Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Neth Heart J ; 18(3): 165-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20390067

ABSTRACT

The risk for lethal ventricular arrhythmias is increased in individuals who carry mutations in genes that encode cardiac ion channels. Loss-of-function mutations in SCN5A, the gene encoding the cardiac sodium channel, are linked to Brugada syndrome (BrS). Arrhythmias in BrS are often preceded by coved-type ST-segment elevation in the right-precordial leads V1 and V2. Loss-of-function mutations in KCNH2, the gene encoding the cardiac ion channel that is responsible for the rapidly activating delayed rectifying potassium current, are linked to long-QT syndrome type 2 (LQT-2). LQT-2 is characterised by delayed cardiac repolarisation and rate-corrected QT interval (QTc) prolongation. Here, we report that the risk for ventricular arrhythmias in BrS and LQT-2 is further increased during fever. Moreover, we demonstrate that fever may aggravate coved-type ST-segment elevation in BrS, and cause QTc lengthening in LQT-2. Finally, we describe molecular mechanisms that may underlie the proarrhythmic effects of fever in BrS and LQT-2. (Neth Heart J 2010;18:165-9.).

2.
Br J Pharmacol ; 153(3): 439-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17965736

ABSTRACT

BACKGROUND AND PURPOSE: Many drugs associated with acquired long QT syndrome (LQTS) directly block human ether-a-go-go-related gene (hERG) K(+) channels. Recently, disrupted trafficking of the hERG channel protein was proposed as a new mechanism underlying LQTS, but whether this defect coexists with the hERG current block remains unclear. This study investigated how ketoconazole, a direct hERG current inhibitor, affects the trafficking of hERG channel protein. EXPERIMENTAL APPROACH: Wild-type hERG and SCN5A/hNa(v) 1.5 Na(+) channels or the Y652A and F656C mutated forms of the hERG were stably expressed in HEK293 cells. The K(+) and Na(+) currents were recorded in these cells by using the whole-cell patch-clamp technique (23 degrees C). Protein trafficking of the hERG was evaluated by Western blot analysis and flow cytometry. KEY RESULTS: Ketoconazole directly blocked the hERG channel current and reduced the amount of hERG channel protein trafficked to the cell surface in a concentration-dependent manner. Current density of the hERG channels but not of the hNa(v) 1.5 channels was reduced after 48 h of incubation with ketoconazole, with preservation of the acute direct effect on hERG current. Mutations in drug-binding sites (F656C or Y652A) of the hERG channel significantly attenuated the hERG current blockade by ketoconazole, but did not affect the disruption of trafficking. CONCLUSIONS AND IMPLICATIONS: Our findings indicate that ketoconazole might cause acquired LQTS via a direct inhibition of current through the hERG channel and by disrupting hERG protein trafficking within therapeutic concentrations. These findings should be considered when evaluating new drugs.


Subject(s)
Antifungal Agents/adverse effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ketoconazole/adverse effects , Long QT Syndrome/chemically induced , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Binding Sites , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Flow Cytometry , Humans , Ketoconazole/administration & dosage , Ketoconazole/pharmacology , Muscle Proteins/metabolism , Mutation , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Protein Transport/drug effects , Sodium Channels/metabolism , Time Factors
3.
Br J Pharmacol ; 149(5): 481-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16967046

ABSTRACT

BACKGROUND AND PURPOSE: Fluoxetine (Prozac) is a widely prescribed drug in adults and children, and it has an active metabolite, norfluoxetine, with a prolonged elimination time. Although uncommon, Prozac causes QT interval prolongation and arrhythmias; a patient who took an overdose of Prozac exhibited a prolonged QT interval (QTc 625 msec). We looked for possible mechanisms underlying this clinical finding by analysing the effects of fluoxetine and norfluoxetine on ion channels in vitro. EXPERIMENTAL APPROACH: We studied the effects of fluoxetine and norfluoxetine on the electrophysiology and cellular trafficking of hERG K+ and SCN5A Na+ channels heterologously expressed in HEK293 cells. KEY RESULTS: Voltage clamp analyses employing square pulse or ventricular action potential waveform protocols showed that fluoxetine and norfluoxetine caused direct, concentration-dependent, block of hERG current (IhERG). Biochemical studies showed that both compounds also caused concentration-dependent reductions in the trafficking of hERG channel protein into the cell surface membrane. Fluoxetine had no effect on SCN5A channel or HEK293 cell endogenous current. Mutations in the hERG channel drug binding domain reduced fluoxetine block of IhERG but did not alter fluoxetine's effect on hERG channel protein trafficking. CONCLUSIONS AND IMPLICATIONS: Our findings show that both fluoxetine and norfluoxetine at similar concentrations selectively reduce IhERG by two mechanisms, (1) direct channel block, and (2) indirectly by disrupting channel protein trafficking. These two effects are not mediated by a single drug binding site. Our findings add complexity to understanding the mechanisms that cause drug-induced long QT syndrome.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Fluoxetine/adverse effects , Long QT Syndrome/chemically induced , Adult , Antidepressive Agents, Second-Generation/adverse effects , Antidepressive Agents, Second-Generation/pharmacology , Blotting, Western , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/physiology , Cisapride/pharmacology , Dose-Response Relationship, Drug , Drug Overdose , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Female , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Humans , Long QT Syndrome/metabolism , Long QT Syndrome/physiopathology , Membrane Potentials/drug effects , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation/genetics , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Piperidines/pharmacology , Protein Transport/drug effects , Pyridines/pharmacology , Sodium Channels/genetics , Sodium Channels/metabolism , Time Factors , Transfection
4.
Handb Exp Pharmacol ; (171): 349-55, 2006.
Article in English | MEDLINE | ID: mdl-16610352

ABSTRACT

The human ether-a-go-go-related gene (hERG) encodes an ion channel subunit underlying IKr, a potassium current required for the normal repolarization of ventricular cells in the human heart. Mutations in hERG cause long QT syndrome (LQTS) by disrupting IKr, increasing cardiac excitability and, in some cases, triggering catastrophic torsades de pointes arrhythmias and sudden death. More than 200 putative disease-causing mutations in hERG have been identified in affected families to date, but the mechanisms by which these mutations cause disease are not well understood. Of the mutations studied, most disrupt protein maturation and reduce the numbers of hERG channels at the membrane. Some trafficking-defective mutants can be rescued by pharmacological agents or temperature. Here we review evidence for rescue of mutant hERG subunits expressed in heterologous systems and discuss the potential for therapeutic approaches to correcting IKr defects associated with LQTS.


Subject(s)
Ether-A-Go-Go Potassium Channels/physiology , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Mutation , Animals , ERG1 Potassium Channel , Humans , Protein Transport
5.
Eur J Pharmacol ; 430(1): 147-8, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11698075

ABSTRACT

The pharmacological characteristics of [3H]dofetilide binding were examined in membranes prepared from human embryonic kidney (HEK293) cells stably expressing human ether-á-go-go related gene (HERG) K+ channels. The classIII antiarrhythmic compounds dofetilide, clofilium, 4'-[[1-[2-(6-methyl-2-pyridyl)ethyl]-4-piperidyl]carbonyl]methanesulfonanilide (E-4031), N-methyl-N-[2-[methyl-(1-methyl-1H-benzimidazol-2-yl)amino]ethyl]-4-[(methylsulfonyl)amino]benzene-sulfonamide (WAY-123,398) and d-sotalol all inhibited [3H]dofetilide binding. In addition, the structurally unrelated compounds pimozide, terfenadine and haloperidol, all of which prolong the QT interval in man, also inhibited binding. These data indicate that a [3H]dofetilide binding assay using HERG membranes may help identify compounds that prolong the QT interval.


Subject(s)
Anti-Arrhythmia Agents/metabolism , Cation Transport Proteins , Cell Membrane/metabolism , DNA-Binding Proteins , Phenethylamines/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Sulfonamides/metabolism , Trans-Activators , Benzimidazoles/pharmacology , Binding, Competitive , Cell Line , Drug Evaluation, Preclinical/methods , ERG1 Potassium Channel , Electrocardiography , Ether-A-Go-Go Potassium Channels , Haloperidol/pharmacology , Humans , Patch-Clamp Techniques , Pimozide/pharmacology , Potassium Channel Blockers , Potassium Channels/genetics , Quaternary Ammonium Compounds/pharmacology , Sulfanilamides/pharmacology , Terfenadine/pharmacology , Transcriptional Regulator ERG , Transfection , Tritium
6.
Mol Pharmacol ; 59(5): 1069-76, 2001 May.
Article in English | MEDLINE | ID: mdl-11306689

ABSTRACT

Cocaine causes cardiac arrhythmias, sudden death, and occasionally long QT syndrome in humans. We investigated the effect of cocaine on the human K(+) channels HERG and KvLQT1+minK that encode native rapidly (I(Kr)) and slowly (I(Ks)) activating delayed rectifier K(+) channels in the heart. HERG and KvLQT1+minK channels were heterologously expressed in human embryonic kidney 293 cells, and whole-cell currents were recorded. Cocaine had no effect on KvLQT1+minK current in concentrations up to 200 microM. In contrast, cocaine reversibly blocked HERG current with half-maximal block of peak tail current of 7.2 microM. By using a protocol to quickly activate HERG channels, we found that cocaine block developed rapidly after channel activation. At 0 mV, the time constants for the development of block were 38.2 +/- 2.1, 15.2 +/- 0.8, and 6.9 +/- 1.1 ms in 10, 50 and 200 microM cocaine, respectively. Cocaine-blocked channels also recovered rapidly from block after repolarization. At -100 mV, recovery from block followed a biphasic time course with fast and slow time constants of 3.5 +/- 0.7 and 100.3 +/- 15.4 ms, respectively. Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. These results indicate that cocaine suppresses HERG, but not KvLQT1+minK, channels by preferentially blocking activated channels, that it unblocks upon repolarization, and does so with unique ultrarapid kinetics. Because the cocaine concentration range we studied is achieved in humans, HERG block may provide an additional mechanism for cocaine-induced arrhythmias and sudden death.


Subject(s)
Cation Transport Proteins , Cocaine/pharmacology , DNA-Binding Proteins , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Cells, Cultured , Cocaine/analogs & derivatives , Cocaine/chemistry , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Patch-Clamp Techniques , Transcriptional Regulator ERG , Transfection , Vasoconstrictor Agents/chemistry , Vasoconstrictor Agents/pharmacology
7.
Br J Pharmacol ; 132(1): 85-92, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11156564

ABSTRACT

1. Long OT syndrome has many causes from both acquired and congenital disorders. For the congenital disorders, their presentation and disease course are not identical. We studied two pharmacological models of long QT syndrome (LQT) to identify differences in cellular electrophysiological properties that may account for this. LQT2 was simulated by suppression of the rapidly activating delayed rectifier potassium current (I(Kr)) with the drug E-4031, and LQT3 was simulated by slowing of the sodium current (I(Na)) decay with the toxin ATX II. 2. Single rabbit ventricular cell action potentials were studied using the amphotericin B perforated patch clamp technique. Action potential and early afterdepolarization (EAD) properties were rigorously defined by the frequency power spectra obtained with fast Fourier transforms. 3. The E-4031 (n=43 myocytes) and ATX II (n=50 myocytes) models produced different effects on action potential and EAD properties. The major differences are that ATX II, compared with E-4031, caused greater action potential prolongation, more positive plateau voltages, lower amplitude EADs with less negative take-off potentials, greater time to the EAD peak voltage, and longer duration EADs. Despite causing greater action potential prolongation, the incidence of EAD induction was much less with the ATX II model (28%) than with the E-4031 model (84%). Thus these two pharmacological models have strikingly different cellular electrophysiological properties. 4. Our findings provide cellular mechanisms that may account for some differences in the clinical presentation of LQT2 and LQT3.


Subject(s)
Long QT Syndrome/physiopathology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Cell Separation , Electric Stimulation , Electrophysiology , Fourier Analysis , Heart/physiopathology , In Vitro Techniques , Long QT Syndrome/chemically induced , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium Channel Blockers , Pyridines/pharmacology , Rabbits , Tetrodotoxin/toxicity
8.
Methods Find Exp Clin Pharmacol ; 22(2): 67-76, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10849888

ABSTRACT

The effects of class III antiarrhythmic agents E-4031 and dofetilide were studied on action potentials and subthreshold delayed afterdepolarizations (DADs) induced by the cardiac glycoside acetylstrophanthidin (AS) in isolated cardiac Purkinje fibers. Action potentials were recorded from cardiac Purkinje fibers using microelectrode techniques. E-4031 and dofetilide consistently increased DAD amplitude, occasionally caused triggered action potentials and shortened action potential duration. The application of E-4031 without prior AS exposure, resulted in the typical class III antiarrhythmic effects of action potential lengthening and the induction of early afterdepolarizations. These findings suggest that under our conditions of AS-induced cell Ca2+ overload, the effects of the "pure" class III antiarrhythmic drugs, E-4031 and dofetilide, are markedly different from those found in non-Ca2+ loaded cells. This may represent an additional electrophysiological mechanism for class III antiarrhythmic drug toxicity.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Phenethylamines/pharmacology , Piperidines/pharmacology , Purkinje Fibers/drug effects , Pyridines/pharmacology , Sulfonamides/pharmacology , Action Potentials/drug effects , Animals , Purkinje Fibers/physiology , Sheep , Time Factors
9.
Mol Pharmacol ; 57(1): 101-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617684

ABSTRACT

Flecainide block of Na(+) current (I(Na)) was investigated in wild-type (WT) or the long QT syndrome 3 (LQT3) sodium channel alpha subunit mutation with three amino acids deleted (DeltaKPQ) stably transfected into human embryonic kidney 293 cells using whole-cell, patch-clamp recordings. Flecainide (1-300 mM) caused tonic and use-dependent block (UDB) of I(Na) in a concentration-dependent manner. Compared with WT, DeltaKPQ I(Na) was more sensitive to flecainide, and flecainide preferentially inhibited late I(Na) (mean current between 20 and 23.5 ms after depolarization) compared with peak I(Na). The IC(50) value of peak and late I(Na) for WT was 127 +/- 6 and 44 +/- 2 microM (n = 20) and for DeltaKPQ was 80 +/- 9 and 19 +/- 2 microM (n = 31) respectively. UDB of peak I(Na) was greater and developed more slowly during pulse trains for DeltaKPQ than for WT. The IC(50) value for UDB of peak I(Na) for WT was 29 +/- 4 microM (n = 20) and for DeltaKPQ was 11 +/- 1 microM (n = 26). For DeltaKPQ, UDB of late I(Na) was greater than for peak I(Na). Recovery from block was slower for DeltaKPQ than for WT. We conclude that DeltaKPQ interacts differently with flecainide than with WT, leading to increased block and slowed recovery, especially for late I(Na). These data provide insights into mechanisms for flecainide block and provide a rationale at the cellular and molecular level that open channel block may be a useful pharmacological property for treatment of LQT3.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Long QT Syndrome/metabolism , Sodium Channel Blockers , Binding Sites , Cells, Cultured , Electrocardiography/drug effects , Electrophysiology , Humans , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Models, Molecular , Mutation , Quality Control , Sodium Channels/genetics , Sodium Channels/physiology
10.
J Cardiovasc Electrophysiol ; 11(12): 1413-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11196567

ABSTRACT

LQT2 is one form of the congenital long QT syndrome. It results from mutations in the human ether-a-go-go-related gene (HERG), and more than 80 mutations, usually causing single amino acid substitutions in the HERG protein, are known. HERG encodes the ion channel pore-forming subunit protein for the rapidly activating delayed rectifier K+ channel (I(Kr)) in the heart. This review summarizes current findings about mutations causing LQT2, the mechanisms by which mutations may cause the clinical phenotype of a reduction in I(Kr) and a prolonged QT interval, and how this may be involved in the generation of ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cation Transport Proteins , DNA-Binding Proteins , Long QT Syndrome/physiopathology , Potassium Channels, Voltage-Gated , Trans-Activators , Amino Acid Substitution , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/therapy , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Genes, Dominant , Humans , Long QT Syndrome/complications , Long QT Syndrome/genetics , Long QT Syndrome/therapy , Mutation , Potassium/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Transport/genetics , Temperature , Transcriptional Regulator ERG
11.
J Biol Chem ; 274(44): 31123-6, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10531299

ABSTRACT

The chromosome 7-linked form of congenital long QT syndrome (LQT2) is caused by mutations in the human ether-a-go-go-related gene (HERG) that encodes the rapidly activating delayed rectifier potassium channel. One mechanism for the loss of normal channel function in LQT2 is defective protein trafficking, which results in the failure of the channel protein to reach the plasma membrane. Here we show that the N470D LQT2 mutant protein is trafficking-deficient when expressed at 37 degrees C in HEK293 cells, whereas at 27 degrees C its trafficking to the plasma membrane and channel function are markedly improved. We further show that the antiarrhythmic drug E-4031, which selectively blocks HERG channels, also corrects defective protein trafficking of the N470D mutant and can restore the generation of HERG current. Similar findings were obtained with the drugs astemizole and cisapride, as well as with high concentrations of glycerol. The effect of E-4031 on HERG protein trafficking was concentration-dependent and required low drug concentrations (saturation present at 5 microM), developed rapidly with drug exposure, and occurred post-translationally. These findings suggest that protein misfolding leading to defective trafficking of some HERG LQT mutations may be corrected by specific pharmacological strategies.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Long QT Syndrome/congenital , Long QT Syndrome/genetics , Mutation , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Anti-Arrhythmia Agents/pharmacology , Astemizole/pharmacology , Biological Transport/drug effects , Cell Compartmentation , Cisapride/pharmacology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Glycerol/pharmacology , Humans , Long QT Syndrome/drug therapy , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium Channel Blockers , Potassium Channels/genetics , Protein Folding , Pyridines/pharmacology , Recombinant Proteins/drug effects , Temperature , Transcriptional Regulator ERG
12.
J Cardiovasc Electrophysiol ; 10(6): 836-43, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10376921

ABSTRACT

INTRODUCTION: The selective H1-receptor antagonist astemizole (Hismanal) causes acquired long QT syndrome. Astemizole blocks the rapidly activating delayed rectifier K+ current I(Kr) and the human ether-a go-go-related gene (HERG) K+ channels that underlie it. Astemizole also is rapidly metabolized. The principal metabolite is desmethylastemizole, which retains H1-receptor antagonist properties, has a long elimination time of 9 to 13 days, and its steady-state serum concentration exceeds that of astemizole by more than 30-fold. A second metabolite is norastemizole, which appears in serum in low concentrations following astemizole ingestion and has undergone development as a new antihistamine drug. Our objective in the present work was to study the effects of desmethylastemizole, norastemizole, and astemizole on HERG K+ channels. METHODS AND RESULTS: HERG channels were expressed in a mammalian (HEK 293) cell line and studied using the patch clamp technique. Desmethylastemizole and astemizole blocked HERG current with similar concentration dependence (half-maximal block of 1.0 and 0.9 nM, respectively) and block was use dependent. Norastemizole also blocked HERG current; however, block was incomplete and required higher drug concentrations (half-maximal block of 27.7 nM). CONCLUSIONS: Desmethylastemizole and astemizole cause equipotent block of HERG channels, and these are among the most potent HERG channel antagonists yet studied. Because desmethylastemizole becomes the dominant compound in serum, these findings support the postulate that it becomes the principal cause of long QT syndrome observed in patients following astemizole ingestion. Norastemizole block of HERG channels is weaker; thus, the risk of producing ventricular arrhythmias may be lower. These findings underscore the potential roles of some H1-receptor antagonist metabolites as K+ channel antagonists.


Subject(s)
Astemizole/analogs & derivatives , Astemizole/pharmacology , Benzimidazoles/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Histamine H1 Antagonists/pharmacology , Piperidines/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels , Trans-Activators , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Terfenadine/pharmacology , Transcriptional Regulator ERG
13.
Circ Res ; 84(9): 989-98, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10325236

ABSTRACT

Calcium channel antagonists have diverse effects on cardiac electrophysiology. We studied the effects of verapamil, diltiazem, and nifedipine on HERG K+ channels that encode IKr in native heart cells. In our experiments, verapamil caused high-affinity block of HERG current (IC50=143.0 nmol/L), a value close to those reported for verapamil block of L-type Ca2+ channels, whereas diltiazem weakly blocked HERG current (IC50=17.3 micromol/L), and nifedipine did not block HERG current. Verapamil block of HERG channels was use and frequency dependent, and verapamil unbound from HERG channels at voltages near the normal cardiac cell resting potential or with drug washout. Block of HERG current by verapamil was reduced by lowering pHO, which decreases the proportion of drug in the membrane-permeable neutral form. N-methyl-verapamil, a membrane-impermeable, permanently charged verapamil analogue, blocked HERG channels only when applied intracellularly. Verapamil antagonized dofetilide block of HERG channels, which suggests that they may share a common binding site. The C-type inactivation-deficient mutations, Ser620Thr and Ser631Ala, reduced verapamil block, which is consistent with a role for C-type inactivation in high-affinity drug block, although the Ser620Thr mutation decreased verapamil block 20-fold more than the Ser631Ala mutation. Our findings suggest that verapamil enters the cell membrane in the neutral form to act at a site within the pore accessible from the intracellular side of the cell membrane, possibly involving the serine at position 620. Thus, verapamil shares high-affinity HERG channel blocking properties with other class III antiarrhythmic drugs, and this may contribute to its antiarrhythmic mechanism.


Subject(s)
Anti-Arrhythmia Agents/metabolism , Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Verapamil/metabolism , Verapamil/pharmacology , Cell Line , Diltiazem/pharmacology , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/physiology , Mutation/physiology , Nifedipine/pharmacology , Phenethylamines/antagonists & inhibitors , Phenethylamines/pharmacology , Potassium Channels/genetics , Potassium Channels/physiology , Sulfonamides/antagonists & inhibitors , Sulfonamides/pharmacology , Transcriptional Regulator ERG , Verapamil/analogs & derivatives
14.
Circulation ; 99(17): 2290-4, 1999 May 04.
Article in English | MEDLINE | ID: mdl-10226095

ABSTRACT

BACKGROUND: The congenital long-QT syndrome (LQTS) is an inherited disorder characterized by a prolonged cardiac action potential and a QT interval that leads to arrhythmia. Mutations in the human ether-a-go-go-related gene (HERG), which encodes the rapidly activating component of the delayed rectifier current (IKr), cause chromosome 7-linked LQTS (LQT2). Studies of mutant HERG channels in heterologous systems indicate that the mechanisms mediating LQT2 are varied and include mutant subunits that form channels with altered kinetic properties or nonfunctional mutant subunits. We recently reported a novel missense mutation of HERG (G601S) in an LQTS family that we have characterized in the present work. METHODS AND RESULTS: To elucidate the electrophysiological properties of the G601S mutant channels, we expressed these channels in mammalian cells and Xenopus oocytes. The G601S mutant produced less current than wild-type channels but exhibited no change in kinetic properties or dominant-negative suppression when coexpressed with wild-type subunits. To examine the cellular trafficking of mutant HERG channel subunits, enhanced green fluorescent protein tagging and Western blot analyses were performed. These showed deficient protein trafficking of the G601S mutant to the plasma membrane. CONCLUSIONS: Our results from both the Xenopus oocyte and HEK293 cell expression systems and green fluorescent protein tagging and Western blot analyses support the conclusion that the G601S mutant is a hypomorphic mutation, resulting in a reduced current amplitude. Thus, it represents a novel mechanism underlying LQT2.


Subject(s)
Arrhythmias, Cardiac/genetics , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Trans-Activators , Animals , Biological Transport , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Female , Humans , Mutation , Potassium Channels/metabolism , Recombinant Proteins/biosynthesis , Transcriptional Regulator ERG , Xenopus
15.
J Cardiovasc Electrophysiol ; 10(12): 1597-604, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10636190

ABSTRACT

INTRODUCTION: Torsades de pointes have been observed during treatment with droperidol, a butyrophenone neuroleptic agent. Our objectives were (1) to characterize the effects of droperidol on cardiac repolarization and (2) to evaluate effects of droperidol on a major time-dependent outward potassium current involved in cardiac repolarization (I(K)r). METHODS AND RESULTS: Isolated, buffer-perfused guinea pig hearts (n = 32) were stimulated at different pacing cycle lengths (150 to 250 msec) and exposed to droperidol in concentrations ranging from 10 to 300 nmol/L. Droperidol increased monophasic action potential duration measured at 90% repolarization (MAPD90) in a concentration-dependent manner by 9.8+/-2.3 msec (7.3%+/-0.7%) at 10 nmol/L but by 32.7+/-3.6 msec (25.7%+/-2.2%) at 300 nmol/L (250-msec cycle length). Increase in MAPD90 also was reverse frequency dependent. As noted previously, droperidol 300 nmol/L increased MAPD90 by 32.7+/-3.6 msec (25.7%+/-2.2%) at a pacing cycle length of 250 msec but by only 14.1+/-1.3 msec (13.6%+/-2.3%) at a pacing cycle length of 150 msec. Patch clamp experiments performed in isolated guinea pig ventricular myocytes demonstrated that droperidol decreases the time-dependent outward K+ current elicited by short depolarizations (250 msec; I(K)250) in a concentration-dependent manner. Estimated IC50 for I(K)250, which mostly underlies I(K)r, was 28 nmol/L. Finally, HERG K+ current elicited in HEK293 cells expressing high levels of HERG protein was decreased 50% by droperidol 32.2 nmol/L. CONCLUSION: Potent block of I(K)r by droperidol is likely to underlie QT prolongation observed in patients treated at therapeutic plasma concentrations (10 to 400 nmol/L) of the drug.


Subject(s)
Action Potentials/drug effects , Dopamine Antagonists/pharmacology , Droperidol/pharmacology , Heart Ventricles/physiopathology , Potassium Channel Blockers , Torsades de Pointes/drug therapy , Animals , Disease Models, Animal , Electrocardiography , Guinea Pigs , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Myocardium/metabolism , Myocardium/pathology , Patch-Clamp Techniques , Potassium Channels/metabolism , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology
16.
Am J Physiol ; 275(6): H2016-24, 1998 12.
Article in English | MEDLINE | ID: mdl-9843800

ABSTRACT

Na+ current (INa) through wild-type human heart Na+ channels (hH1) is important for normal cardiac excitability and conduction, and it participates in the control of repolarization and refractoriness. INa kinetics depend strongly on temperature, but INa for hH1 has been studied previously only at room temperature. We characterized early INa (the peak and initial decay) and late INa of the wild-type hH1 channel and a mutant channel (DeltaKPQ) associated with congenital long Q-T syndrome. Channels were stably transfected in HEK-293 cells and studied at 23 and 33 degreesC using whole cell patch clamp. Activation and inactivation kinetics for early INa were twofold faster at higher temperature for both channels and shifted activation and steady-state inactivation in the positive direction, especially for DeltaKPQ. For early INa (<24 ms), DeltaKPQ decayed faster than the wild type for voltages negative to -20 mV but slower for more positive voltages, suggesting a reduced voltage dependence of fast inactivation. Late INa at 240 ms was significantly greater for DeltaKPQ than for the wild type at both temperatures. The majority of late INa for DeltaKPQ was not persistent; rather, it decayed slowly, and this late component exhibited slower recovery from inactivation compared with peak INa. Additional kinetic changes for early and peak INa for DeltaKPQ compared with the wild type at both temperatures were 1) reduced voltage dependence of steady-state inactivation with no difference in midpoint, 2) positive shift for activation kinetics, and 3) more rapid recovery from inactivation. This study represents the first description of human Na+ channel kinetics near physiological temperature and also demonstrates complex gating changes in the DeltaKPQ that are present at 33 degreesC and that may underlie the electrophysiological and clinical phenotype of congenital long Q-T Na+ channel syndromes.


Subject(s)
Long QT Syndrome/metabolism , Mutation/physiology , Myocardium/metabolism , Sodium Channels/genetics , Sodium Channels/physiology , Temperature , Cell Line , Electric Conductivity , Homeostasis/physiology , Humans , Kinetics , Long QT Syndrome/genetics , Reaction Time/physiology , Time Factors
17.
J Biol Chem ; 273(33): 21061-6, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9694858

ABSTRACT

Mutations in HERG are associated with human chromosome 7-linked congenital long QT (LQT-2) syndrome. We used electrophysiological, biochemical, and immunohistochemical methods to study the molecular mechanisms of HERG channel dysfunction caused by LQT-2 mutations. Wild type HERG and LQT-2 mutations were studied by stable and transient expression in HEK 293 cells. We found that some mutations (Y611H and V822M) caused defects in biosynthetic processing of HERG channels with the protein retained in the endoplasmic reticulum. Other mutations (I593R and G628S) were processed similarly to wild type HERG protein, but these mutations did not produce functional channels. In contrast, the T474I mutation expressed HERG current but with altered gating properties. These findings suggest that the loss of HERG channel function in LQT-2 mutations is caused by multiple mechanisms including abnormal channel processing, the generation of nonfunctional channels, and altered channel gating.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Long QT Syndrome/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Biological Transport , Blotting, Western , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Long QT Syndrome/physiopathology , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Processing, Post-Translational , Transcriptional Regulator ERG , Transfection
18.
Mol Pharmacol ; 54(1): 113-21, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658196

ABSTRACT

In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Cetirizine was completely devoid of any inhibitory action on HERG K+ channels heterologously expressed in Xenopus laevis oocytes in concentrations up to 30 microM. On the other hand, terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (the estimated IC50 values were 330 and 480 nM, respectively), whereas loratadine was approximately 300-fold less potent (IC50 approximately 100 microM). In addition, in contrast to terfenadine, cetirizine did not show use-dependent blockade. In SH-SY5Y cells, a human neuroblastoma clone that constitutively expresses K+ currents carried by HERG channels (IHERG), as well as in human embryonic kidney 293 cells stably transfected with HERG cDNA, extracellular perfusion with 3 microM cetirizine did not exert any inhibitory action on IHERG. Astemizole (3 microM), on the other hand, was highly effective. Terfenadine (3 microM) caused a marked (approximately 80%) inhibition of IHERG in SH-SY5Y cells, whereas loratadine, at the same concentration, caused a 40% blockade. Furthermore, the application of cetirizine (3 microM) on the intracellular side of the membrane of HERG-transfected human embryonic kidney 293 cells did not affect IHERG, whereas the same intracellular concentration of astemizole caused a complete block. The results of the current study suggest that second-generation antihistamines display marked differences in their ability to block HERG K+ channels. Cetirizine in particular, which possesses more polar and smaller substituent groups attached to the tertiary amine compared with other antihistamines, lacks HERG-blocking properties, possibly explaining the absence of torsade de pointes ventricular arrhythmias associated with its therapeutical use.


Subject(s)
Cation Transport Proteins , Cetirizine/pharmacology , DNA-Binding Proteins , Heart/drug effects , Histamine H1 Antagonists/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Receptors, Histamine H1/drug effects , Trans-Activators , Animals , Astemizole/pharmacology , ERG1 Potassium Channel , Electrophysiology , Ether-A-Go-Go Potassium Channels , Female , Heart/physiology , Humans , Loratadine/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Potassium Channels/metabolism , Receptors, Histamine H1/metabolism , Terfenadine/pharmacology , Transcriptional Regulator ERG , Transfection , Tumor Cells, Cultured/drug effects , Xenopus laevis
19.
Biophys J ; 74(4): 1830-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9545045

ABSTRACT

Although L-type Ca2+ channels have been shown to play a central role in cardiac excitation-contraction (E-C) coupling, little is known about the role of T-type Ca2+ channels in this process. We used the amphotericin B perforated patch method to study the possible role of T-type Ca2+ current in E-C coupling in isolated canine Purkinje myocytes where both Ca2+ currents are large. T-type Ca2+ current was separated from L-type Ca2+ current using protocols employing the different voltage dependencies of the channel types and their different sensitivities to pharmacological blockade. We showed that Ca2+ admitted through either T- or L-type Ca2+ channels is capable of initiating contraction and that the contractions depended on Ca2+-induced Ca2+ release from the sarcoplasmic reticulum (SR). The contractions, however, had different properties. Those initiated by Ca2+ entry through T-type Ca2+ channels had a longer delay to the onset of shortening, slower rates of shortening and relaxation, lower peak shortening, and longer time to peak shortening. These differences were present even when L-type Ca2+ current amplitude, or charge entry, was less than that of T-type Ca2+ current, suggesting that Ca2+ entry through the T-type Ca2+ channel is a less effective signal transduction mechanism to the SR than is Ca2+ entry through the L-type Ca2+ channel. We conclude that under our experimental conditions in cardiac Purkinje cells Ca2+ entry through the T-type Ca2+ channel can activate cell contraction. However, Ca2+ entry through the L-type Ca2+ channel is a more effective signal transduction mechanism. Our findings support the concept that different structural relationships exist between these channel types and the SR Ca2+ release mechanism.


Subject(s)
Calcium Channels/metabolism , Myocardial Contraction/physiology , Purkinje Cells/metabolism , Animals , Biophysical Phenomena , Biophysics , Cadmium/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/classification , Calcium Channels/drug effects , Dogs , In Vitro Techniques , Membrane Potentials , Myocardial Contraction/drug effects , Nickel/pharmacology , Patch-Clamp Techniques , Purkinje Cells/drug effects , Purkinje Cells/physiology , Sarcoplasmic Reticulum/metabolism , Signal Transduction
20.
Biophys J ; 74(1): 230-41, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9449325

ABSTRACT

We have established stably transfected HEK 293 cell lines expressing high levels of functional human ether-a go-go-related gene (HERG) channels. We used these cells to study biochemical characteristics of HERG protein, and to study electrophysiological and pharmacological properties of HERG channel current at 35 degrees C. HERG-transfected cells expressed an mRNA band at 4.0 kb. Western blot analysis showed two protein bands (155 and 135 kDa) slightly larger than the predicted molecular mass (127 kDa). Treatment with N-glycosidase F converted both bands to smaller molecular mass, suggesting that both are glycosylated, but at different levels. HERG current activated at voltages positive to -50 mV, maximum current was reached with depolarizing steps to -10 mV, and the current amplitude declined at more positive voltages, similar to HERG channel current expressed in other heterologous systems. Current density at 35 degrees C, compared with 23 degrees C, was increased by more than twofold to a maximum of 53.4 +/- 6.5 pA/pF. Activation, inactivation, recovery from inactivation, and deactivation kinetics were rapid at 35 degrees C, and more closely resemble values reported for the rapidly activating delayed rectifier K+ current (I(Kr)) at physiological temperatures. HERG channels were highly selective for K+. When we used an action potential clamp technique, HERG current activation began shortly after the upstroke of the action potential waveform. HERG current increased during repolarization to reach a maximum amplitude during phases 2 and 3 of the cardiac action potential. HERG contributed current throughout the return of the membrane to the resting potential, and deactivation of HERG current could participate in phase 4 depolarization. HERG current was blocked by low concentrations of E-4031 (IC50 7.7 nM), a value close to that reported for I(Kr) in native cardiac myocytes. Our data support the postulate that HERG encodes a major constituent of I(Kr) and suggest that at physiological temperatures HERG contributes current throughout most of the action potential and into the postrepolarization period.


Subject(s)
Action Potentials/physiology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Trans-Activators , Anti-Arrhythmia Agents/pharmacology , Cell Line , Cell Membrane/physiology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Kidney , Kinetics , Membrane Potentials/drug effects , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium Channels/biosynthesis , Potassium Channels/drug effects , Pyridines/pharmacology , Recombinant Proteins/biosynthesis , Temperature , Time Factors , Transcriptional Regulator ERG , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...