the K(+) dependence of hERG-S624T and F627Y-also abolished the febrile temperature-induced hERG reduction. In neonatal rat ventricular myocytes, febrile temperature prolonged the action potential duration and selectively reduced IKr in a manner similar to low K(+) culture. In an in vivo rabbit model, fever and hypokalemia synergistically prolonged the QT interval. CONCLUSION Febrile temperature facilitates the development of LQTS by expediting hERG degradation through altered K(+) dependence.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Jun Guo
- 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
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jennifer Koichopolos
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shawn M Lamothe
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Yudi Kang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada,.
| |
Collapse
|
45
|
Li Z, Dutta S, Sheng J, Tran PN, Wu W, Colatsky T. A temperature-dependent in silico model of the human ether-à-go-go-related (hERG) gene channel. J Pharmacol Toxicol Methods 2016; 81:233-9. [PMID: 27178106 DOI: 10.1016/j.vascn.2016.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/20/2016] [Accepted: 05/09/2016] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Current regulatory guidelines for assessing the risk of QT prolongation include in vitro assays assessing drug effects on the human ether-à-go-go-related (hERG; also known as Kv11.1) channel expressed in cell lines. These assays are typically conducted at room temperature to promote the ease and stability of recording hERG currents. However, the new Comprehensive in vitro Proarrhythmia Assay (CiPA) paradigm proposes to use an in silico model of the human ventricular myocyte to assess risk, requiring as input hERG channel pharmacology data obtained at physiological temperatures. To accommodate current industry safety pharmacology practices for measuring hERG channel activity, an in silico model of hERG channel that allows for the extrapolation of hERG assay data across different temperatures is desired. Because temperature may have an effect on both channel gating and drug binding rate, such models may need to have two components: a base model dealing with temperature-dependent gating changes without drug, and a pharmacodynamic component simulating temperature-dependent drug binding kinetics. As a first step, a base mode that can capture temperature effects on hERG channel gating without drug is needed. METHODS AND RESULTS To meet this need for a temperature-dependent base model, a Markov model of the hERG channel with state transition rates explicitly dependent on temperature was developed and calibrated using data from a variety of published experiments conducted over a range of temperatures. The model was able to reproduce observed temperature-dependent changes in key channel gating properties and also to predict the results obtained in independent sets of new experiments. DISCUSSION This new temperature-sensitive model of hERG gating represents an attempt to improve the predictivity of safety pharmacology testing by enabling the translation of room temperature hERG assay data to more physiological conditions. With further development, this model can be incorporated into the CiPA paradigm and also be used as a tool for developing insights into the thermodynamics of hERG channel gating mechanisms and the temperature-dependence of hERG channel block by drugs.
Collapse
Affiliation(s)
- Zhihua Li
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States.
| | - Sara Dutta
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States
| | - Jiansong Sheng
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States
| | - Phu N Tran
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States
| | - Wendy Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States
| | - Thomas Colatsky
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, United States
| |
Collapse
|
46
|
Guo J, Cheng YM, Lees-Miller JP, Perissinotti LL, Claydon TW, Hull CM, Thouta S, Roach DE, Durdagi S, Noskov SY, Duff HJ. NS1643 interacts around L529 of hERG to alter voltage sensor movement on the path to activation. Biophys J 2016; 108:1400-1413. [PMID: 25809253 DOI: 10.1016/j.bpj.2014.12.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 11/25/2014] [Accepted: 12/08/2014] [Indexed: 12/17/2022] Open
Abstract
Activators of hERG1 such as NS1643 are being developed for congenital/acquired long QT syndrome. Previous studies identify the neighborhood of L529 around the voltage-sensor as a putative interacting site for NS1643. With NS1643, the V1/2 of activation of L529I (-34 ± 4 mV) is similar to wild-type (WT) (-37 ± 3 mV; P > 0.05). WT and L529I showed no difference in the slope factor in the absence of NS1643 (8 ± 0 vs. 9 ± 0) but showed a difference in the presence of NS1643 (9 ± 0.3 vs. 22 ± 1; P < 0.01). Voltage-clamp-fluorimetry studies also indicated that in L529I, NS1643 reduces the voltage-sensitivity of S4 movement. To further assess mechanism of NS1643 action, mutations were made in this neighborhood. NS1643 shifts the V1/2 of activation of both K525C and K525C/L529I to hyperpolarized potentials (-131 ± 4 mV for K525C and -120 ± 21 mV for K525C/L529I). Both K525C and K525C/K529I had similar slope factors in the absence of NS1643 (18 ± 2 vs. 34 ± 5, respectively) but with NS1643, the slope factor of K525C/L529I increased from 34 ± 5 to 71 ± 10 (P < 0.01) whereas for K525C the slope factor did not change (18 ± 2 at baseline and 16 ± 2 for NS1643). At baseline, K525R had a slope factor similar to WT (9 vs. 8) but in the presence of NS1643, the slope factor of K525R was increased to 24 ± 4 vs. 9 ± 0 mV for WT (P < 0.01). Molecular modeling indicates that L529I induces a kink in the S4 voltage-sensor helix, altering a salt-bridge involving K525. Moreover, docking studies indicate that NS1643 binds to the kinked structure induced by the mutation with a higher affinity. Combining biophysical, computational, and electrophysiological evidence, a mechanistic principle governing the action of some activators of hERG1 channels is proposed.
Collapse
Affiliation(s)
- Jiqing Guo
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Yen May Cheng
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada
| | - James P Lees-Miller
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Laura L Perissinotti
- Centre for Molecular Simulations, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Tom W Claydon
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Christina M Hull
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Samrat Thouta
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Daniel E Roach
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Serdar Durdagi
- Centre for Molecular Simulations, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Sergei Y Noskov
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada; Centre for Molecular Simulations, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
47
|
Perissinotti LL, Guo J, De Biase PM, Clancy CE, Duff HJ, Noskov SY. Kinetic model for NS1643 drug activation of WT and L529I variants of Kv11.1 (hERG1) potassium channel. Biophys J 2015; 108:1414-1424. [PMID: 25809254 PMCID: PMC4375712 DOI: 10.1016/j.bpj.2014.12.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 11/24/2014] [Accepted: 12/08/2014] [Indexed: 11/28/2022] Open
Abstract
Congenital and acquired (drug-induced) forms of the human long-QT syndrome are associated with alterations in Kv11.1 (hERG) channel-controlled repolarizing IKr currents of cardiac action potentials. A mandatory drug screen implemented by many countries led to a discovery of a large group of small molecules that can activate hERG currents and thus may act as potent antiarrhythmic agents. Despite significant progress in identification of channel activators, little is known about their mechanism of action. A combination of electrophysiological studies with molecular and kinetic modeling was used to examine the mechanism of a model activator (NS1643) action on the hERG channel and its L529I mutant. The L529I mutant has gating dynamics similar to that of wild-type while its response to application of NS1643 is markedly different. We propose a mechanism compatible with experiments in which the model activator binds to the closed (C3) and open states (O). We suggest that NS1643 is affecting early gating transitions, probably during movements of the voltage sensor that precede the opening of the activation gate.
Collapse
Affiliation(s)
- Laura L Perissinotti
- Centre for Molecular Modeling, Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Jiqing Guo
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pablo M De Biase
- Centre for Molecular Modeling, Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Colleen E Clancy
- Department of Pharmacology, University of California at Davis, Davis, California.
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Sergei Y Noskov
- Centre for Molecular Modeling, Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
48
|
Vonderlin N, Fischer F, Zitron E, Seyler C, Scherer D, Thomas D, Katus HA, Scholz EP. Anesthetic drug midazolam inhibits cardiac human ether-à-go-go-related gene channels: mode of action. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:867-77. [PMID: 25733807 PMCID: PMC4338257 DOI: 10.2147/dddt.s72765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Midazolam is a short-acting benzodiazepine that is in wide clinical use as an anxiolytic, sedative, hypnotic, and anticonvulsant. Midazolam has been shown to inhibit ion channels, including calcium and potassium channels. So far, the effects of midazolam on cardiac human ether-à-go-go-related gene (hERG) channels have not been analyzed. The inhibitory effects of midazolam on heterologously expressed hERG channels were analyzed in Xenopus oocytes using the double-electrode voltage clamp technique. We found that midazolam inhibits hERG channels in a concentration-dependent manner, yielding an IC50 of 170 μM in Xenopus oocytes. When analyzed in a HEK 293 cell line using the patch-clamp technique, the IC50 was 13.6 μM. Midazolam resulted in a small negative shift of the activation curve of hERG channels. However, steady-state inactivation was not significantly affected. We further show that inhibition is state-dependent, occurring within the open and inactivated but not in the closed state. There was no frequency dependence of block. Using the hERG pore mutants F656A and Y652A we provide evidence that midazolam uses a classical binding site within the channel pore. Analyzing the subacute effects of midazolam on hERG channel trafficking, we further found that midazolam does not affect channel surface expression. Taken together, we show that the anesthetic midazolam is a low-affinity inhibitor of cardiac hERG channels without additional effects on channel surface expression. These data add to the current understanding of the pharmacological profile of the anesthetic midazolam.
Collapse
Affiliation(s)
- Nadine Vonderlin
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Fathima Fischer
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Edgar Zitron
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Claudia Seyler
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Scherer
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Dierk Thomas
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Eberhard P Scholz
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
49
|
Combined action potential- and dynamic-clamp for accurate computational modelling of the cardiac IKr current. J Mol Cell Cardiol 2015; 79:187-94. [DOI: 10.1016/j.yjmcc.2014.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/29/2014] [Accepted: 11/12/2014] [Indexed: 11/18/2022]
|
50
|
Wiśniowska B, Mendyk A, Fijorek K, Polak S. Computer-based prediction of the drug proarrhythmic effect: problems, issues, known and suspected challenges. Europace 2015; 16:724-35. [PMID: 24798962 DOI: 10.1093/europace/euu009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
It is likely that computer modelling and simulations will become an element of comprehensive cardiac safety testing. Their role would be primarily the integration and the interpretation of previously gathered data. There are still unanswered questions and issues which we list and describe below. They include sources of data used for the development of the models as well as data utilized as input information, which can come from the in vitro studies and the quantitative structure-activity relationship models. The pharmacokinetics of the drugs in question play a crucial role as their active concentration should be considered, yet the question remains where is the right place to assess it. The pharmacodynamic angle includes complications coming from multiple drugs (i.e. active metabolites) acting in parallel as well as the type of interaction with (potentially) multiple affected channels. Once established, the model and the methodology of its use should be further validated, optimistically against individual data reported at the clinical level as the physiological, anatomical, and genetic parameters play a crucial role in the drug-triggered arrhythmia induction. All the abovementioned issues should be at least considered and-hopefully-resolved, to properly utilize the mathematical models for a cardiac safety assessment.
Collapse
Affiliation(s)
- Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, 30-688 Kraków, Poland
| | | | | | | |
Collapse
|