1
|
Huang H, Pugsley MK, Fermini B, Curtis MJ, Koerner J, Accardi M, Authier S. Cardiac voltage-gated ion channels in safety pharmacology: Review of the landscape leading to the CiPA initiative. J Pharmacol Toxicol Methods 2017; 87:11-23. [PMID: 28408211 DOI: 10.1016/j.vascn.2017.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 03/27/2017] [Accepted: 04/06/2017] [Indexed: 12/15/2022]
Abstract
Voltage gated ion channels are central in defining the fundamental properties of the ventricular cardiac action potential (AP), and are also involved in the development of drug-induced arrhythmias. Many drugs can inhibit cardiac ion currents, including the Na+ current (INa), L-type Ca2+ current (Ica-L), and K+ currents (Ito, IK1, IKs, and IKr), and thereby affect AP properties in a manner that can trigger or sustain cardiac arrhythmias. Since publication of ICH E14 and S7B over a decade ago, there has been a focus on drug effects on QT prolongation clinically, and on the rapidly activating delayed rectifier current (IKr), nonclinically, for evaluation of proarrhythmic risk. This focus on QT interval prolongation and a single ionic current likely impacted negatively some drugs that lack proarrhythmic liability in humans. To rectify this issue, the Comprehensive in vitro proarrhythmia assay (CiPA) initiative has been proposed to integrate drug effects on multiple cardiac ionic currents with in silico modelling of human ventricular action potentials, and in vitro data obtained from human stem cell-derived ventricular cardiomyocytes to estimate proarrhythmic risk of new drugs with improved accuracy. In this review, we present the physiological functions and the molecular basis of major cardiac ion channels that contribute to the ventricle AP, and discuss the CiPA paradigm in drug development.
Collapse
Affiliation(s)
- Hai Huang
- CiToxLAB North America, 445, Armand-Frappier Boul, Laval H7V 4B3, QC, Canada
| | - Michael K Pugsley
- Department of Toxicology, Purdue Pharma L.P., Cranbury, NJ 08512, USA
| | | | - Michael J Curtis
- Cardiovascular Division, Faculty of Life Sciences & Medicine, King's College London, Rayne Institute, St Thomas' Hospital, London SE17EH, UK
| | - John Koerner
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Michael Accardi
- CiToxLAB North America, 445, Armand-Frappier Boul, Laval H7V 4B3, QC, Canada
| | - Simon Authier
- CiToxLAB North America, 445, Armand-Frappier Boul, Laval H7V 4B3, QC, Canada.
| |
Collapse
|
2
|
Hoeker GS, Skarsfeldt MA, Jespersen T, Poelzing S. Electrophysiologic effects of the IK1 inhibitor PA-6 are modulated by extracellular potassium in isolated guinea pig hearts. Physiol Rep 2017; 5:e13120. [PMID: 28087819 PMCID: PMC5256165 DOI: 10.14814/phy2.13120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/14/2016] [Indexed: 12/02/2022] Open
Abstract
The pentamidine analog PA-6 was developed as a specific inward rectifier potassium current (IK1) antagonist, because established inhibitors either lack specificity or have side effects that prohibit their use in vivo. We previously demonstrated that BaCl2, an established IK1 inhibitor, could prolong action potential duration (APD) and increase cardiac conduction velocity (CV). However, few studies have addressed whether targeted IK1 inhibition similarly affects ventricular electrophysiology. The aim of this study was to determine the effects of PA-6 on cardiac repolarization and conduction in Langendorff-perfused guinea pig hearts. PA-6 (200 nm) or vehicle was perfused into ex-vivo guinea pig hearts for 60 min. Hearts were optically mapped with di-4-ANEPPS to quantify CV and APD at 90% repolarization (APD90). Ventricular APD90 was significantly prolonged in hearts treated with PA-6 (115 ± 2% of baseline; P < 0.05), but not vehicle (105 ± 2% of baseline). PA-6 slightly, but significantly, increased transverse CV by 7%. PA-6 significantly prolonged APD90 during hypokalemia (2 mmol/L [K+]o), although to a lesser degree than observed at 4.56 mmol/L [K+]o In contrast, the effect of PA-6 on CV was more pronounced during hypokalemia, where transverse CV with PA-6 (24 ± 2 cm/sec) was significantly faster than with vehicle (13 ± 3 cm/sec, P < 0.05). These results show that under normokalemic conditions, PA-6 significantly prolonged APD90, whereas its effect on CV was modest. During hypokalemia, PA-6 prolonged APD90 to a lesser degree, but profoundly increased CV Thus, in intact guinea pig hearts, the electrophysiologic effects of the IK1 inhibitor, PA-6, are [K+]o-dependent.
Collapse
Affiliation(s)
- Gregory S Hoeker
- Biomedical Engineering and Mechanics, Center for Heart and Regenerative Medicine, Virginia Tech Virginia Tech Carilion Research Institute, Roanoke, Virginia
| | - Mark A Skarsfeldt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steven Poelzing
- Biomedical Engineering and Mechanics, Center for Heart and Regenerative Medicine, Virginia Tech Virginia Tech Carilion Research Institute, Roanoke, Virginia
| |
Collapse
|
3
|
Allocryptopine and benzyltetrahydropalmatine block hERG potassium channels expressed in HEK293 cells. Acta Pharmacol Sin 2013; 34:847-58. [PMID: 23524574 DOI: 10.1038/aps.2012.176] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
AIM Allocryptopine (ALL) is an alkaloid extracted from Corydalis decumbens (Thunb) Pers. Papaveraceae, whereas benzyltetrahydropalmatine (BTHP) is a derivative of tetrahydropalmatine extracted from Corydalis ambigua (Pall) Cham et Schlecht. The aim of this study was to investigate the effects of ALL and BTHP on the human ether-a-go-go related gene (hERG) current expressed in HEK293 cells. METHODS Cultured HEK293 cells were transiently transfected with hERG channel cDNA plasmid pcDNA3.1 using Lipofectamine. The whole-cell current IHERG was evoked and recorded using Axon MultiClamp 700B amplifier. The drugs were applied via supserfusion. RESULTS Both ALL and BTHP reversibly suppressed the amplitude and density of IHERG in concentration- and voltage-dependent manners (the respective IC50 value was 49.65 and 22.38 μmol/L). BTHP (30 μmol/L) caused a significant negative shift of the steady-state inactivation curve of IHERG, while ALL (30 μmol/L) did not affect the steady-state inactivation of IHERG. Furthermore, BTHP, but not ALL, shortened the time constants of fast inactivation and slow time constants of deactivation of IHERG. But both the drugs markedly lengthened the time constants for recovery of IHERG from inactivation. Using action potential waveform pulses, it was found that both the drugs at 30 μmol/L significantly suppressed the current densities in the late phase of action potential, but did not significantly affect the current densities in the early phase of action potential. CONCLUSION Both ALL and BTHP derived from Chinese herbs potently block hERG current.
Collapse
|
4
|
Kulzer M, Seyler C, Welke F, Scherer D, Xynogalos P, Scholz EP, Thomas D, Becker R, Karle CA, Katus HA, Zitron E. Inhibition of cardiac Kir2.1–2.3 channels by beta3 adrenoreceptor antagonist SR 59230A. Biochem Biophys Res Commun 2012; 424:315-20. [DOI: 10.1016/j.bbrc.2012.06.114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 06/21/2012] [Indexed: 11/30/2022]
|
5
|
Grilo LS, Carrupt PA, Abriel H. Stereoselective Inhibition of the hERG1 Potassium Channel. Front Pharmacol 2010; 1:137. [PMID: 21833176 PMCID: PMC3153011 DOI: 10.3389/fphar.2010.00137] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 11/03/2010] [Indexed: 12/11/2022] Open
Abstract
A growing number of drugs have been shown to prolong cardiac repolarization, predisposing individuals to life-threatening ventricular arrhythmias known as Torsades de Pointes. Most of these drugs are known to interfere with the human ether à-gogo related gene 1 (hERG1) channel, whose current is one of the main determinants of action potential duration. Prolonged repolarization is reflected by lengthening of the QT interval of the electrocardiogram, as seen in the suitably named drug-induced long QT syndrome. Chirality (presence of an asymmetric atom) is a common feature of marketed drugs, which can therefore exist in at least two enantiomers with distinct three-dimensional structures and possibly distinct biological fates. Both the pharmacokinetic and pharmacodynamic properties can differ between enantiomers, as well as also between individuals who take the drug due to metabolic polymorphisms. Despite the large number of reports about drugs reducing the hERG1 current, potential stereoselective contributions have only been scarcely investigated. In this review, we present a non-exhaustive list of clinically important molecules which display chiral toxicity that may be related to hERG1-blocking properties. We particularly focus on methadone cardiotoxicity, which illustrates the importance of the stereoselective effect of drug chirality as well as individual variations resulting from pharmacogenetics. Furthermore, it seems likely that, during drug development, consideration of chirality in lead optimization and systematic assessment of the hERG1 current block with all enantiomers could contribute to the reduction of the risk of drug-induced LQTS.
Collapse
Affiliation(s)
- Liliana Sintra Grilo
- School of Pharmaceutical Sciences, University of Geneva, University of LausanneGeneva, Switzerland
- Department of Clinical Research, University of BernBern, Switzerland
| | - Pierre-Alain Carrupt
- School of Pharmaceutical Sciences, University of Geneva, University of LausanneGeneva, Switzerland
| | - Hugues Abriel
- Department of Clinical Research, University of BernBern, Switzerland
| |
Collapse
|
6
|
Polak S, Wiśniowska B, Brandys J. Collation, assessment and analysis of literature in vitro data on hERG receptor blocking potency for subsequent modeling of drugs' cardiotoxic properties. J Appl Toxicol 2009; 29:183-206. [PMID: 18988205 DOI: 10.1002/jat.1395] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The assessment of the torsadogenic potency of a new chemical entity is a crucial issue during lead optimization and the drug development process. It is required by the regulatory agencies during the registration process. In recent years, there has been a considerable interest in developing in silico models, which allow prediction of drug-hERG channel interaction at the early stage of a drug development process. The main mechanism underlying an acquired QT syndrome and a potentially fatal arrhythmia called torsades de pointes is the inhibition of potassium channel encoded by hERG (the human ether-a-go-go-related gene). The concentration producing half-maximal block of the hERG potassium current (IC(50)) is a surrogate marker for proarrhythmic properties of compounds and is considered a test for cardiac safety of drugs or drug candidates. The IC(50) values, obtained from data collected during electrophysiological studies, are highly dependent on experimental conditions (i.e. model, temperature, voltage protocol). For the in silico models' quality and performance, the data quality and consistency is a crucial issue. Therefore the main objective of our work was to collect and assess the hERG IC(50) data available in accessible scientific literature to provide a high-quality data set for further studies.
Collapse
Affiliation(s)
- Sebastian Polak
- Toxicology Department, Faculty of Pharmacy, Medical Collage, Jagiellonian University, Poland.
| | | | | |
Collapse
|
7
|
WANG XJ, YANG Q, YIN DL, CHEN YD, YOU QD. A Pharmacophore Modeling Study of Drugs Inducing Cardiotoxic Side Effects. CHINESE J CHEM 2008. [DOI: 10.1002/cjoc.200890380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
8
|
Diller DJ, Hobbs DW. Understanding hERG inhibition with QSAR models based on a one-dimensional molecular representation. J Comput Aided Mol Des 2007; 21:379-93. [PMID: 17549583 DOI: 10.1007/s10822-007-9122-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 04/29/2007] [Indexed: 10/23/2022]
Abstract
Blockage of the potassium channel encoded by the human ether-a-go-go related gene (hERG) is well understood to be the root cause of the cardio-toxicity of numerous approved and investigational drugs. As such, a cascade of in vitro and in vivo assays have been developed to filter compounds with hERG inhibitory activity. Quantitative structure activity relationship (QSAR) models are used at the very earliest part of this cascade to eliminate compounds that are likely to have this undesirable activity prior to synthesis. Here a new QSAR technique based on the one-dimensional representation is described in the context of the development of a model to predict hERG inhibition. The model is shown to perform close to the limits of the quality of the data used for model building. In order to make optimal use of the available data, a general robust mathematical scheme was developed and is described to simultaneously incorporate quantitative data, such as IC50 = 50 nM, and qualitative data, such as inactive or IC50 > 30 microM into QSAR models without discarding any experimental information.
Collapse
Affiliation(s)
- David J Diller
- Department of Molecular Modeling, Pharmacopeia Inc, CN5350, Princeton, NJ 08543-5350, USA.
| | | |
Collapse
|
9
|
Ekins S, Balakin KV, Savchuk N, Ivanenkov Y. Insights for Human Ether-a-Go-Go-Related Gene Potassium Channel Inhibition Using Recursive Partitioning and Kohonen and Sammon Mapping Techniques. J Med Chem 2006; 49:5059-71. [PMID: 16913696 DOI: 10.1021/jm060076r] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The human ether-a-go-go related gene (hERG) can be inhibited by marketed drugs, and this inhibition may lead to QT prolongation and possibly fatal cardiac arrhythmia. We have collated literature data for 99 diverse hERG inhibitors to generate Kohonen maps, Sammon maps, and recursive partitioning models. Our aim was to investigate whether these computational models could be used either individually or together in a consensus approach to predict the binding of a prospectively selected test set of 35 diverse molecules and at the same time to offer further insights into hERG inhibition. The recursive partitioning model provided a quantitative prediction, which was markedly improved when Tanimoto similarity was included as a filter to remove molecules from the test set that were too dissimilar to the training set (r2 = 0.83, Spearman rho = 0.75, p = 0.0003 for the 18 remaining molecules, >0.77 similarity). This model was also used to screen and prioritize a database of drugs, recovering several hERG inhibitors not used in model building. The mapping approaches used molecular descriptors required for hERG inhibition that were not reported previously and in particular highlighted the importance of molecular shape. The Sammon map model provided the best qualitative classification of the test set (95% correct) compared with the Kohonen map model (81% correct), and this result was also superior to the consensus approach. This study illustrates that patch clamping data from various literature sources can be combined to generate valid models of hERG inhibition for prospective predictions.
Collapse
Affiliation(s)
- Sean Ekins
- ACT LLC, 601 Runnymede Avenue, Jenkintown, Pennsylvania 19046, USA.
| | | | | | | |
Collapse
|
10
|
Diaz GJ, Daniell K, Leitza ST, Martin RL, Su Z, McDermott JS, Cox BF, Gintant GA. The [3H]dofetilide binding assay is a predictive screening tool for hERG blockade and proarrhythmia: Comparison of intact cell and membrane preparations and effects of altering [K+]o. J Pharmacol Toxicol Methods 2005; 50:187-99. [PMID: 15519905 DOI: 10.1016/j.vascn.2004.04.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Accepted: 04/06/2004] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The human ether-a-go-go-related gene (hERG) encodes a potassium channel responsible for the cardiac delayed rectifier current (IKr) involved in ventricular repolarization. Drugs that block hERG have been associated with QT interval prolongation and serious, sometimes fatal, cardiac arrhythmias (including torsade de pointes). While displacement of [3H]dofetilide, a potent methanesulfonanilide hERG blocker, from cells heterologously expressing hERG has been suggested as a screening assay, questions have been raised about its predictive value. METHODS To validate the utility of this assay as a screening tool, we performed a series of saturation and competition binding studies using [3H]dofetilide as ligand and either intact cells or membrane preparations from HEK 293 cells stably transfected with hERG K+ channels. The object of these experiments was to (1) compare binding Ki values for 22 hERG blockers using intact cells or membrane homogenates to determine whether maintaining cell integrity enhanced assay reliability; (2) evaluate the ability of different K+ concentrations (2, 5, 10, 20, and 60 mM) to modulate hERG binding; and (3) to establish the predictive value of the assay by comparing Ki values from binding studies at 5 and 60 mM [K+]o to functional IC50 values for hERG current block using 56 structurally diverse drugs. RESULTS We found (a) comparable Ki values in the intact cell and isolated membrane binding assays, although there were some differences in rank order; (b) increasing [K+]o lowered the Kd and increased the Bmax for [3H]dofetilide, particularly in the membrane assay; and (c) good correlation between binding Ki values and functional IC50 values for hERG current block. DISCUSSION In conclusion, increasing K+ concentrations results in an increase in both [3H]dofetilide affinity for hERG and available binding sites, particularly when using membrane homogenates. There are no meaningful differences between Ki values when comparing intact cell versus membrane assay, neither are there meaningful trends with increasing [K+]o within assays. There is good correlation between binding Ki values and functional (whole-cell patch clamp) IC50 values at both 5 and 60 mM K+ concentrations (R2 values of .824 and .863, respectively). The simplicity, predictability, and adaptability to high-throughput platforms make the [3H]dofetilide membrane binding assay a useful tool for screening and ranking compounds for their potential to block the hERG K+ channel.
Collapse
Affiliation(s)
- Gilbert J Diaz
- Department of Integrative Pharmacology, R46R, AP9-1, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL 60064-6119, USA.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Biliczki P, Acsai K, Virág L, Tálosi L, Jost N, Biliczki A, Papp JG, Varró A. Cellular electrophysiological effect of terikalant in the dog heart. Eur J Pharmacol 2005; 510:161-6. [PMID: 15763239 DOI: 10.1016/j.ejphar.2004.12.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Revised: 12/21/2004] [Accepted: 12/24/2004] [Indexed: 11/29/2022]
Abstract
The cellular mechanism of action of terikalant, an investigational antiarrhythmic agent known to block the inward rectifier and other potassium currents, has not yet been fully clarified. The aim of the present study was therefore to analyse the in vitro electrophysiological effects of terikalant in canine isolated ventricular muscle and Purkinje fibers by applying the standard microelectrode technique. The effects of terikalant on the duration of action potential at a stimulation cycle length of 1000 ms and on the maximum upstroke velocity of the action potential in right ventricular papillary muscle were examined at 1, 2.5, 10, and 20 microM concentrations. Terikalant significantly prolonged the action potential duration measured both at 50% and 90% of repolarization in concentration-dependent manner. The maximum upstroke velocity of the action potential was unaffected at 1 and 2.5 microM concentrations. However, this parameter was significantly reduced at 10 and 20 microM concentrations of terikalant. In Purkinje fibers terikalant (2.5 microM) also produced a marked action potential lengthening effect. Frequency dependence (cycle length of 300-5000 ms) of the action potential lengthening effect of terikalant was studied at a concentration of 2.5 microM. Prolongation of the duration of action potential occurred in a reverse frequency-dependent manner both in papillary muscle and Purkinje fibers, with a more pronounced frequency-dependence observed in Purkinje fibers. The onset kinetics of the terikalant (10 microM) induced block of the maximum upstroke velocity of the action potential was rapid (0.6+/-0.1 beat(-1), n=6) like that of Class I/B antiarrhythmics, and the offset (recovery) kinetics of the drug (2956+/-696 ms, n=6) best resembled that of Class I/A antiarrhythmic drugs. It was concluded that terikalant, unlike pure Class III antiarrhythmic drugs, has combined mode of action by lengthening repolarization and blocking the inward sodium current in a use-dependent manner.
Collapse
Affiliation(s)
- Péter Biliczki
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Clancy CE, Kass RS. Inherited and acquired vulnerability to ventricular arrhythmias: cardiac Na+ and K+ channels. Physiol Rev 2005; 85:33-47. [PMID: 15618477 DOI: 10.1152/physrev.00005.2004] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in cardiac Na(+) and K(+) channels can disrupt the precise balance of ionic currents that underlies normal cardiac excitation and relaxation. Disruption of this equilibrium can result in arrhythmogenic phenotypes leading to syncope, seizures, and sudden cardiac death. Congenital defects result in an unpredictable expression of phenotypes with variable penetrance, even within single families. Additionally, phenotypically opposite and overlapping cardiac arrhythmogenic syndromes can stem from one mutation. A number of these defects have been characterized experimentally with the aim of understanding mechanisms of mutation-induced arrhythmia. Improving understanding of abnormalities may provide a basis for the development of therapeutic approaches.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | |
Collapse
|
13
|
Kocic I, Hirano Y, Hiraoka M. The effects of K+ channels modulators terikalant and glibenclamide on membrane potential changes induced by hypotonic challenge of guinea pig ventricular myocytes. J Pharmacol Sci 2005; 95:27-32. [PMID: 15153647 DOI: 10.1254/jphs.95.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Contribution of inward rectifier K(+) currents (I(K1)) and ATP-sensitive K(+) currents (I(KATP)) to membrane potential changes of ventricular myocytes appearing during hypotonic challenge is unclear. We used here the whole cell patch clamp technique, voltage and current clamp modes, to record membrane potentials and ionic currents in isolated guinea pig ventricular myocytes under isotonic or hypotonic perfusion. The difference in osmolarity between iso- and hypotonic solutions was about 100 mOsm. Exposure to hypotonic solution for 60 s induced initial prolongation of action potential duration at 90% of repolarization (APD(90)) (from 176 +/- 10 to 189 +/- 11 ms, P<0.05, n = 13). Further perfusion for the next 300 s shorthened APD(90) to 135 +/- 9 ms (P<0.01, in comparison with control values, n = 13) and depolarized resting potential from -79.2 +/- 1.5 to -75.0 +/- 0.9 mV, (P<0.05, n = 13). Neither pretreatment with a blocker of I(K1) channels, terikalant at 10 microM, nor with a blocker of I(KATP) channels, glibenclamide at 1 microM, prevented the above-mentioned changes in membrane potential induced by hypotonic challenge when a pipette solution containing 5 mM ATP was used. Also, glibenclamide and terikalant did not affect the hypotonic-sensitive current, obtained by ramp or voltage-step protocols, respectively. Additionally, the current-voltage relationship (I-V curve) of the whole cell hypotonic-sensitive current shifted from an isotonic I-V curve in a parallel way. Our results indicate that I(K1) and I(KATP) do not participate in membrane potential changes induced by hypotonic solution at least in the guinea pig ventricular myocytes with sufficient intracellular ATP.
Collapse
Affiliation(s)
- Ivan Kocic
- Department of Cardiovascular Disease, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | |
Collapse
|
14
|
Li J, McLerie M, Lopatin AN. Transgenic upregulation ofIK1in the mouse heart leads to multiple abnormalities of cardiac excitability. Am J Physiol Heart Circ Physiol 2004; 287:H2790-802. [PMID: 15271672 DOI: 10.1152/ajpheart.00114.2004] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To assess the functional significance of upregulation of the cardiac current ( IK1), we have produced and characterized the first transgenic (TG) mouse model of IK1upregulation. To increase IK1density, a pore-forming subunit of the Kir2.1 (green fluorescent protein-tagged) channel was expressed in the heart under control of the α-myosin heavy chain promoter. Two lines of TG animals were established with a high level of TG expression in all major parts of the heart: line 1 mice were characterized by 14% heart hypertrophy and a normal life span; line 2 mice displayed an increased mortality rate, and in mice ≤1 mo old, heart weight-to-body weight ratio was increased by >100%. In adult ventricular myocytes expressing the Kir2.1-GFP subunit, IK1conductance at the reversal potential was increased ∼9- and ∼10-fold in lines 1 and 2, respectively. Expression of the Kir2.1 transgene in line 2 ventricular myocytes was heterogeneous when assayed by single-cell analysis of GFP fluorescence. Surface ECG recordings in line 2 mice revealed numerous abnormalities of excitability, including slowed heart rate, premature ventricular contractions, atrioventricular block, and atrial fibrillation. Line 1 mice displayed a less severe phenotype. In both TG lines, action potential duration at 90% repolarization and monophasic action potential at 75–90% repolarization were significantly reduced, leading to neuronlike action potentials, and the slow phase of the T wave was abolished, leading to a short Q-T interval. This study provides a new TG model of IK1upregulation, confirms the significant role of IK1in cardiac excitability, and is consistent with adverse effects of IK1upregulation on cardiac electrical activity.
Collapse
Affiliation(s)
- Jingdong Li
- Dept. of Molecular and Integrative Physiology, Univ. of Michigan, Rm. 7812 Medical Science II, 1150 W. Medical Center Dr., Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
15
|
Bains W, Basman A, White C. HERG binding specificity and binding site structure: evidence from a fragment-based evolutionary computing SAR study. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2004; 86:205-33. [PMID: 15288759 DOI: 10.1016/j.pbiomolbio.2003.09.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We describe the application of genetic programming, an evolutionary computing method, to predicting whether small molecules will block the HERG cardiac potassium channel. Models based on a molecular fragment-based descriptor set achieve an accuracy of 85-90% in predicting whether the IC(50) of a 'blind' set of compounds is <1 microM. Analysis of the models provides a 'meta-SAR', which predicts a pharmacophore of two hydrophobic features, one preferably aromatic and one preferably nitrogen-containing, with a protonatable nitrogen asymmetrically situated between them. Our experience of the approach suggests that it is robust, and requires limited scientist input to generate valuable predictive results and structural understanding of the target.
Collapse
Affiliation(s)
- William Bains
- Amedis Pharmaceuticals, Unit 162 Cambridge Science Park, Milton Road, Cambridge, UK
| | | | | |
Collapse
|
16
|
Du LP, Tsai KC, Li MY, You QD, Xia L. The pharmacophore hypotheses of IKr potassium channel blockers: novel class III antiarrhythmic agents. Bioorg Med Chem Lett 2004; 14:4771-7. [PMID: 15324906 DOI: 10.1016/j.bmcl.2004.06.070] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2004] [Accepted: 06/17/2004] [Indexed: 11/24/2022]
Abstract
Predictive pharmacophore models were developed for a large series of I(Kr) potassium channel blockers as class III antiarrhythmic agents using HypoGen in Catalyst software. The pharmacophore hypotheses were generated using a training set consisting of 34 compounds carefully selected from documents. Their biological data, expressed as IC(50), spanned from 1.5 nM to 2.8 mM with 7 orders difference. The most predictive hypothesis (Hypo1), consisting of four features (one positive ionizable feature, two aromatic rings and one hydrophobic group), had a best correlation coefficient of 0.825, a lowest rms deviation of 1.612, and a highest cost difference (null cost-total cost) of 77.552, which represents a true correlation and a good predictivity. The hypothesis Hypo1 was then validated by a test set consisting of 21 compounds and by a cross-validation of 95% confidence level with randomizing the data using CatScramble program. Accordingly, our model has strong predictivity to identify structural diverse I(Kr) potassium channel blockers with desired biological activity by virtual screening
Collapse
Affiliation(s)
- Lü-Pei Du
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | |
Collapse
|
17
|
Clancy CE, Kurokawa J, Tateyama M, Wehrens XHT, Kass RS. K+ channel structure-activity relationships and mechanisms of drug-induced QT prolongation. Annu Rev Pharmacol Toxicol 2003; 43:441-61. [PMID: 12540747 DOI: 10.1146/annurev.pharmtox.43.100901.140245] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pharmacological intervention, often for the purpose of treating syndromes unrelated to cardiac disease, can increase the vulnerability of some patients to life-threatening rhythm disturbances. This may be due to an underlying propensity stemming from genetic defects or polymorphisms, or structural abnormalities that provide a substrate allowing for the initiation of arrhythmic triggers. A number of pharmacological agents that have proven useful in the treatment of allergic reactions, gastrointestinal disorders, and psychotic disorders, among others, have been shown to reduce repolarizing K(+) currents and prolong the QT interval on the electrocardiogram. Understanding the structural determinants of K(+) channel blockade may provide new insights into the mechanism and rate-dependent effects of drugs on cellular physiology. Drug-induced disruption of cellular repolarization underlies electrocardiographic abnormalities that are diagnostic indicators of arrhythmia susceptibility.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | | | | | |
Collapse
|
18
|
Abstract
BRL-32872 is a new antiarrhythmic drug with balanced class-III and class-IV actions as categorized by the Vaughan-Williams classification. BRL-32872 blocks the rapid component of the cardiac delayed rectifier potassium channel IK(r) (IC(50) = 28 nM) and its molecular correlate HERG ("Human-ether-a-go-go related gene," IC(50) of 19.8 nM in cell lines) at low concentrations. It also inhibits the L-type calcium current (ICa) at higher concentrations (IC(50) = 2.8 microM). This dual concentration-dependent profile of action at higher concentrations may possibly prevent "torsades de pointes" ventricular arrhythmias, which is a dangerous side effect of many other class-III antiarrhythmic drugs. With BRL-32872, an excessive prolongation of the action potential duration and consecutive QTc prolongation is prevented by a concentration-dependent increase of calcium channel block, resulting in the so-called "bell-shaped" profile of antiarrhythmic drug action. BRL-32872 is very effective in the treatment of ventricular arrhythmias in animal models of cardiac ischemia. In the ischemic hearts of animals the drug significantly reduced early afterdepolarization and ventricular tachycardia. The antiarrhythmic effect of BRL-32872 has not yet been demonstrated in humans.
Collapse
Affiliation(s)
- Christoph A Karle
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | | | | |
Collapse
|
19
|
Zitron E, Karle CA, Wendt-Nordahl G, Kathöfer S, Zhang W, Thomas D, Weretka S, Kiehn J. Bertosamil blocks HERG potassium channels in their open and inactivated states. Br J Pharmacol 2002; 137:221-8. [PMID: 12208779 PMCID: PMC1573484 DOI: 10.1038/sj.bjp.0704859] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. Bertosamil is chemically related to the class-III anti-arrhythmic drug tedisamil and has been developed as a bradycardic, anti-ischemic and anti-arrhythmic drug. Its anti-arrhythmic properties might in part be attributed to its block of voltage-dependent potassium channels Kv(1.2), Kv(1.4). and Kv(1.5). However, HERG-potassium channel block as an important target for class-III drugs has not yet been investigated. 2. We investigated the effect of bertosamil on the HERG potassium channel heterologously expressed in Xenopus oocytes with the two-electrode voltage-clamp technique. 3. Bertosamil (70 microM) inhibited HERG tail currrent after a test pulse to 30 mV by 49.3+/-8.4% (n=5) and the IC(50) was 62.7 microM. Onset of block was fast, i.e. 90% of inhibition developed within 180+/-8.22 s (n=5), and block was totally reversible upon washout within 294+/-38.7 s (n=5). 4. Bertosamil-induced block of HERG potassium channels was state-dependent with block mainly to open- and inactivated channels. Half-maximal activation voltage was slightly shifted towards more negative potentials. 5. Steady-state inactivation of HERG was not influenced by bertosamil. Bertosamil block elicited voltage-but no frequency-dependent effects. 6. In summary, bertosamil blocked the HERG potassium channel. These blocking properties may contribute to the anti-arrhythmic effects of bertosamil in the treatment of atrial and particular ventricular arrhythmias.
Collapse
Affiliation(s)
- Edgar Zitron
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Christoph A Karle
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Gunnar Wendt-Nordahl
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Sven Kathöfer
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Wei Zhang
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Dierk Thomas
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Slawomir Weretka
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
| | - Johann Kiehn
- 3rd Department of Internal Medicine (Cardiology), University of Heidelberg Medical School, Heidelberg, Germany
- Author for correspondence:
| |
Collapse
|
20
|
Farkas A, Coker SJ. Limited induction of torsade de pointes by terikalant and erythromycin in an in vivo model. Eur J Pharmacol 2002; 449:143-53. [PMID: 12163118 DOI: 10.1016/s0014-2999(02)01992-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The proarrhythmic activities of the selective I(Kr) blocker erythromycin and the less selective K(+) channel blockers, terikalant and clofilium, have been compared in an alpha(1)-adrenoceptor-stimulated, anaesthetized rabbit model. Terikalant (2.5, 7.5 and 25 nmol kg(-1) min(-1); n = 10), erythromycin (133, 400 and 1330 nmol kg(-1) min(-1); n = 8), clofilium (20, 60 and 200 nmol kg(-1) min(-1); n=10) or vehicle (n = 8) was infused intravenously over 19 min and there was a 15-min interval between each infusion [corrected]. QT and QTc intervals, and epicardial monophasic action potential duration were prolonged significantly (and to a similar extent) only by clofilium and terikalant. The total incidences of torsade de pointes were 60%*, 20%, 0% and 0% in clofilium-, terikalant-, erythromycin- and vehicle-treated animals, respectively (*P < 0.05 compared to vehicle control). In conclusion, terikalant exerted mild proarrhythmic activity though it prolonged repolarisation markedly. Despite being given in high doses, erythromycin neither prolonged repolarisation nor induced proarrhythmia.
Collapse
Affiliation(s)
- András Farkas
- Department of Pharmacology and Therapeutics, The University of Liverpool, Ashton Street, Liverpool L69 3GE, UK.
| | | |
Collapse
|
21
|
Qi X, Varma P, Newman D, Mamalias N, Dorian P. Terikalant and barium decrease the area of vulnerability to ventricular fibrillation induction by T-wave shocks. J Cardiovasc Pharmacol 2002; 39:242-50. [PMID: 11791010 DOI: 10.1097/00005344-200202000-00011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The area of vulnerability (AOV) to ventricular fibrillation (VF) induction by high-voltage shocks has been proposed as a measure of vulnerability to VF. Biphasic shocks spanning the T wave and ranging between 50 V and the upper limit of vulnerability (ULV) to VF were delivered before and after terikalant (1 mg/kg) and barium (1.1 mg/kg load followed by 0.05-0.10 mg/kg/min maintenance) or vehicle in dogs. The AOV decreased by 34% and 28% (p < 0.01) after terikalant and barium (n = 8 dogs each), respectively. Mean ULV, defibrillation threshold (DFT), and ventricular vulnerability period (VVP) decreased by 16%, 23%, and 31% (p < 0.01), respectively, after terikalant, and by 25%, 17% (p < 0.01), and 13% (p = 0.08), respectively, after barium. Vehicle (n = 14) did not significantly alter any of these variables. The ULV was correlated with the DFT before and after terikalant (r = 0.78, p < 0.01) and barium (r = 0.83, p < 0.01). Potassium channel blockers of the current reduce the ability to induce VF; this effect may be related to the anti-fibrillatory action of class III anti-arrhythmic drugs and their ability to decrease DFT.
Collapse
Affiliation(s)
- Xiangqian Qi
- Division of Cardiology, Department of Medicine, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
22
|
Dumotier BM, Bastide M, Adamantidis MM. "Use-dependent" effects of cisapride on postrest action potentials in rabbit ventricular myocardium. Eur J Pharmacol 2001; 422:137-48. [PMID: 11430924 DOI: 10.1016/s0014-2999(01)01062-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Repercussions of cisapride-induced blocking effects on repolarisation of K(+) channels in open and inactivated states investigated in rabbit ventricular myocardium during rest and under stimulation were compared with effects of K(+)-blocking drugs (4-aminopyridine, dofetilide, terikalant). Major lengthening in the first postrest action potential indicates affinity for closed channels. Gradual lengthening during stimulation implies affinity for open channels. Four (control, add-in, steady-state, washout) 20-min rest periods were alternated with regular stimulation (0.5 Hz). Each drug was added during add-in and steady-state periods. Similarly to dofetilide (10 nM) and terikalant (0.3 microM), cisapride (1 microM) increasingly lengthened action potentials during stimulation, whereas 4-aminopyridine (1 mM) prolonged mostly the first postrest action potential. Our results indicate that cisapride induced use-dependent lengthening of repolarisation, compatible with an affinity for open K(+) channels. We also found that in isolated rabbit ventricular myocytes, cisapride (1-10 microM) decreased the inward rectifier K(+) current, an effect contributing to the proarrhythmic potential.
Collapse
Affiliation(s)
- B M Dumotier
- Novartis Pharma AG, Toxicology-Pathology, WSH 2881.230A, CH 4002, Bale, Switzerland
| | | | | |
Collapse
|
23
|
Brandts B, Van Bracht M, Tüttelmann F, Allessie MA, Trappe HJ. Inhibition of muscarinic potassium current by the class III antiarrhythmic drug RP58866 in guinea-pig atrial myocytes. Pacing Clin Electrophysiol 2000; 23:1812-5. [PMID: 11139931 DOI: 10.1111/j.1540-8159.2000.tb07026.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RP58866 is a potent antiarrhythmic drug that maintains its antiarrhythmic properties during ischemia. Since interstitial concentrations of adenosine increase during ischemia, we examined the properties of the drug with respect to the muscarinic K+ current (IK(ACh)), with a main emphasis on the adenosine (Ado)-induced current (IK(Ado)). Using different Gi-coupled receptors (M2, A1, sphingolipid), we studied the effect of RP58866 in isolated guinea-pig atrial myocytes by the whole-cell voltage clamp technique. Application of 50 microM RP58866 resulted in complete inhibition of the muscarinic K+ current. Inhibition was observed during activation of IK(ACh) by each of the three receptors. IC50 was approximately 2.0 microM. GTP-gamma-S induced IK(ACh) was reduced by RP58866. The drug was active from the outside only, and its intracellular application via the patch pipet had no inhibitory effect. Despite the structural homologies between inward rectifying K+ channels, the adenosine triphosphate-sensitive K+ current (IK(ATP)) was not inhibited by the compound. It is concluded that muscarinic K+ current is inhibited by RP58866, an inhibition not limited to IK1, Ito, and IKr. High interstitial adenosine concentrations during ischemia are expected to increase the participation of IK(Ado) on repolarization. RP58866-induced inhibition of IK(Ado) would, therefore, be of particular relevance during ischemia. The high sensitivity of IK(Ado) to RP58866 may partially explain the unique properties of the drug toward arrhythmias developing in the ischemic myocardium.
Collapse
Affiliation(s)
- B Brandts
- Department of Cardiology and Angiology, Ruhr-University Bochum, Hoelkeskampring 40, 44625 Herne, Germany.
| | | | | | | | | |
Collapse
|
24
|
|
25
|
Wang H, Shi H, Wang Z. Inactivation block of the HERG human cardiac K+ channels by RP58866. Br J Pharmacol 1999; 127:1899-1907. [PMID: 10482922 PMCID: PMC1566185 DOI: 10.1038/sj.bjp.0702741] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/1999] [Revised: 05/17/1999] [Accepted: 05/27/1999] [Indexed: 11/09/2022] Open
Abstract
1. RP58866 possesses a unique electrophysiological property: highly effective against various types of arrhythmias including ventricular fibrillation in animal models, noticeably those occurring during ischaemia with depolarized membrane due to elevated extracellular K+ concentrations. To understand the potential ionic mechanisms, we performed detailed studies on the effects of RP58866 on the HERG channels expressed in Xenopus oocytes, which are believed to be important compositions of the rapid component of delayed rectifier K+ current in the hearts. 2. RP58866 significantly inhibited the HERG channels in a concentration-dependent manner, with approximately 50% decrease in the current amplitude at a concentration of 1 microM. RP58866 produced more pronounced inhibition with voltage protocols which favoured inactivation of the HERG channels. It caused substantial negative shift of the inactivation curves but did not alter the activation properties. The inhibition was considerably relieved by elevating [K+]o from 5 - 20 mM, which weakened the channel inactivation. More importantly, the potency was reduced by approximately 100 fold on the mutated HERG channels (S631A) in which the C-type inactivation was substantially weakened. 4. We conclude that blockade of the HERG channels by RP58866 is mainly associated with the binding of the drugs to the inactivated channels. This unique property of HERG blockade might explain some previously reported but unexplained observations: RP58866 maintains its efficacy in APD prolongation with depolarized membrane potential and in arrhythmias during ischaemia with manifested membrane depolarization.
Collapse
Affiliation(s)
- Huizhen Wang
- Research Center, Montreal Heart Institute, 5000 Belanger East, Montreal, Quebec H1T 1C8, Canada
| | - Hong Shi
- Research Center, Montreal Heart Institute, 5000 Belanger East, Montreal, Quebec H1T 1C8, Canada
| | - Zhiguo Wang
- Research Center, Montreal Heart Institute, 5000 Belanger East, Montreal, Quebec H1T 1C8, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
26
|
Dumotier BM, Adamantidis MM, Puisieux FL, Bastide MM, Dupuis BA. Repercussions of pharmacologic reduction in ionic currents on action potential configuration in rabbit Purkinje fibers: Are they indicative of proarrhythmic potential? Drug Dev Res 1999. [DOI: 10.1002/(sici)1098-2299(199906)47:2<63::aid-ddr2>3.0.co;2-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Wang GX, Zhou XB, Eschenhagen T, Korth M. Effects of mitoxantrone on action potential and membrane currents in isolated cardiac myocytes. Br J Pharmacol 1999; 127:321-30. [PMID: 10385229 PMCID: PMC1566021 DOI: 10.1038/sj.bjp.0702547] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/1998] [Revised: 02/10/1999] [Accepted: 02/18/1999] [Indexed: 01/25/2023] Open
Abstract
1. The effects of mitoxantrone (MTO), an anticancer drug, on the membrane electrical properties of cardiac myocytes were investigated using the whole-cell clamp technique. 2. In isolated guinea-pig ventricular myocytes, 30 microM MTO induced a time-dependent prolongation of action potential duration (APD) which was occasionally accompanied by early afterdepolarizations. APD prolongation was preserved in the presence of 10 microM tetrodotoxin and showed reverse rate-dependence. 3. Both the inward rectifier K+ current (I(KI)) and the delayed rectifier K+ current (I(K)) of guinea-pig ventricular myocytes were significantly depressed by 30 microM MTO. The rapidly activating component of I(k) (I(Kr)) seemed to be preferentially blocked by MTO. The transient outward current was not affected by MTO in rat ventricular myocytes. 4. Thirty microM MTO had no direct effect on the L-type Ca2+ current (I(Ca(L))), but reversed the inhibitory effect of 1 microM carbamylcholine but not the A1-adenosine receptor agonist (-)-N6-phenylisopropyladenosine (1 microM) on I(Ca(L)) enhanced by 50 nM isoprenaline in guinea-pig ventricular myocytes. In guinea-pig atrial mycotyes, 30 microM MTO inhibited by 93% the muscarinic receptor gated K+ current (I(K,ACh)) evoked by 1 microM carbamylcholine, whereas I(K,ACh) elicited by 100 microM GTPgammaS, a nonhydrolysable GTP analogue, was only decreased by 12%. 5. The specific binding of [3H]QNB, a muscarinic receptor ligand, to human atrial membranes was concentration-dependently displaced by MTO (1-1000 microM). 6. In conclusion, MTO blocks cardiac muscarinic receptors and prolongs APD by inhibition of I(KI) and I(Kr). The occasionally observed early afterdepolarizations may signify a potential cardiac hazard of the drug.
Collapse
Affiliation(s)
- Ge-Xin Wang
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universitäts-Krankenhaus Eppendorf, Martinistrasse 52, D-20251 Hamburg, Germany
| | - Xiao-Bo Zhou
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universitäts-Krankenhaus Eppendorf, Martinistrasse 52, D-20251 Hamburg, Germany
| | - Thomas Eschenhagen
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universitäts-Krankenhaus Eppendorf, Martinistrasse 52, D-20251 Hamburg, Germany
| | - Michael Korth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universitäts-Krankenhaus Eppendorf, Martinistrasse 52, D-20251 Hamburg, Germany
| |
Collapse
|
28
|
Jones SE, Kasamaki Y, Ogura T, Shuba LM, McCullough JR, McDonald TF. Inhibition of cardiac inward-rectifier K+ current by terodiline. Eur J Pharmacol 1999; 370:319-27. [PMID: 10334509 DOI: 10.1016/s0014-2999(99)00130-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The antispasmodic agent terodiline has cardiotoxic effects that include QT lengthening. To determine whether inhibition of inwardly-rectifying K+ current (I(K1)) might be a factor in the cardiotoxicity, we measured I(K1) in guinea pig ventricular myocytes. Terodiline reduced outward I(K1) with an IC50 of 7 microM; maximal reduction was 60% with 100-300 microM concentration. Inhibition was independent of current direction, and persisted after removal of the drug. Terodiline (3-5 microM) lengthened action potentials in guinea pig papillary muscles by ca. 10%, primarily by slowing phase 3 repolarization; higher concentrations abbreviated the plateau and markedly slowed late repolarization. Terodiline washout provoked an extra lengthening, consistent with persistent inhibition of I(K1) and rapid recovery of net inward plateau current. The results suggest that inhibition of I(K1) is a likely factor in the cardiotoxicity of the drug.
Collapse
Affiliation(s)
- S E Jones
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Williams BA, Dickenson DR, Beatch GN. Kinetics of rate-dependent shortening of action potential duration in guinea-pig ventricle; effects of IK1 and IKr blockade. Br J Pharmacol 1999; 126:1426-36. [PMID: 10217537 PMCID: PMC1565915 DOI: 10.1038/sj.bjp.0702443] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The kinetics of shortening of action potential duration (APD) following an increase in pacing rate, from 2 to 3.3 Hz, was characterized in guinea-pig ventricular preparations. Terikalant (RP62719), an inhibitor of the inwardly rectifying K+ current (IK1), and dofetilide, a specific inhibitor of the rapidly activating delayed-rectifier current (IKr), were applied to determine the effect of inhibition of these ion currents on slow APD shortening. 2. Action potentials were recorded from isolated guinea-pig ventricular myocytes using the perforated-patch patch-clamp technique, and monophasic action potentials were recorded from Langendorff-perfused guinea-pig ventricles using a contact epicardial probe. 3. Under control conditions, after an increase in pacing rate, APD immediately decreased, and then shortened slowly with an exponential time course. In ventricular myocytes, the time constant of this exponential shortening was 28+/-4 s and the amount of slow shortening was 21.9+/-0.9 ms (n=8) for an increase in rate from 2 to 3.3 Hz. Similar values were observed in Langendorff-perfused ventricles. 4. Terikalant dose-dependently increased APD and the increase was enhanced by rapid pacing ('positive' rate-dependence). The drug dose-dependently decreased the time constant of shortening and amount of slow APD shortening. In contrast, dofetilide, an inhibitor of IKr, which shows 'reverse' rate-dependent APD widening, had no significant effect on the time constant or amount of slow shortening. 5. These observations suggest that IK1 plays a role in rate-dependent shortening of APD. The results appear to support the hypothesis that 'reverse' rate-dependent effects of IKr blockers are due to these drugs not affecting the ion current(s) mediating intrinsic rate-dependent slow shortening of APD.
Collapse
Affiliation(s)
- B A Williams
- Division of Cardiology, Faculty of Medicine, University of Ottawa Heart Institute, Ontario, Canada
| | | | | |
Collapse
|
30
|
Nuss HB, Marbán E, Johns DC. Overexpression of a human potassium channel suppresses cardiac hyperexcitability in rabbit ventricular myocytes. J Clin Invest 1999; 103:889-96. [PMID: 10079110 PMCID: PMC408140 DOI: 10.1172/jci5073] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/1998] [Accepted: 02/09/1999] [Indexed: 11/17/2022] Open
Abstract
The high incidence of sudden death in heart failure may reflect abnormalities of repolarization and heightened susceptibility to arrhythmogenic early afterdepolarizations (EADs). We hypothesized that overexpression of the human K+ channel HERG (human ether-a-go-go-related gene) could enhance repolarization and suppress EADs. Adult rabbit ventricular myocytes were maintained in primary culture, which suffices to prolong action potentials and predisposes to EADs. To achieve efficient gene transfer, we created AdHERG, a recombinant adenovirus containing the HERG gene driven by a Rous sarcoma virus (RSV) promoter. The virally expressed HERG current exhibited pharmacologic and kinetic properties like those of native IKr. Transient outward currents in AdHERG-infected myocytes were similar in magnitude to those in control cells, while stimulated action potentials (0.2 Hz, 37 degrees C) were abbreviated compared with controls. The occurrence of EADs during a train of action potentials was reduced by more than fourfold, and the relative refractory period was increased in AdHERG-infected myocytes compared with control cells. Gene transfer of delayed rectifier potassium channels represents a novel and effective strategy to suppress arrhythmias caused by unstable repolarization.
Collapse
Affiliation(s)
- H B Nuss
- Section of Molecular and Cellular Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
31
|
Abstract
Virtually all clinical antiarrhythmic agents act by reducing ion channel conductance, with sodium (Na+), potassium (K+), and calcium (Ca++) channels the primary targets. Na+ channel blockers increase the risk of ischemic ventricular fibrillation and are relatively contraindicated in the presence of active coronary heart disease. Ca++ channel blockers suppress AV nodal conduction and are used to terminate reentrant supraventricular arrhythmias and control the ventricular response to atrial fibrillation. K+ channels constitute the most diverse group of cardiac ion channels. They are the primary targets of Class III antiarrhythmic drugs, the category of such agents presently undergoing the most active development. The rapid delayed rectifier, IKr, plays a key role in repolarization of all cardiac tissues and is the most common (and often only) target of action potential-prolonging drugs. Unfortunately, because of the ubiquity of IKr and the reverse use-dependent action potential prolongation that results from blocking it, IKr blockers are likely to cause torsades de pointes ventricular proarrhythmia. K+ channel blockers, such as amiodarone and azimilide, that affect the slow delayed rectifier IKs as well as IKr, appear to produce a more desirable rate-dependent profile of Class III action. Recently, much has been learned about the molecular basis of K+ channels based on their role in the congenital long QT syndrome. The availability of molecular clones that encode many of the channels in the human heart allows for the rapid screening of many potential new drugs, making possible the development of "designer" antiarrhythmic drugs with specific profiles of channel-blocking selectivity.
Collapse
Affiliation(s)
- S Nattel
- Research Center and Department of Medicine, Montreal Heart Institute, University of Montreal, Quebec, Canada.
| |
Collapse
|
32
|
Carmeliet E, Mubagwa K. Antiarrhythmic drugs and cardiac ion channels: mechanisms of action. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 1998; 70:1-72. [PMID: 9785957 DOI: 10.1016/s0079-6107(98)00002-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this review a description and an analysis are given of the interaction of antiarrhythmic drugs with their molecular target, i.e. ion channels and receptors. Our approach is based on the concept of vulnerable parameter, i.e. the electrophysiological property which plays a crucial role in the genesis of arrhythmias. To prevent or stop the arrhythmia a drug should modify the vulnerable parameter by its action on channel or receptor targets. In the first part, general aspects of the interaction between drugs channel molecules are considered. Drug binding depends on the state of the channel: rested, activated pre-open, activated open, or inactivated state. The change in channel behaviour with state is presented in the framework of the modulated-receptor hypothesis. Not only inhibition but also stimulation can be the result of drug binding. In the second part a detailed and systematic description and an analysis are given of the interaction of drugs with specific channels (Na+, Ca2+, K+, "pacemaker") and non-channel receptors. Emphasis is given to the type of state-dependent block involved (rested, activated and inactivated state block) and the change in channel kinetics. These properties vary and determine the voltage- and frequency-dependence of the change in ionic current. Finally, the question is asked as to whether the available drugs by their action on channels and receptors modify the vulnerable parameter in the desired way to stop or prevent arrhythmias.
Collapse
Affiliation(s)
- E Carmeliet
- Centre for Experimental Surgery and Anaesthesiology, University of Leuven, Belgium.
| | | |
Collapse
|
33
|
Rials SJ, Xu X, Wu Y, Marinchak RA, Kowey PR. Regression of LV hypertrophy with captopril normalizes membrane currents in rabbits. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:H1216-24. [PMID: 9746469 DOI: 10.1152/ajpheart.1998.275.4.h1216] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies indicate that regression of left ventricular hypertrophy (LVH) normalizes the in situ electrophysiological abnormalities of the left ventricle. This study was designed to determine whether regression of LVH also normalizes the abnormalities of individual membrane currents. LVH was induced in rabbits by renal artery banding. Single ventricular myocytes from rabbits with LVH at 3 mo after renal artery banding demonstrated increased cell membrane capacitance, prolonged action potential duration, decreased inward rectifier K+ current density, and increased transient outward K+ current density compared with myocytes from age-matched controls. Additional rabbits were randomized at 3 mo after banding to treatment with either vehicle or captopril for an additional 3 mo. Myocytes from LVH rabbits treated with vehicle showed persistent membrane current abnormalities. However, myocytes isolated from LVH rabbits treated with captopril had normal cell membrane capacitance, action potential duration, and membrane current densities. Captopril had no direct effect on membrane currents of either control or LVH myocytes. These data support the hypothesis that the action potential prolongation and membrane current abnormalities of LVH are reversed by regression. Normalization of membrane currents probably explains the reduced vulnerability to ventricular arrhythmia observed in this LVH model after treatment with captopril.
Collapse
Affiliation(s)
- S J Rials
- Cardiovascular Division, The Lankenau Hospital and Medical Research Center, Wynnewood 19096, USA
| | | | | | | | | |
Collapse
|
34
|
Salata JJ, Jurkiewicz NK, Wang J, Evans BE, Orme HT, Sanguinetti MC. A novel benzodiazepine that activates cardiac slow delayed rectifier K+ currents. Mol Pharmacol 1998; 54:220-30. [PMID: 9658209 DOI: 10.1124/mol.54.1.220] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The slowly activating delayed rectifier K+ current, IKs, is an important modulator of cardiac action potential repolarization. Here, we describe a novel benzodiazepine, [L-364,373 [(3-R)-1, 3-dihydro-5-(2-fluorophenyl)-3-(1H-indol-3-ylmethyl)-1-methyl-2H- 1,4-benzodiazepin-2-one] (R-L3), that activates IKs and shortens action potentials in guinea pig cardiac myocytes. These effects were additive to isoproterenol, indicating that channel activation by R-L3 was independent of beta-adrenergic receptor stimulation. The increase of IKs by R-L3 was stereospecific; the S-enantiomer, S-L3, blocked IKs at all concentrations examined. The increase in IKs by R-L3 was greatest at voltages near the threshold for normal channel activation, caused by a shift in the voltage dependence of IKs activation. R-L3 slowed the rate of IKs deactivation and shifted the half-point of the isochronal (7.5 sec) activation curve for IKs by -16 mV at 0.1 microM and -24 mV at 1 microM. R-L3 had similar effects on cloned KvLQT1 channels expressed in Xenopus laevis oocytes but did not affect channels formed by coassembly of KvLQT1 and hminK subunits. These findings indicate that the association of minK with KvLQT1 interferes with the binding of R-L3 or prevents its action once bound to KvLQT1 subunits.
Collapse
Affiliation(s)
- J J Salata
- Department of Pharmacology, Merck Research Laboratories, West Point, Pennsylvania 19486, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Taglialatela M, Castaldo P, Pannaccione A, Giorgio G, Annunziato L. Human ether-a-gogo related gene (HERG) K+ channels as pharmacological targets: present and future implications. Biochem Pharmacol 1998; 55:1741-6. [PMID: 9714291 DOI: 10.1016/s0006-2952(98)00002-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Electrophysiological and molecular biology techniques have widely expanded our knowledge of the diverse functions where K+ channels are implicated as potential and proven pharmacological targets. The aim of the present commentary is to review the recent progress in the understanding of the functional role of the K+ channels encoded by the human ether-a-gogo related gene (HERG), with particular emphasis on their direct pharmacological modulation by drugs, or on their regulation by pharmacologically relevant phenomena. About 3 years have passed since the cloning, expression, and description of the pathophysiological role of HERG K+ channels in human cardiac repolarization. Despite this short lapse of time, these K+ channels have already gained considerable attention as pharmacological targets. In fact, interference with HERG K+ channels seems to be the main mechanism explaining both the therapeutic actions of the class III antiarrhythmics and the potential cardiotoxicity of second-generation H1 receptor antagonists such as terfenadine and astemizole, as well as of psychotropic drugs such as some antidepressants and neuroleptics. It seems possible to anticipate that the main tasks for future investigation will be, on the one side, the better understanding of the intimate mechanism of action of HERG K+ channel-blocking drugs in order to elucidate the conditions regulating the delicate balance between antiarrhythmic and proarrhythmic potential and, on the other, to unravel the pathophysiological role of this K+ channel in the function of the brain and of other excitable tissues.
Collapse
Affiliation(s)
- M Taglialatela
- Department of Neuroscience, School of Medicine, University of Naples Federico II, Italy.
| | | | | | | | | |
Collapse
|
36
|
Busch AE, Eigenberger B, Jurkiewicz NK, Salata JJ, Pica A, Suessbrich H, Lang F. Blockade of HERG channels by the class III antiarrhythmic azimilide: mode of action. Br J Pharmacol 1998; 123:23-30. [PMID: 9484850 PMCID: PMC1565134 DOI: 10.1038/sj.bjp.0701575] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
1. The class III antiarrhythmic azimilide has previously been shown to inhibit I(Ks) and I(Kr) in guinea-pig cardiac myocytes and I(Ks) (minK) channels expressed in Xenopus oocytes. Because HERG channels underly the conductance I(Kr), in human heart, the effects of azimilide on HERG channels expressed in Xenopus oocytes were the focus of the present study. 2. In contrast to other well characterized HERG channel blockers, azimilide blockade was reverse use-dependent, i.e., the relative block and apparent affinity of azimilide decreased with an increase in channel activation frequency. Azimilide blocked HERG channels at 0.1 and 1 Hz with IC50s of 1.4 microM and 5.2 microM respectively. 3. In an envelope of tail test, HERG channel blockade increased with increasing channel activation, indicating binding of azimilide to open channels. 4. Azimilide blockade of HERG channels expressed in Xenopus oocytes and I(Kr) in mouse AT-1 cells was decreased under conditions of high [K+]e, whereas block of slowly activating I(Ks) channels was not affected by changes in [K+]e. 5. In summary, azimilide is a blocker of cardiac delayed rectifier channels, I(Ks) and HERG. Because of the distinct effects of stimulation frequency and [K+]e on azimilide block of I(Kr) and I(Ks) channels, we conclude that the relative contribution of block of each of these cardiac delayed rectifier channels depends on heart frequency. [K+]e and regulatory status of the respective channels.
Collapse
Affiliation(s)
- A E Busch
- Institute of Physiology I, Eberhard-Karls University of Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Lakhe M, Furukawa Y, Yonezawa T, Nagashima Y, Chiba S. Chronotropic and inotropic effects of terikalant on isolated, blood-perfused atrial and ventricular preparations of dogs. Fundam Clin Pharmacol 1997; 11:331-8. [PMID: 9263763 DOI: 10.1111/j.1472-8206.1997.tb00846.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We investigated the effects of terikalant, which blocks inward rectifier K+ current, on the sinus rate, atrial and ventricular contractile force in the isolated, blood-perfused right atrial and left ventricular preparations of dogs, and the effects of terikalant on the negative cardiac responses to acetylcholine, adenosine or pinacidil (an ATP-sensitive K+ channel opener) and on the positive cardiac responses to norepinephrine. Terikalant (1-100 nmol) decreased sinus rate and briefly and slightly increased atrial contractile force in isolated atria. However, terikalant did not increase ventricular contractile force in isolated ventricles. Neither propranolol nor atropine inhibited the positive inotropic and negative chronotropic responses to terikalant, respectively. Terikalant (10 or 30 nmol) did not significantly affect the negative cardiac responses to acetylcholine, adenosine nor pinacidil and the positive responses to norepinephrine. These results suggest that terikalant decreases sinus rate with a small changes in myocardial contractile force and does not affect the cardiac responses to muscarinic and adenosine receptor agonists, ATP-sensitive K+ channel openers nor beta-adrenoceptor agonists in the dog heart.
Collapse
Affiliation(s)
- M Lakhe
- Department of Pharmacology, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | | | |
Collapse
|