Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Gen Physiol ; 113(4): 581-600, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10102938

RESUMO

The biophysical characteristics and alpha subunits underlying calcium-independent transient outward potassium current (Ito) phenotypes expressed in ferret left ventricular epicardial (LV epi) and endocardial (LV endo) myocytes were analyzed using patch clamp, fluorescent in situ hybridization (FISH), and immunofluorescent (IF) techniques. Two distinct Ito phenotypes were measured (21-22 degrees C) in the majority of LV epi and LV endo myocytes studied. The two Ito phenotypes displayed marked differences in peak current densities, activation thresholds, inactivation characteristics, and recovery kinetics. Ito,epi recovered rapidly [taurec, -70 mV = 51 +/- 3 ms] with minimal cumulative inactivation, while Ito,endo recovered slowly [taurec, -70 mV = 3,002 +/- 447 ms] with marked cumulative inactivation. Heteropoda toxin 2 (150 nM) blocked Ito,epi in a voltage-dependent manner, but had no effect on Ito,endo. Parallel FISH and IF measurements conducted on isolated LV epi and LV endo myocytes demonstrated that Kv1.4, Kv4.2, and Kv4.3 alpha subunit expression in LV myocyte types was quite heterogenous: (a) Kv4.2 and Kv4.3 were more predominantly expressed in LV epi than LV endo myocytes, and (b) Kv1.4 was expressed in the majority of LV endo myocytes but was essentially absent in LV epi myocytes. In combination with previous measurements on recovery kinetics (Kv1.4, slow; Kv4.2/4.3, relatively rapid) and Heteropoda toxin block (Kv1.4, insensitive; Kv4.2, sensitive), our results strongly support the hypothesis that, in ferret heart, Kv4.2/Kv4.3 and Kv1.4 alpha subunits, respectively, are the molecular substrates underlying the Ito,epi and Ito,endo phenotypes. FISH and IF measurements were also conducted on ferret ventricular tissue sections. The three Ito alpha subunits again showed distinct patterns of distribution: (a) Kv1.4 was localized primarily to the apical portion of the LV septum, LV endocardium, and approximate inner 75% of the LV free wall; (b) Kv4. 2 was localized primarily to the right ventricular free wall, epicardial layers of the LV, and base of the heart; and (c) Kv4.3 was localized primarily to epicardial layers of the LV apex and diffusely distributed in the LV free wall and septum. Therefore, in intact ventricular tissue, a heterogeneous distribution of candidate Ito alpha subunits not only exists from LV epicardium to endocardium but also from apex to base.


Assuntos
Furões/metabolismo , Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Eletrofisiologia , Técnica Direta de Fluorescência para Anticorpo , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Técnicas In Vitro , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.4 , Masculino , Potenciais da Membrana/fisiologia , Dados de Sequência Molecular , Miocárdio/citologia , Técnicas de Patch-Clamp , Fenótipo , Canais de Potássio/genética , Canais de Potássio Shal
2.
Can J Cardiol ; 14(2): 275-80, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9520865

RESUMO

The nature of open state block was characterized in isolated canine cardiac sarcoplasmic reticulum (SR) potassium channel incorporated into planar lipid bilayers. 4-Aminopyridine (4-AP) blocked the open conductance state of the potassium channels in a voltage-dependent manner. Blockade was reversible, occurred from either the cis (cytoplasmic) or the trans (lumenal) side and was competitive with potassium ions. Reversal potential measurements indicated that this channel was impermeable to 4-AP. Measured effective electrical distances were roughly symmetrical and indicated penetration of 0.39 and 0.42 of the membrane electrical field from the cis and trans sides, respectively. Effective electrical distance was insensitive to potassium ion concentration in the range 50 to 200 mM and indicated that 4-AP was able to penetrate relatively deeply into the pore compared with blockade of sarcolemmal potassium channels. Potassium ion concentration and voltage dependence of 4-AP blockade were consistent with a two binding site blockade model, similar to the model used previously to describe calcium ion blockade of the SR potassium ion channel. Unlike calcium blockade, however, 4-AP blocked from either cis or trans in a similar manner, suggesting a distinct binding site for each of these two blockers. Open channel, voltage-dependent blockade of the SR potassium channel by 4-AP is in marked contrast to its action on sarcolemmal potassium channels and suggests that either 4-AP penetrates much farther into the potassium channel permeation pathway than was previously believed, or the SR potassium channel has a very different physical pore arrangement from that of sarcolemmal potassium channels.


Assuntos
4-Aminopiridina/farmacologia , Coração/efeitos dos fármacos , Canais de Potássio/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Animais , Cães , Humanos , Miocárdio/metabolismo
3.
Circ Res ; 82(7): 739-50, 1998 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-9562433

RESUMO

Inactivation is the process by which an open channel enters a stable nonconducting conformation after a depolarizing change in membrane potential. Inactivation is a widespread property of many different types of voltage-gated ion channels. Recent advances in the molecular biology of K+ channels have elucidated two mechanistically distinct types of inactivation, N-type and C-type. N-type inactivation involves occlusion of the intracellular mouth of the pore through binding of a short segment of residues at the extreme N-terminal. In contrast to this "tethered ball" mechanism of N-type inactivation, C-type inactivation involves movement of conserved core domain residues that result in closure of the external mouth of the pore. Although C-type inactivation can show rapid kinetics that approach those observed for N-type inactivation, it is often thought of as a slowly developing and slowly recovering process. Current models of C-type inactivation also suggest that this process involves a relatively localized change in conformation of residues near the external mouth of the permeation pathway. The rate of C-type inactivation and recovery can be strongly influenced by other factors, such as N-type inactivation, drug binding, and changes in [K+]o. These interactions make C-type inactivation an important biophysical process in determining such physiologically important properties as refractoriness and drug binding. C-type inactivation is currently viewed as arising from small-scale rearrangements at the external mouth of the pore. This review will examine the multiplicity of interactions of C-type inactivation with N-terminal-mediated inactivation and drug binding that suggest that our current view of C-type inactivation is incomplete. This review will suggest that C-type inactivation must involve larger-scale movements of transmembrane-spanning domains and that such movements contribute to the diversity of kinetic properties observed for C-type inactivation.


Assuntos
Coração/fisiologia , Ativação do Canal Iônico , Canais de Potássio/fisiologia , Regulação Alostérica , Animais , Humanos , Potenciais da Membrana/fisiologia , Fragmentos de Peptídeos/fisiologia , Ligação Proteica
4.
FEBS Lett ; 417(1): 43-7, 1997 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-9395071

RESUMO

Rectification of HERG is due to a rapid inactivation process that has been labeled C-type inactivation and is believed to be due to closure of the external mouth of the pore. We examined the effects of mutation of extracellular residues that remove C-type inactivation on binding of the intracellularly acting methanesulfonanilide drug E-4031. Removal of inactivation through mutation reduced drug affinity by more than an order of magnitude. Elevation of [K+]o in the wild-type channel reduces channel affinity for E-4031. Elevation of [K+]o also interferes with the extracellular pore mouth closure associated with C-type inactivation through a 'foot in the door' mechanism. We examined the possibility that [K+]o elevation reduces drug binding through inhibition of C-type inactivation by comparing drug block in the wild-type and inactivation-removed mutant channels. Elevation of [K+]o decreased affinity in both channel constructs by a roughly equal amount. These results suggest that [K+]o alters drug binding affinity independently of its effects on C-type inactivation. They further suggest that inhibition of pore mouth closure by elevated [K+]o does not have same effect on drug affinity as mutations removing C-type inactivation.


Assuntos
Proteínas de Transporte de Cátions , Piperidinas/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Potássio/metabolismo , Piridinas/metabolismo , Animais , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio/fisiologia , Xenopus laevis
5.
Circ Res ; 81(1): 128-35, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9201036

RESUMO

Repolarization of the cardiac action potential varies widely throughout the heart. This could be due to the differential distribution of ion channels responsible for repolarization, especially the K+ channels. We have therefore studied the cardiac localization of ERG, a channel protein known to play an important role in generation of the rapid component of the delayed rectifier K+ current (IKr), an important determinant of the repolarization waveform, Cryosections of the ferret atrium and ventricle were prepared to determine the localization of ERG by fluorescence in situ hybridization (FISH) and immunofluorescence. We found that in the ferret, ERG transcript and protein expression was most abundant in the epicardial cell layers throughout most of the ventricle, except at the base. In the atrium, we found that ERG is most abundant in the medial right atrium, especially in the trabeculae and the crista terminalis of the right atrial appendage. It also is present in areas within the sinoatrial node. In all regions studied, FISH and immunofluorescence showed concordant localization patterns. These data suggest that repolarization mediated by IKr is not uniform throughout the ferret heart and provide a molecular explanation for heterogeneity in action potential repolarization throughout the mammalian heart.


Assuntos
Furões , Genes , Coração/fisiologia , Miocárdio/metabolismo , Canais de Potássio/análise , Canais de Potássio/genética , Proteínas/análise , Potenciais de Ação , Animais , Anticorpos/análise , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Genes/imunologia , Genes/fisiologia , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Hibridização in Situ Fluorescente , Técnicas In Vitro , Miocárdio/citologia , Canais de Potássio/fisiologia
6.
J Physiol ; 502 ( Pt 1): 45-60, 1997 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9234196

RESUMO

1. The human ether à-go-go-related gene (HERG) encodes a K+ channel that is believed to be the basis of the delayed rectified current, IKr, in cardiac muscle. We studied HERG expressed in Xenopus oocytes using a two-electrode and cut-open oocyte clamp technique with [K+]0 of 2 and 98 mM. 2. The time course of activation of the channel was measured using an envelope of tails protocol and demonstrated that activation of the heterologously expressed HERG current (IHERG) was sigmoidal in onset. At least three closed states were required to reproduce the sigmoid time course. 3. The voltage dependence of the activation process and its saturation at positive voltages suggested the existence of at least one relatively voltage-insensitive step. A three closed state activation model with a single voltage-insensitive intermediate closed state was able to reproduce the time and voltage dependence of activation, deactivation and steady-state activation. Activation was insensitive to changes in [K+]0. 4. Both inactivation and recovery time constants increased with a change of [K+]0 from 2 to 98 mM. Steady-state inactivation shifted by approximately 30 mV in the depolarized direction with a change from 2 to 98 mM K+0. 5. Simulations showed that modulation of inactivation is a minimal component of the increase of this current by [K+]0, and that a large increase in total conductance must also occur.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Ativação do Canal Iônico/fisiologia , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Transativadores , Animais , Cálcio/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Feminino , Expressão Gênica/fisiologia , Humanos , Cinética , Potenciais da Membrana/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Canais de Potássio/genética , Homologia de Sequência de Aminoácidos , Regulador Transcricional ERG , Transfecção , Xenopus laevis
7.
Pacing Clin Electrophysiol ; 20(2 Pt 2): 388-96, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9058843

RESUMO

The molecular basis of the potassium ion (K+) channels that generate repolarization in heart tissue remains uncertain, in part because of the molecular diversity of the voltage-gated K+ channel family. In our investigation, we used fluorescent labeled oligonucleotide probes to perform in situ hybridization studies on enzymatically isolated myocytes to determine the identity, regional distribution, and cellular distribution of voltage-gated K+ channel, alpha-subunit mRNA expressed in ferret heart. The regions studied were from the sinoatrial node (SA), right and left atrium, right and left ventricle, and interatrial and interventricular septa. Kv1.5 and Kv1.4 were the most widely distributed K+ channel transcripts in the ferret heart (present in approximately 70%-86% and approximately 46%-95% of tested myocytes, respectively), followed by Kv1.2, Kv2.1, and Kv4.2. In addition, many myocytes contain transcripts for Kv1.3, Kv2.2, Kv4.1, Kv5.1, and members of the Kv3 family. Kv1.1, Kv1.6, and Kv6.1 were rarely expressed in working myocytes, but were more commonly expressed in SA nodal cells. Two other transcripts whose genes have been implicated in the long QT syndrome, erg and KvLQT1, were common in all regions (approximately 41%-58% and 52%-72%, respectively). These results show that both the diversity and heterogeneity of K+ channel mRNA in heart tissue is greater than previously suspected.


Assuntos
Furões , Miocárdio/citologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Canais de Potássio/fisiologia , Transcrição Gênica , Animais , Nó Atrioventricular/metabolismo , Nó Atrioventricular/fisiologia , Canais de Potássio de Retificação Tardia , Sistema de Condução Cardíaco/fisiologia , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Canal de Potássio Kv1.1 , Canal de Potássio Kv1.2 , Canal de Potássio Kv1.3 , Canal de Potássio Kv1.4 , Canal de Potássio Kv1.5 , Masculino , Miocárdio/metabolismo , Canais de Potássio/metabolismo , RNA Mensageiro/química , Canais de Potássio Shab , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiologia , Troponina I/química , Troponina I/genética
8.
Adv Exp Med Biol ; 430: 29-37, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9330716

RESUMO

A recently discovered class of ancillary subunits has been shown to modify the inactivation properties of alpha-subunits belonging to the Kv1 family of potassium channels. One of these subunits, Kv beta 1.2, modifies intrinsic alpha-subunit C-type inactivation. N-type inactivation and open channel block have been proposed to increase the rate of development of C-type inactivation. We demonstrate here that Kv beta 1.2 has kinetic properties which are consistent with rapid open channel block.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Animais , Simulação por Computador , Condutividade Elétrica , Furões , Ativação do Canal Iônico/fisiologia , Cinética , Canal de Potássio Kv1.2 , Canal de Potássio Kv1.4 , Substâncias Macromoleculares , Canais de Potássio/química
9.
Proc Natl Acad Sci U S A ; 93(26): 15119-23, 1996 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-9005448

RESUMO

Voltage-gated K+ channels are complexes of membrane-bound, ion-conducting alpha and cytoplasmic ancillary (beta) subunits. The primary physiologic effect of coexpression of alpha and beta subunits is to increase the intrinsic rate of inactivation of the alpha subunit. For one beta subunit, Kv beta 1.1, inactivation is enhanced through an N-type mechanism. A second beta subunit, Kv beta 1.2, has been shown to increase inactivation, but through a distinct mechanism. Here we show that the degree of enhancement of Kv beta 1.2 inactivation is dependent on the amino acid composition in the pore mouth of the alpha subunit and the concentration of extracellular K+. Experimental conditions that promote C-type inactivation also enhance the stimulation of inactivation by Kv beta 1.2, showing that this beta subunit directly stimulates C-type inactivation. Chimeric constructs containing just the nonconserved N-terminal region of Kv beta 1.2 fused with an alpha subunit behave in a similar fashion to coexpressed Kv beta 1.2 and alpha subunit. This shows that it is the N-terminal domain of Kv beta 1.2 that mediates the increase in C-type inactivation from the cytoplasmic side of the pore. We propose a model whereby the N terminus of Kv beta 1.2 acts as a weakly binding "ball" domain that associates with the intracellular vestibule of the alpha subunit to effect a conformational change leading to enhancement of C-type inactivation.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Estrutura Secundária de Proteína , Sequência de Aminoácidos , Animais , Sequência de Bases , Membrana Celular/fisiologia , Citoplasma/fisiologia , Feminino , Furões , Canal de Potássio Kv1.1 , Canal de Potássio Kv1.2 , Substâncias Macromoleculares , Modelos Estruturais , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos , Oócitos/fisiologia , Mutação Puntual , Reação em Cadeia da Polimerase , Canais de Potássio/biossíntese , Canais de Potássio/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Xenopus
10.
J Gen Physiol ; 108(4): 277-93, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8894977

RESUMO

The effects of NO-related activity and cellular thiol redox state on basal L-type calcium current, ICa,L, in ferret right ventricular myocytes were studied using the patch clamp technique. SIN-1, which generates both NO. and O2-, either inhibited or stimulated ICa,L. In the presence of superoxide dismutase only inhibition was seen. 8-Br-cGMP also inhibited ICa,L, suggesting that the NO inhibition is cGMP-dependent. On the other hand, S-nitrosothiols (RSNOs), which donate NO+, stimulated ICa,L. RSNO effects were not dependent upon cell permeability, modulation of SR Ca2+ release, activation of kinases, inhibition of phosphatases, or alterations in cGMP levels. Similar activation of ICa,L by thiol oxidants, and reversal by thiol reductants, identifies an allosteric thiol-containing "redox switch" on the L-type calcium channel subunit complex by which NO/O2- and NO+ transfer can exert effects opposite to those produced by NO. In sum, our results suggest that: (a) both indirect (cGMP-dependent) and direct (S-nitrosylation/oxidation) regulation of ventricular ICa,L, and (b) sarcolemma thiol redox state may be an important determinant of ICa,L activity.


Assuntos
Canais de Cálcio/metabolismo , Miocárdio/metabolismo , Animais , Canais de Cálcio/efeitos dos fármacos , Espaço Extracelular/metabolismo , Furões , Ventrículos do Coração , Masculino , Molsidomina/análogos & derivados , Molsidomina/farmacologia , Miocárdio/citologia , Óxido Nítrico/fisiologia , Compostos Nitrosos/metabolismo , Oxirredução , Técnicas de Patch-Clamp , Compostos de Sulfidrila/fisiologia
11.
FEBS Lett ; 389(2): 167-73, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8766823

RESUMO

The rapid delayed rectifier, IKr, is believed to have h-erg (human ether-à-go-go related gene) as its molecular basis. A recent study has shown that rectification of h-erg involves a rapid inactivation process that involves rapid closure of the external mouth of the pore or C-type inactivation. We measured the instantaneous current to voltage relationship for h-erg channels using the saponin permeabilized variation of the cut-open oocyte clamp technique. In contrast to C-type inactivation in other voltage-gated K+ channels, the rate of inactivation was strongly voltage dependent at depolarized potentials. This voltage dependence could be modulated independently of activation by increasing [K+]0 from 2 to 98 mM. These results suggest that inactivation of h-erg has its own intrinsic voltage sensor.


Assuntos
Proteínas de Transporte de Cátions , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/biossíntese , Canais de Potássio/química , Animais , Relação Dose-Resposta a Droga , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go , Feminino , Regulação da Expressão Gênica , Oócitos/química , Potássio/química , Potássio/metabolismo , Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Fatores de Tempo , Xenopus laevis/fisiologia
13.
Biophys J ; 70(6): 2704-15, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8744308

RESUMO

Ferret atrial myocytes can display an E-4031-sensitive current (IKr) that is similar to that previously described for guinea pig cardiac myocytes. We examined the ferret atrial IKr as the E-4031-sensitive component of current using the amphotericin B perforated patch-clamp technique. Steady-state IKr during depolarizing pulses showed characteristic inward rectification. Activation time constants during a single pulse were voltage dependent, consistent with previous studies. However, for potentials positive to +30 mV, IKr time course became complex and included a brief transient component. We examined the envelope of tails of the drug-sensitive current for activation in the range -10 to +50 mV and found that the tail currents for IKr do not activate with the same time course as the current during the depolarizing pulse. The activation time course determined from tail currents was relatively voltage insensitive over the range +30 to +50 mV (n = 5), but was voltage sensitive for potentials between -10 and +30 mV and appeared to show some sigmoidicity in this range. These data indicate that activation of IKr occurs in at least two steps, one voltage sensitive and one voltage insensitive, the latter of which becomes rate limiting at positive potentials. We also examined the rapid time-dependent inactivation process that mediates rectification at positive potentials. The time constants for this process were only weakly voltage dependent over the range of potentials from -50 to +60 mV. From these data we constructed a simple linear four-state model that reproduces the general features of ferret IKr, including the initial transient at positive potentials and the apparent discrepancy between the currents during the initial depolarizing pulse and the tail current.


Assuntos
Miocárdio/metabolismo , Canais de Potássio/metabolismo , Animais , Fenômenos Biofísicos , Biofísica , Furões , Cobaias , Átrios do Coração/metabolismo , Técnicas In Vitro , Ativação do Canal Iônico , Cinética , Potenciais da Membrana , Modelos Cardiovasculares , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio
14.
Circ Res ; 78(6): 1083-9, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8635239

RESUMO

The molecular basis of K+ currents that generate repolarization in the heart is uncertain. In part, this reflects the similar functional properties different K+ channel clones display when heterologously expressed, in addition to the molecular diversity of the voltage-gated K+ channel family. To determine the identity, regional distribution, and cellular distribution of voltage-sensitive K+ channel mRNA subunits expressed in ferret heart, we used fluorescent labeled oligonucleotide probes to perform in situ hybridization studies on enzymatically isolated myocytes from the sinoatrial (SA) node, right and left atria, right and left ventricles, and interatrial and interventricular septa. The most widely distributed K+ channel transcripts in the ferret heart were Kv1.5 (present in 69.3% to 85.6% of myocytes tested, depending on the anatomic region from which myocytes were isolated) and Kv1.4 (46.1% to 93.7%), followed by kv1.2, Kv2.1, and Kv4.2. Surprisingly, many myocytes contain transcripts for Kv1.3, Kv2.2, Kv4.1, Kv5.1, and members of the Kv3 family. Kv1.1, Kv1.6, and Kv6.1, which were rarely expressed in working myocytes, were more commonly expressed in SA nodal cells. IRK was expressed in ventricular (84.3% to 92.8%) and atrial (52.4% to 64.0%) cells but was nearly absent (6.6%) in SA nodal cells; minK was most frequently expressed in SA nodal cells (33.7%) as opposed to working myocytes (10.3% to 29.3%). Two gene products implicated in long-QT syndrome, ERG and KvLQT1, were common in all anatomic regions (41.1% to 58.2% and 52.1% to 71.8%, respectively). These results show that the diversity of K+ channel mRNA in heart is greater than previously suspected and that the molecular basis of K+ channels may vary from cell to cell within distinct regions of the heart and also between major anatomic regions.


Assuntos
Hibridização In Situ , Miocárdio/metabolismo , Canais de Potássio/genética , RNA Mensageiro/análise , Animais , Sequência de Bases , Furões , Técnicas In Vitro , Masculino , Dados de Sequência Molecular
15.
J Physiol ; 489 ( Pt 3): 709-21, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8788936

RESUMO

1. A fast inactivating transient K+ current (FK1) cloned from ferret ventricle and expressed in Xenopus oocytes was studied using the two-electrode voltage clamp technique. Removal of the NH2-terminal domain of FK1 (FK1 delta 2-146) removed fast inactivation consistent with previous findings in Kv1.4 channels. The NH2-terminal deletion mutation revealed a slow inactivation process, which matches the criteria for C-type inactivation described for Shaker B channels. 2. Inactivation of FK1 delta 2-146 at depolarized potentials was well described by a single exponential process with a voltage-insensitive time constant. In the range -90 to +20 mV, steady-state C-type inactivation was well described by a Boltzmann relationship that compares closely with inactivation measured in the presence of the NH2-terminus. These results suggest that C-type inactivation is coupled to activation. 3. The coupling of C-type inactivation to activation was assessed by mutation of the fourth positively charged residue (arginine 454) in the S4 voltage sensor to glutamine (R454Q). This mutation produced a hyperpolarizing shift in the inactivation relationship of both FK1 and FK1 delta 2-146 without altering the rate of inactivation of either clone. 4. The rates of recovery from inactivation are nearly identical in FK1 and FK1 delta 2-146. 5. To assess the mechanisms underlying recovery from inactivation the effects of elevated [K+]o and selective mutations in the extracellular pore and the S4 voltage sensor were compared in FK1 and FK1 delta 2-146. The similarity in recovery rates in response to these perturbations suggests that recovery from C-type inactivation governs the overall rate of recovery of inactivated channels for both FK1 and FK1 delta 2-146. 6. Analysis of the rate of recovery of FK1 channels for inactivating pulses of different durations (70-2000 ms) indicates that recovery rate is insensitive to the duration of the inactivating pulse.


Assuntos
Miocárdio/metabolismo , Oócitos/metabolismo , Canais de Potássio/metabolismo , Animais , Sequência de Bases , Clonagem Molecular , Feminino , Furões , Coração/efeitos dos fármacos , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Dados de Sequência Molecular , Mutação , Oócitos/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , RNA Complementar/metabolismo , Xenopus
16.
Circ Res ; 77(2): 284-93, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7614715

RESUMO

Shear stress causes the vascular endothelium to release nitric oxide (NO), which is an important regulator of vascular tone. However, direct measurement of NO release after the imposition of laminar flow has not been previously accomplished because of chemical (oxidative degradation) and physical (diffusion, convection, and washout) complications. Consequently, the mechanism, time course, kinetics, and Ca2+ dependence of NO release due to shear stress remain incompletely understood. In this study, we characterized these parameters by using fura 2 fluorescence and a polymeric porphyrin/Nafion-coated carbon fiber microsensor (detection limit, 5 nmol/L; response time, 1 millisecond) to directly measure changes in [Ca2+]i and NO release due to shear stress or agonist (ATP or brominated Ca2+ ionophore [Br-A23187]) from bovine aortic endothelial cells. The cells were grown to confluence on glass coverslips, loaded with fura 2-AM, and mounted in a parallel-plate flow chamber (volume, 25 microL). The microsensor was positioned approximately 100 microns above the cells with its long axis parallel to the direction of flow. Laminar flow of perfusate was maintained from 0.04 to 1.90 mL/min, which produced shear stresses of 0.2 to 10 dyne/cm2. Shear stress caused transient NO release 3 to 5 seconds after the initiation of flow and 1 to 3 seconds after the rise in [Ca2+]i, which reached a plateau after 35 to 70 seconds. Although the amount (peak rate) of NO release increased as a function of the shear stress (0.08 to 3.80 pmol/s), because of the concomitant increase in the flow rate, the peak NO concentration (133 +/- 9 nmol/L) remained constant. Maintenance of flow resulted in additional transient NO release, with peak-to-peak intervals of 15.5 +/- 2.5 minutes. During this 13- to 18-minute period, when the cells were unresponsive to shear stress, exogenous ATP (10 mumol/L) or Br-A23187 (10 mumol/L) evoked NO release. Prior incubation of the cells with exogenous NO or the removal and EGTA (100 mumol/L) chelation of extracellular Ca2+ blocked shear stress but not ATP-dependent NO release. The kinetics of shear stress-induced NO release (2.23 +/- 0.07 nmol/L per second) closely resembled the kinetics of Ca2+ flux but differed markedly from the kinetics of ATP-induced NO release (5.64 +/- 0.32 nmol/L per second). These data argue that shear stress causes a Ca(2+)-mediated ATP-independent transient release of NO, where the peak rate of release but not the peak concentration depends on the level of shear stress.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Trifosfato de Adenosina/metabolismo , Técnicas Biossensoriais , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Óxido Nítrico/metabolismo , Porfirinas , Animais , Aorta , Cálcio/metabolismo , Bovinos , Células Cultivadas , Modelos Biológicos , Óxido Nítrico/análise , Estresse Mecânico , Fatores de Tempo
17.
Am J Physiol ; 269(1 Pt 2): H385-91, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7631872

RESUMO

In mammals, voltage-gated K+ channels can be made of complexes containing alpha-subunits similar to the Shaker K+ channel and smaller cytoplasmic beta-subunits. Recent studies have suggested that these ancillary beta-subunits can modulate K+ channel gating properties. We studied the effects of a K+ channel beta-subunit, Kv beta 3, coexpressed with a Kv1.4 alpha-subunit, FK1, on the time and voltage dependence of channel activation, inactivation, recovery from inactivation, and deactivation, using an oocyte expression system. Kv beta 3 was found to accelerate both the fast and the slow component of Kv1.4 inactivation. Kv beta 3 also altered the relative contributions of the two components of inactivation by increasing the contribution of the slow component to the inactivation process. Kv beta 3 slowed recovery from inactivation for Kv1.4, but not for a Kv1.4 deletion mutant lacking N-type inactivation. Finally, steady-state activation and the time course of Kv1.4 current activation were not strongly influenced by Kv beta 3; however, deactivation was slowed in the presence of Kv beta 3. This study suggests that Kv beta 3 alters channel states which follow activation.


Assuntos
Clonagem Molecular , Ativação do Canal Iônico , Miocárdio/metabolismo , Canais de Potássio/química , Canais de Potássio/fisiologia , Animais , Eletrofisiologia , Furões , Ventrículos do Coração , Oócitos/metabolismo , Fatores de Tempo , Xenopus laevis
18.
J Physiol ; 485 ( Pt 1): 59-71, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7658383

RESUMO

1. Using the two-microelectrode, 'cut open' oocyte, and 'torn off' macropatch voltage clamp techniques, we studied the blocking effects of 4-aminopyridine (4-AP) on two cloned K+ channels expressed in Xenopus oocytes, an inactivating K+ channel isolated from ferret ventricle (FK1), and its NH2-terminal deletion mutant (delta NCO) which lacks fast N-type inactivation. 2. Experiments with a permanently charged, impermeant 4-AP derivative, 4-aminopyridine-methyliodide, indicated that the cationic form of 4-AP blocks at an intracellular site. 3. Block accumulated from pulse to pulse and was sensitive to the applied potential during hyperpolarizing deactivating pulses, indicating trapping of 4-AP in deactivated channels. For long trains of depolarizing pulses (-90 to +50 mV, 0.1 Hz), 4-AP block increased with decreasing pulse duration. Block of FK1 was much more sensitive to pulse duration than was block of delta NCO, consistent with competition between N-type inactivation and 4-AP binding. 4. To elucidate these mechanisms further, in the absence of fast N-type inactivation the following results were obtained on delta NCO channels: (1) application of 4-AP caused the appearance of apparent inactivation; (2) 4-AP, however, did not cause cross-over of deactivating tail currents; (3) 4-AP block developed with time for potentials positive to -40 mV; and (4) trapping of 4-AP by delta NCO was insensitive to the degree of C-type inactivation. 5. We conclude that the kinetics of 4-AP block of FK1 and delta NCO channels cannot be accounted for by either a pure open channel or closed channel blocking scheme.


Assuntos
4-Aminopiridina/farmacologia , Miocárdio/metabolismo , Oócitos/metabolismo , Canais de Potássio/efeitos dos fármacos , Animais , Eletrofisiologia , Feminino , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Microeletrodos , Conformação Molecular , Miocárdio/citologia , Oócitos/efeitos dos fármacos , RNA Complementar/biossíntese , Xenopus laevis
19.
J Biol Chem ; 270(11): 6272-7, 1995 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-7890764

RESUMO

Voltage-gated potassium channel beta subunits are cytoplasmic proteins that co-purify with the pore-forming alpha subunits. One of these subunits, Kv beta 1 from rat brain, was previously demonstrated to increase the rate of inactivation of Kv1.1 and Kv1.4 when co-expressed in Xenopus oocytes. We have cloned and characterized a novel voltage-gated K+ channel beta subunit. The cDNA, designated Kv beta 3, has a 408-amino acid open reading frame. It possesses a unique 79-amino acid N-terminal leader, but is identical with rat Kv beta 1 over the 329 C-terminal amino acids. The Kv beta 3 transcript was found in many tissues, but was most abundant in aorta and left ventricle of the heart. Co-expression of Kv beta 3 with K+ channel alpha subunits shows that this beta subunit can increase the rate of inactivation from 4- to 7-fold in a Kv1.4 or Shaker B channel. Kv beta 3 had no effect on Kv1.1, unlike Kv beta 1 which can increase rate of inactivation of this alpha subunit more than 100-fold. Other kinetic parameters were unaffected. This study shows that voltage-gated K+ channel beta subunits are present outside the central nervous system, and that at least one member of this family selectively modulates inactivation of K+ channel alpha subunits.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais de Potássio/fisiologia , Sequência de Aminoácidos , Animais , Aorta/metabolismo , Sequência de Bases , Bovinos , Clonagem Molecular , Primers do DNA , Furões , Expressão Gênica , Ventrículos do Coração , Humanos , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta , Substâncias Macromoleculares , Potenciais da Membrana , Dados de Sequência Molecular , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Bloqueadores dos Canais de Potássio , Canais de Potássio/biossíntese , RNA Mensageiro/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Transcrição Gênica
20.
Adv Exp Med Biol ; 382: 11-22, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8540388

RESUMO

We studied the blocking effects of 4-aminopyridine (4-AP) on a Kv1.4 K+ channel. A permanently charged 4-AP derivative only produced block when applied intracellularly. 4-AP block accumulated from pulse to pulse indicating trapping of 4-AP in deactivated channels. For long trains of depolarizing pulses, 4-AP block increased with decreasing pulse duration. This increase took many pulses (> 10) to accumulate and was relieved by two to three subsequent pulses of 500 msec duration. We conclude that the time- and voltage-dependence of 4-AP block can not be accounted for solely by either simple pure open channel or pure closed channel blocking schemes. We propose that the data can be explained by a model in which 4-AP binding is most stable when the channel has a symmetric arrangement in the binding regions.


Assuntos
4-Aminopiridina/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Miocárdio/metabolismo , Bloqueadores dos Canais de Potássio , Animais , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Eletrofisiologia , Furões , Canais de Potássio/genética , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA