Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 54(1): 220-30, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658209

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.


Subject(s)
Benzodiazepines/pharmacology , Heart/drug effects , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Action Potentials/drug effects , Animals , Delayed Rectifier Potassium Channels , Guinea Pigs , Heart/physiology , Heart Ventricles/drug effects , Humans , Ion Channels/drug effects , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Potassium Channels/metabolism , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Ventricular Function , Xenopus laevis/genetics
2.
Br J Pharmacol ; 123(1): 23-30, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9484850

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.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Imidazoles/pharmacology , Imidazolidines , Piperazines/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Trans-Activators , Animals , Cell Line , ERG1 Potassium Channel , Electric Stimulation , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Hydantoins , Membrane Potentials/physiology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels/metabolism , RNA, Messenger/biosynthesis , Xenopus
4.
Circulation ; 94(11): 2938-46, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8941124

ABSTRACT

BACKGROUND: The class III antiarrhythmic agent RP 58866 and its active enantiomer, terikalant, are reported to selectively block the inward rectifier K+ current, IK1. These drugs have demonstrated efficacy in animal models of cardiac arrhythmias, suggesting that block of IK1 may be a useful antiarrhythmic mechanism. The symmetrical action potential (AP)-prolonging and bradycardic effects of these drugs, however, are inconsistent with a sole effect on IK1. METHODS AND RESULTS: We studied the effects of RP 58866 and terikalant on AP and outward K+ currents in guinea pig ventricular myocytes. RP 58866 and terikalant potently blocked the rapidly activating delayed rectifier K+ current, IKr, with IC50S of 22 and 31 nmol/L, respectively. Block of IK1 was approximately 250-fold less potent; IC50S were 8 and 6 mumol/L, respectively. No significant block of the slowly activating delayed rectifier, IK1, was observed at < or = 10 mumol/L. The phenotypical IKr currents in mouse AT-1 cells and Xenopus oocytes expressing HERG were also blocked 50% by 200 to 250 nmol/L RP 58866 or terikalant, providing further conclusive evidence for potent block of IKr. RP 58866 < or = 1 mumol/L and dofetilide increased AP duration symmetrically, consistent with selective block of IKr. Only higher concentrations (> or = 10 mumol/L) of RP 58866 slowed the rate of AP repolarization and decreased resting membrane potential, consistent with an additional but substantially less potent block of IK1. CONCLUSIONS: These data demonstrate that RP 58866 and terikalant are potent blockers of IKr and prompt a reinterpretation of previous studies that assumed specific block of IK1 by these drugs.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , Chromans/pharmacology , DNA-Binding Proteins , Heart/drug effects , Heart/physiology , Piperidines/pharmacology , Potassium Channels, Voltage-Gated , Potassium/antagonists & inhibitors , Trans-Activators , Action Potentials , Animals , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Mice , Myocardium/cytology , Oocytes/metabolism , Osmolar Concentration , Patch-Clamp Techniques , Potassium/physiology , Potassium Channels/physiology , Reaction Time/drug effects , Stereoisomerism , Xenopus
5.
Am J Physiol ; 271(6 Pt 2): H2477-89, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8997308

ABSTRACT

The delayed rectifier K+ current (IK) in rabbit heart has long been thought to consist of only a single, rapidly activating, dofetilide-sensitive current, IKr. However, we find that IK of rabbit ventricular myocytes actually consists of both rapid and slow components, IKr and IKs, respectively, that can be isolated pharmacologically. Thus, after complete blockade of IKr with dofetilide, the remaining current, IKs, is homogeneous as judged by an envelope of tails test. IKs activates and deactivates slowly, continues to activate during sustained depolarizations, has a half-activation potential of 7.0 +/- 0.8 mV and slope factor of 11.0 +/- 0.7 mV, reverses at -77.2 +/- 1.3 mV (extracellular K+ concentration = 4 mM), is increased by removing extracellular K+, and is enhanced by isoproterenol and stocked by azimilide. Northern analysis demonstrates that the minK (IsK) gene, which encodes a subunit of the channel that underlies the IKs current, is expressed in rabbit heart. Expression of the rabbit protein in Xenopus oocytes elicits a slowly activating, voltage-dependent current, IsK, similar to those expressed previously from mouse, rat, guinea pig, and human genes. The results demonstrate that IKs is present in rabbit ventricle and therefore contributes to cardiac repolarization in this species.


Subject(s)
Imidazolidines , Potassium Channels/physiology , Ventricular Function , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Animals , Anti-Arrhythmia Agents/pharmacology , Electric Conductivity , Electrophysiology , Gene Expression , Guinea Pigs , Heart/physiology , Humans , Hydantoins , Imidazoles/pharmacology , Isoproterenol/pharmacology , Mice , Molecular Sequence Data , Mutation , Oocytes/metabolism , Piperazines/pharmacology , Potassium Channels/genetics , Rabbits , Rats , Xenopus laevis
6.
J Cardiovasc Pharmacol ; 26(2): 259-71, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7475051

ABSTRACT

We studied the effects of NE-10064 (azimilide), a new antiarrhythmic agent reported to be a selective blocker of the slowly activating component of the delayed rectifier, IKs. In ferret papillary muscles, NE-10064 increased effective refractory period (ERP) and decreased isometric twitch tension in a concentration-dependent manner (0.3-30 microM). Increases in ERP showed reverse use-dependence, and were greater at 1 than at 3 Hz. In contrast, changes in tension were use dependent, with larger decreases observed at 3 than at 1 Hz. In guinea pig ventricular myocytes, NE-10064 (0.3-3 microM) significantly prolonged action potential duration (APD) at 1 Hz. At 3 Hz, NE-10064 (0.3-1 microM) increased APD only slightly, and at 10 microM decreased APD and the plateau potential. NE-10064 potently blocked the rapidly activating component of the delayed rectifier, IKr (IC50 0.4 microM), and inhibited IKs (IC50 3 microM) with nearly 10-fold less potency. NE-10064 (10 microM) did not block the inward rectifier potassium current (IKl). NE-10064 (10 microM) blocked the L-type calcium current (ICa) in a use-dependent manner; block was greater at 3 than at 1 Hz. We conclude that (a) NE-10064's block of potassium currents is relatively selective for IKr over IKs, (b) NE-10064 inhibits ICa in a use-dependent fashion, and (c) NE-10064's effects on ERP and tension in papillary muscle as well as APD and action potential plateau level in myocytes may be explained by its potassium and calcium channel blocking properties.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Heart/drug effects , Imidazoles/pharmacology , Imidazolidines , Piperazines/pharmacology , Potassium Channel Blockers , Action Potentials/drug effects , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Ferrets , Guinea Pigs , Heart/physiology , Hydantoins , In Vitro Techniques , Male , Refractory Period, Electrophysiological/drug effects
7.
Circ Res ; 76(1): 110-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-8001268

ABSTRACT

We compared the cardiac electrophysiological actions of two types of H1-receptor antagonists--the piperidines, astemizole and terfenadine, and the nonpiperidines, chlorpheniramine and pyrilamine-in vitro in guinea pig ventricular myocytes and in vivo in chloralose-anesthetized dogs. Astemizole and terfenadine significantly increased action potential duration of guinea pig myocytes. This concentration-dependent prolongation of action potential duration was reverse frequency dependent and led to development of early afterdepolarizations, which occurred more frequently at higher concentrations and slower pacing frequencies. Astemizole and terfenadine potently blocked the rapidly activating component of the delayed rectifier, IKr, with IC50 values of 1.5 and 50 nmol/L, respectively. At 10 mumol/L, terfenadine but not astemizole blocked the slowly activating component of the delayed rectifier, IKs (58.4 +/- 3.1%), and the inward rectifier, IK1 (20.5 +/- 3.4%). Chlorpheniramine and pyrilamine blocked IKr relatively weakly (IC50 = 1.6 and 1.1 mumol/L, respectively) and IKs and IK1 less than 20% at 10 mumol/L. Astemizole and terfenadine (1.0 to 3.0 mg/kg IV) significantly prolonged the QTc interval and ventricular effective refractory period in vivo. Chlorpheniramine and pyrilamine (< or = 3.0 mg/kg) did not significantly affect these parameters. Block of repolarizing K+ currents, particularly IK1, by astemizole and terfenadine produces reverse rate-dependent prolongation of action potential duration and development of early afterdepolarizations, delays ventricular repolarization, and may underlie the development of torsade de pointes ventricular arrhythmias observed with the use and abuse of these agents.


Subject(s)
Astemizole/pharmacology , Chlorpheniramine/pharmacology , Heart/drug effects , Pyrilamine/pharmacology , Terfenadine/pharmacology , Action Potentials/drug effects , Animals , Dose-Response Relationship, Drug , Electrocardiography , Guinea Pigs , Heart/physiology , In Vitro Techniques , Potassium Channels/drug effects , Refractory Period, Electrophysiological/drug effects
8.
J Pharmacol Exp Ther ; 269(2): 541-54, 1994 May.
Article in English | MEDLINE | ID: mdl-7802864

ABSTRACT

The cardiac electrophysiologic and antiarrhythmic actions of two Class III ketone- and alcohol-containing spirobenzopyran piperidine analogs, L-702,958 and L-706,000 [MK-499], respectively, were assessed in vitro and in vivo. L-702,958 and L-706,000 [MK-499] selectively blocked the rapidly activating component of the delayed rectifier K+ current in guinea pig isolated ventricular myocytes (IC50 values, 14.6 and 43.9 nM, respectively), and prolonged effective refractory period in ferret isolated papillary muscles (EC25 values, 10.5 and 53.8 nM, respectively). In anesthetized dogs, L-702,958 and L-706,000 [MK-499] increased ventricular refractory periods (ED20 values, 3.3 and 9.2 micrograms/kg i.v., respectively) and concomitantly increased ECG QT interval and left ventricular+dP/dt. Cumulative i.v. administrations of up to 100 micrograms/kg of L-702,958 and 300 micrograms/kg L-706,000 [MK-499] in anesthetized dogs increased atrial and ventricular refractoriness and prolonged the ECG QT interval, but did not alter atrial, atrioventricular nodal, His-Purkinje or ventricular conduction indices. In anesthetized dogs studied chronically (9.2 +/- 1.1 days) after anterior myocardial infarction, the cumulative i.v. administrations of 100 micrograms/kg of L-702,958 and 300 of micrograms/kg L-706,000 [MK-499] suppressed the induction of ventricular tachyarrhythmia by programmed ventricular stimulation (suppression rates: 8 of 10, 80% and 9 of 11, 82%, respectively) and reduced the incidence of lethal ventricular arrhythmias (incidence of lethal ischemic arrhythmias: 4 of 10, 40% and 1 of 11 9%, respectively, compared to 34 of 40, 85%, in vehicle controls. L-702,958 and L-706,000 [MK-499] (cumulative 100 and 300 micrograms/kg i.v., respectively) did not facilitate the induction of arrhythmias by programmed ventricular stimulation in postinfarction dogs. After equivalently effective p.o. doses in conscious dogs, L-702,958 (10 micrograms/kg) and L-706,000 [MK-499] (30 micrograms/kg) increased ECG QT interval with long durations of action of approximately 9 and 14 hr, respectively. L-706,000 [MK-499] elicited a more consistent and sustained prolongation of the QT interval than L-702,958. These findings show that both L-702,958 and L-706,000 [MK-499] are potentially useful agents for the prevention of malignant ventricular arrhythmias in the setting of myocardial ischemic injury.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzopyrans/pharmacology , Heart/drug effects , Piperidines/pharmacology , Animals , Dogs , Electrocardiography/drug effects , Female , Ferrets , Guinea Pigs , Heart/physiology , Hemodynamics/drug effects , In Vitro Techniques , Male , Molecular Structure , Myocardial Ischemia/physiopathology
9.
Proc Natl Acad Sci U S A ; 91(5): 1766-70, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-7510407

ABSTRACT

We have isolated cardiac cDNA and genomic clones encoding the guinea pig IsK protein. The deduced amino acid sequence is approximately 78% identical to the rat, mouse, and human variants of this channel, and the structure of the gene encoding the protein is also similar to that in other species. For example, the gene is present only once in the haploid genome, the protein-coding sequence is present on a single uninterrupted exon, an intron exists in the 5' untranslated domain, and multiple alternative polyadenylation sites are used in processing the transcript. Expression of the guinea pig protein in Xenopus oocytes results in a slowly activating, voltage-dependent K+ current, IsK, similar to those expressed previously from the rat, mouse, and human genes. However, in sharp contrast to the rat and mouse currents, activation of protein kinase C with phorbol esters increases the amplitude of the guinea pig IsK current, analogous to its effects on the endogenous IKs current in guinea pig cardiac myocytes. Mutagenesis of the guinea pig cDNA to alter four cytoplasmic amino acid residues alters the phenotype of the current response to protein kinase C from enhancement to inhibition, mimicking that of rat and mouse IsK currents. This mutation is consistent with reports that phosphorylation of Ser-102 by protein kinase C decreases the current amplitude. These data explain previously reported differences in the regulatory properties between recombinant rat or mouse IsK channels and native guinea pig IKs channels and provide further evidence that the IsK protein forms the channels that underlie the IKs current in the heart.


Subject(s)
Membrane Proteins/metabolism , Myocardium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels , Potassium/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Enzyme Activation , Female , Guinea Pigs , Humans , Ion Channels/metabolism , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Rats , Xenopus
10.
Circ Res ; 72(1): 75-83, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8417848

ABSTRACT

Class III antiarrhythmic agents act by selective prolongation of cardiac action potential duration (APD). Methanesulfonanilide class III agents (e.g., E-4031 and dofetilide) are extremely potent and lengthen action potentials in a "reverse" rate-dependent manner; i.e., effects are greater at low compared with high rates of stimulation. By using the whole-cell current-clamp technique in isolated guinea pig ventricular myocytes, APD was shortened by rapid pacing (244 +/- 16 msec at 30 pulses per minute, 166 +/- 8 msec at 240 pulses per minute; n = 8). Dofetilide (1 microM) prolonged APD more when cells were stimulated at the rate of 30 pulses per minute (44 +/- 10-msec increase) than at 240 pulses per minute (21 +/- 5-msec increase). We investigated the mechanism of APD prolongation using voltage-clamp techniques. Dofetilide selectively inhibited IKr (IC50, 31.5 nM), defined as the rapidly activating inward rectifying component of net delayed rectifier K+ current (IK), without effects on the larger but more slowly activating component of IK (IKs) or on the inward rectifier K+ current (IK1). To examine the rate-dependent effects of dofetilide on APD, trains of conditioning pulses to 0 mV (200-msec duration) were applied at either 30 or 240 pulses per minute to mimic the action potential experiments. Test pulses or ramps were given after the conditioning train to quantitate changes in IK1, IKr, or IKs. The magnitude of neither IK1 nor IKr was dependent on the rate of the preceding train of depolarizations. Sensitivity to block of IKr by dofetilide was rate independent.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Myocardium/metabolism , Phenethylamines/pharmacology , Potassium/metabolism , Sulfonamides/pharmacology , Action Potentials/drug effects , Animals , Dose-Response Relationship, Drug , Guinea Pigs , Male , Myocardium/cytology
11.
Pflugers Arch ; 420(2): 180-6, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1620577

ABSTRACT

We sought to determine whether extracellular Ca2+ (Ca2+e) and K+ (K+e) play essential roles in the normal functioning of cardiac K+ channels. Reports by others have shown that removal of Ca2+e and K+e alters the gating properties of neural delayed rectifier (IK) and A-type K+ currents, resulting in a loss of normal cation selectivity and voltage-dependent gating. We found that removal of Ca2+e and K+e from the solution bathing guinea pig ventricular myocytes often induced a leak conductance, but did not affect the ionic selectivity or time-dependent activation and deactivation properties of IK. The effect of [K+]e on the magnitude of the two components of cardiac IK was also examined. IK in guinea pig myocytes is comprised of two distinct types of currents: IKr (rapidly activating, rectifying) and IKs (slowly activating). The differential effect of Ca2+e on the two components of IK (previously shown to shift the voltage dependence of activation of the two currents in opposite directions) was exploited to determine the role of K+e on the magnitude of IKs and IKr. Lowering [K+]e from 4 to 0 mM increased IKs, as expected from the change in driving force for K+, but decreased IKr. The differential effect of [K+]e on the two components of cardiac IK may explain the reported discrepancies regarding modulation of cardiac IK conductance by this cation.


Subject(s)
Calcium/physiology , Extracellular Space/physiology , Ion Channel Gating , Myocardium/metabolism , Potassium Channels/metabolism , Potassium/physiology , Animals , Cells, Cultured , Guinea Pigs , Membrane Potentials , Myocardium/cytology , Potassium Channels/physiology
12.
Am J Physiol ; 260(2 Pt 2): H393-9, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1899980

ABSTRACT

The delayed rectifier outward K+ current (IK) was studied in isolated guinea pig atrial myocytes using the whole cell voltage-clamp technique. Similar to previous findings in ventricular cells, IK of atrial cells is the composite of two distinct components: IK,r, a rapidly activating current that exhibits strong inward rectification and IK,s, a slowly activating current with only modest rectification. IK,r was defined by its sensitivity to block by Co2+ and the class III antiarrhythmic agent, E-4031. IK,r underlies the prominent outward "hump" (between -30 and +40 mV) in the steady-state current-voltage relationship. Activation of IK,r was not dependent on transient changes in intracellular Ca2+ concentration. Block of Ca2+ current by nisoldipine or nitrendipine did not prevent activation of IK,r. Peak IK,r was not decreased in cells when intracellular Ca2+ was strongly buffered with 1,2-bis(aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. The activation curve for IK,r in atrial cells had a threshold of -40 mV, a half-point of -19 mV, and a slope factor of 5.2 mV. The activation curve for IK,s had a half-point of +24 mV and a slope factor of 15.7 mV. The peak tail currents of fully activated IK,s (21.1 pA/pF) and IK,r (2.53 pA/pF) are about two times that previously measured in guinea pig ventricular cells. This difference in current density may partly explain why action potentials of atrial cells are shorter than those of ventricular cells in guinea pig hearts.


Subject(s)
Heart/physiology , Potassium/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Buffers , Calcium/antagonists & inhibitors , Calcium/physiology , Cobalt/pharmacology , Egtazic Acid/pharmacology , Electric Conductivity , Guinea Pigs , Heart Atria , Kinetics , Myocardium/cytology , Nisoldipine/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology
13.
Circ Res ; 68(1): 77-84, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1984874

ABSTRACT

The mechanism by which isoproterenol (ISO) prevents the prolongation of action potential duration (APD) and refractory period (RP) by the class III antiarrhythmic agent E-4031 was studied. E-4031 (1 microM) increased RP by 50% with no effect on contractile force in papillary muscles isolated from guinea pig heart. ISO (1 microM) increased force of contraction more than fivefold and decreased RP by 25%. The prolongation of RP by E-4031 was prevented by pretreatment of muscles with ISO. The prolongation of APD in isolated guinea pig ventricular myocytes by 5 microM E-4031 also was antagonized by prior exposure of the cells to 1 microM ISO. Instantaneous currents and delayed rectifier K+ currents, IK, were measured in isolated myocytes using the suction microelectrode voltage-clamp technique. Currents were measured in response to 225-msec depolarizing pulses from a holding potential of -40 mV. Previous studies have demonstrated that IK in these cells results from activation of two distinct outward K+ currents, IKs and IKr (specifically blocked by E-4031). ISO doubled the magnitude of IKs without significant effect on IKr. The instantaneous current, putatively identified as a Cl- current, also was doubled by ISO but was unaffected by E-4031. The augmented conductance of IKs and instantaneous current by ISO results in a decrease in RP. The small effect of E-4031 on APD and RP in the presence of ISO results from the smaller contribution of IKr relative to the augmented repolarizing currents.


Subject(s)
Heart/drug effects , Isoproterenol/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Refractory Period, Electrophysiological/drug effects , Action Potentials , Animals , Anti-Arrhythmia Agents/pharmacology , Cell Separation , Electric Conductivity , Guinea Pigs , Heart/physiology , In Vitro Techniques , Myocardium/cytology , Papillary Muscles/drug effects , Piperidines/antagonists & inhibitors , Pyridines/antagonists & inhibitors
14.
Am J Physiol ; 259(6 Pt 2): H1881-9, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2260712

ABSTRACT

Delayed rectifier outward K+ current (IK) in guinea pig ventricular myocytes represents the sum of two currents: a slowly activating delayed rectifier K+ current (IK.s) and a relatively rapidly activating delayed rectifier K+ current (IK.r), which rectifies at positive potentials and is specifically blocked by the class III antiarrhythmic agent, E-4031. La3+ was previously reported to block an unidentified component of IK in these cells. We used the whole cell voltage-clamp technique on isolated myocytes and confirmed these results: we show that the current blocked by La3+ (greater than or equal to 1 microM) is IK.r. This block is not caused by La3+ displacement of surface-bound Ca2+. Thus, in the presence of either E-4031 or La3+, IK represents the activation of a single current, IK.s. La3+ (10 microM-1 mM) also caused a positive shift in the voltage dependence of the activation curve of IK.s. When we assumed that La3+ acts to bind and screen negative surface charges on the outer sarcolemmal membrane, the external surface potential of these cells (in 1.8 mM Ca2+) could be estimated to be -19 mV. A modification of the Gouy-Chapman equation was used to estimate the equilibrium constant for La3+ binding (10.7 mM-1) and the minimum spacing between the negative charges on the surface membrane (22 A).


Subject(s)
Heart/physiology , Lanthanum/pharmacology , Potassium/physiology , Animals , Calcium/pharmacology , Cell Membrane/physiology , Electric Conductivity , Electrophysiology , Models, Cardiovascular , Myocardium/cytology , Myocardium/ultrastructure
15.
J Gen Physiol ; 96(1): 195-215, 1990 Jul.
Article in English | MEDLINE | ID: mdl-2170562

ABSTRACT

An envelope of tails test was used to show that the delayed rectifier K+ current (IK) of guinea pig ventricular myocytes results from the activation of two outward K+ currents. One current was specifically blocked by the benzenesulfonamide antiarrhythmic agent, E-4031 (IC50 = 397 nM). The drug-sensitive current, "IKr" exhibits prominent rectification and activates very rapidly relative to the slowly activating drug-insensitive current, "IKs." IKs was characterized by a delayed onset of activation that occurs over a voltage range typical of the classically described cardiac IK. Fully activated IKs, measured as tail current after 7.5-s test pulses, was 11.4 times larger than the fully activated IKr. IKr was also blocked by d-sotalol (100 microM), a less potent benzenesulfonamide Class III antiarrhythmic agent. The activation curve of IKr had a steep slope (+7.5 mV) and a negative half-point (-21.5 mV) relative to the activation curve of IKs (slope = +12.7 mV, half-point = +15.7 mV). The reversal potential (Erev) of IKr (-93 mV) was similar to EK (-94 mV for [K+]o = 4 mM), whereas Erev of IKs was -77 mV. The time constants for activation and deactivation of IKr made up a bell-shaped function of membrane potential, peaking between -30 and -40 mV (170 ms). The slope conductance of the linear portion of the fully activated IKr-V relation was 22.5 S/F. Inward rectification of this relation occurred at potentials greater than -50 mV, resulting in a voltage-dependent decrease in peak IKr at test potentials greater than 0 mV. Peak IKr at 0 mV averaged 0.8 pA/pF (n = 21). Although the magnitude of IKr was small relative to fully activated IKs, the two currents were of similar magnitude when measured during a relatively short pulse protocol (225 ms) at membrane potentials (-20 to +20 mV) typical of the plateau phase of cardiac action potentials.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/physiology , Myocardium/metabolism , Potassium Channels/drug effects , Animals , Calcium Channels/drug effects , Guinea Pigs , In Vitro Techniques , Kinetics , Membrane Potentials/drug effects , Microelectrodes , Myocardium/cytology , Nisoldipine/pharmacology , Ouabain/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Sodium Channels/metabolism
16.
J Cardiovasc Pharmacol ; 9(4): 435-44, 1987 Apr.
Article in English | MEDLINE | ID: mdl-2438506

ABSTRACT

Recainam, [N-2,6-dimethylphenyl-N'-3-(1-methylethyl-amino)propylurea] hydrochloride (Wy-42,362), is a new class I antiarrhythmic agent that has been shown to be very effective in suppressing premature ventricular contractions in humans. To clarify the mechanism of antiarrhythmic action, the electrophysiologic effects of recainam were examined in canine cardiac Purkinje fibers using standard microelectrode techniques. Recainam at 3-100 microM (1-30 micrograms/ml) produced concentration-dependent decreases in action potential duration (APD), membrane responsiveness, and maximal upstroke velocity (Vmax). The reduction in Vmax was strongly modulated by the frequency of stimulation--i.e., Vmax block was use dependent. The rate of development of use-dependent block produced by recainam was much slower than typically seen with lidocaine, but comparable with that of the class Ia agents disopyramide and procainamide. However, unlike agents of the Ia subclass, recainam did not prolong APD at any concentration or cycle length tested. In summary, recainam appears to possess a novel cardiac cellular electrophysiologic profile, in that it shares characteristics with all three current class I antiarrhythmic subclasses.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart Conduction System/drug effects , Phenylurea Compounds/pharmacology , Purkinje Fibers/drug effects , Action Potentials/drug effects , Animals , Cardiac Pacing, Artificial , Dogs , In Vitro Techniques , Membrane Potentials/drug effects , Purkinje Fibers/physiology
17.
Eur J Pharmacol ; 126(1-2): 37-45, 1986 Jul 15.
Article in English | MEDLINE | ID: mdl-3758165

ABSTRACT

The antiarrhythmic activity of iprindole was compared to that of imipramine in a variety of experimental arrhythmia models. Iprindole at 20 mg/kg i.v. showed efficacy in reverting ouabain- and aconitine-induced arrhythmias in pentobarbital anesthetized dogs, and at 15-30 mg/kg i.v. reduced the severity of the ventricular arrhythmias following acute coronary artery occlusion in anesthetized pigs. Imipramine (5-10 mg/kg i.v.) was also effective in reverting ouabain- and aconitine-induced arrhythmias, but appeared to exacerbate arrhythmias during coronary occlusion. In microelectrode experiments on isolated dog Purkinje fibers, iprindole reduced maximal upstroke velocity (Vmax) and action potential duration (characteristics of Class Ib antiarrhythmic agents) at concentrations greater than 1 microgram/ml. Significant decreases in Vmax occurred at lower iprindole concentrations when membrane potential was reduced by increasing external potassium from 4 to 10 mM, suggesting that electrical activity in depolarized cells may be selectively suppressed by iprindole. The present data indicate that iprindole may exert beneficial therapeutic effects in the treatment of cardiac arrhythmias, mediated, at least in part, through a Class I mechanism of action.


Subject(s)
Anti-Arrhythmia Agents , Indoles/pharmacology , Iprindole/pharmacology , Aconitine , Action Potentials/drug effects , Anesthesia , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Dogs , Female , Kinetics , Male , Ouabain , Purkinje Fibers/drug effects , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...