Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 97(1): 89-134, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27807201

RESUMEN

Ion channels represent the molecular entities that give rise to the cardiac action potential, the fundamental cellular electrical event in the heart. The concerted function of these channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syndrome. We examine mechanisms of dysfunction associated with three critical cardiac currents that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the study of mutations that cause congenital Long QT Syndrome, the scientific community has advanced understanding of ion channel structure-function relationships, physiology, and pharmacological response to clinically employed and experimental pharmacological agents. Our understanding of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-driven clinical management of the disease has improved patient care as precision medicine becomes even more a reality.


Asunto(s)
Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Animales , Humanos
2.
J Physiol ; 588(Pt 14): 2643-55, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20498233

RESUMEN

Computer modelling has emerged as a particularly useful tool in understanding the physiology and pathophysiology of cardiac tissues. Models of ventricular, atrial and nodal tissue have evolved and include detailed ion channel kinetics and intercellular Ca(2+) handling. Purkinje fibre cells play a central role in the electrophysiology of the heart and in the genesis of cardiac arrhythmias. In this study, a new computational model has been constructed that incorporates the major membrane currents that have been isolated in recent experiments using Purkinje fibre cells. The model, which integrates mathematical models of human ion channels based on detailed biophysical studies of their kinetic and voltage-dependent properties, recapitulates distinct electrophysiological characteristics unique to Purkinje fibre cells compared to neighbouring ventricular myocytes. These characteristics include automaticity, hyperpolarized voltage range of the action potential plateau potential, and prolonged action potential duration. Simulations of selective ion channel blockade reproduce responses to pharmacological challenges characteristic of isolated Purkinje fibres in vitro, and importantly, the model predicts that Purkinje fibre cells are prone to severe arrhythmogenic activity in patients harbouring long QT syndrome 3 but much less so for other common forms of long QT. This new Purkinje cellular model can be a useful tool to study tissue-specific drug interactions and the effects of disease-related ion channel dysfunction on the cardiac conduction system.


Asunto(s)
Simulación por Computador , Síndrome de QT Prolongado/fisiopatología , Modelos Neurológicos , Ramos Subendocárdicos/fisiología , Potenciales de Acción/efectos de los fármacos , Arritmias Cardíacas/fisiopatología , Humanos , Canales Iónicos/efectos de los fármacos , Canales Iónicos/fisiología , Moduladores del Transporte de Membrana/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Ramos Subendocárdicos/efectos de los fármacos
3.
J Mol Cell Cardiol ; 47(2): 326-34, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19371746

RESUMEN

Long QT syndrome variant 3 (LQT-3) is a channelopathy in which mutations in SCN5A, the gene coding for the primary heart Na(+) channel alpha subunit, disrupt inactivation to elevate the risk of mutation carriers for arrhythmias that are thought to be calcium (Ca(2+))-dependent. Spontaneous arrhythmogenic diastolic activity has been reported in myocytes isolated from mice harboring the well-characterized Delta KPQ LQT-3 mutation but the link to altered Ca(2+) cycling related to mutant Na(+) channel activity has not previously been demonstrated. Here we have investigated the relationship between elevated sarcoplasmic reticulum (SR) Ca(2+) load and induction of spontaneous diastolic inward current (I(TI)) in myocytes expressing Delta KPQ Na(+) channels, and tested the sensitivity of both to the antianginal compound ranolazine. We combined whole-cell patch clamp measurements, imaging of intracellular Ca(2+), and measurement of SR Ca(2+) content using a caffeine dump methodology. We compared the Ca(2+) content of Delta KPQ(+/-) myocytes displaying I(TI) to those without spontaneous diastolic activity and found that I(TI) induction correlates with higher sarcoplasmic reticulum (SR) Ca(2+). Both spontaneous diastolic I(TI) and underlying Ca(2+) waves are inhibited by ranolazine at concentrations that preferentially target I(NaL) during prolonged depolarization. Furthermore, ranolazine I(TI) inhibition is accompanied by a small but significant decrease in SR Ca(2+) content. Our results provide the first direct evidence that induction of diastolic transient inward current (I(TI)) in Delta KPQ(+/-) myocytes occurs under conditions of elevated SR Ca(2+) load.


Asunto(s)
Acetanilidas/farmacología , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Diástole/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Síndrome de QT Prolongado/fisiopatología , Piperazinas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Ratones , Mutación/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ranolazina
4.
Science ; 242(4875): 67-9, 1988 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-2845575

RESUMEN

The enzymes adenosine 3',5'-monophosphate (cAMP)-dependent protein kinase (protein kinase A) and protein kinase C regulate the activity of a diverse group of cellular proteins including membrane ion channel proteins. When protein kinase A was stimulated in cardiac ventricular myocytes with the membrane-soluble cAMP analog 8-chlorphenylthio cAMP (8-CPT cAMP), the amplitude of the delayed-rectifier potassium current (IK) doubled when recorded at 32 degrees C but was not affected at 22 degrees C. In contrast, modulation of the calcium current (ICa) by 8-CPT cAMP was independent of temperature with similar increases in ICa occurring at both temperatures. Stimulation of protein kinase C by phorbol 12,13-dibutyrate also enhanced IK in a temperature-dependent manner but failed to increase ICa at either temperature. Thus, cardiac delayed-rectifier potassium but not calcium channels are regulated by two distinct protein kinases in a similar temperature-dependent fashion.


Asunto(s)
AMP Cíclico/análogos & derivados , Corazón/fisiología , Canales de Potasio/fisiología , Proteína Quinasa C/metabolismo , Proteínas Quinasas/metabolismo , Tionucleótidos/farmacología , Animales , AMP Cíclico/farmacología , Cobayas , Homeostasis , Técnicas In Vitro , Cinética , Potenciales de la Membrana , Termodinámica , Función Ventricular
6.
Circ Res ; 88(7): 740-5, 2001 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-11304498

RESUMEN

Variant 3 of the congenital long-QT syndrome (LQTS-3) is caused by mutations in the gene encoding the alpha subunit of the cardiac Na(+) channel. In the present study, we report a novel LQTS-3 mutation, E1295K (EK), and describe its functional consequences when expressed in HEK293 cells. The clinical phenotype of the proband indicated QT interval prolongation in the absence of T-wave morphological abnormalities and a steep QT/R-R relationship, consistent with an LQTS-3 lesion. However, biophysical analysis of mutant channels indicates that the EK mutation changes channel activity in a manner that is distinct from previously investigated LQTS-3 mutations. The EK mutation causes significant positive shifts in the half-maximal voltage (V(1/2)) of steady-state inactivation and activation (+5.2 and +3.4 mV, respectively). These gating changes shift the window of voltages over which Na(+) channels do not completely inactivate without altering the magnitude of these currents. The change in voltage dependence of window currents suggests that this alteration in the voltage dependence of Na(+) channel gating may cause marked changes in action potential duration because of the unique voltage-dependent rectifying properties of cardiac K(+) channels that underlie the plateau and terminal repolarization phases of the action potential. Na(+) channel window current is likely to have a greater effect on net membrane current at more positive potentials (EK channels) where total K(+) channel conductance is low than at more negative potentials (wild-type channels), where total K(+) channel conductance is high. These findings suggest a fundamentally distinct mechanism of arrhythmogenesis for congenital LQTS-3.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Corazón/fisiopatología , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/genética , Canales de Sodio/genética , Adolescente , Sustitución de Aminoácidos , Arritmias Cardíacas/genética , Línea Celular , Secuencia Conservada , Análisis Mutacional de ADN , Electrocardiografía , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Fenotipo , Sodio/metabolismo , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología , Transfección
7.
Handb Exp Pharmacol ; (171): 287-304, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16610349

RESUMEN

The congenital long QT syndrome is a rare disease in which inherited mutations of genes coding for ion channel subunits, or channel interacting proteins, delay repolarization of the human ventricle and predispose mutation carriers to the risk of serious or fatal arrhythmias. Though a rare disorder, the long QT syndrome has provided invaluable insight from studies that have bridged clinical and pre-clinical (basic science) medicine. In this brief review, we summarize some of the key clinical and genetic characteristics of this disease and highlight novel findings about ion channel structure, function, and the causal relationship between channel dysfunction and human disease, that have come from investigations of this disorder.


Asunto(s)
Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/genética , Mutación , Canales de Sodio/genética , Potenciales de Acción , Anestésicos Locales/farmacología , Corazón/fisiología , Humanos , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/etiología , Factores de Riesgo , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/química , Canales de Sodio/efectos de los fármacos , Canales de Sodio/fisiología , Relación Estructura-Actividad
8.
Circulation ; 107(25): 3216-22, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12796143

RESUMEN

BACKGROUND: Mutations in SCN5A, the gene coding for the human cardiac Na+ channel alpha-subunit, are associated with variant 3 of the long-QT syndrome (LQT-3). Several LQT-3 mutations promote a mode of Na+ channel gating in which a fraction of channels fail to inactivate, contributing sustained Na+ channel current (Isus), which can delay repolarization and prolong the QT interval. Here, we investigate the possibility that stimulation of protein kinase C (PKC) may modulate Isus, which is prominent in disease-related Na+ channel mutations. METHODS AND RESULTS: We measured the effects of PKC stimulation on Na+ currents in human embryonic kidney (HEK 293) cells expressing 3 previously reported disease-associated Na+ channel mutations (Y1795C, Y1795H, and DeltaKPQ). We find that the PKC activator 1-oleoyl-2-acetyl-sn-glycerol (OAG) significantly reduced Isus in the mutant but not wild-type channels. The effect of OAG on Isus was reduced by the PKC inhibitor staurosporine (2.5 micromol/L), ablated by the mutation S1503A, and mimicked by the mutation S1503D. Isus recorded in myocytes isolated from mice expressing DeltaKPQ channels was similarly inhibited by OAG exposure or stimulation of alpha1-adrenergic receptors by phenylephrine. The actions of phenylephrine on Isus were blocked by the PKC inhibitor chelerythrine. CONCLUSIONS: We conclude that stimulation of PKC inhibits channel bursting in disease-linked mutations via phosphorylation-induced alteration of the charge at residue 1503 of the Na+ channel alpha-subunit. Sympathetic nerve activity may contribute directly to suppression of mutant channel bursting via alpha-adrenergic receptor-mediated stimulation of PKC.


Asunto(s)
Activación del Canal Iónico , Síndrome de QT Prolongado/fisiopatología , Proteína Quinasa C/metabolismo , Canales de Sodio/metabolismo , Animales , Células Cultivadas , Diglicéridos/farmacología , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Síndrome de QT Prolongado/genética , Ratones , Ratones Mutantes , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Sodio/metabolismo , Canales de Sodio/genética , Relación Estructura-Actividad , Sistema Nervioso Simpático/fisiología , Transfección
9.
Circulation ; 102(5): 584-90, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10920073

RESUMEN

BACKGROUND: D1790G, a mutation of SCN5A, the gene that encodes the human Na(+) channel alpha-subunit, is linked to 1 form of the congenital long-QT syndrome (LQT-3). In contrast to other LQT-3-linked SCN5A mutations, D1790G does not promote sustained Na(+) channel activity but instead alters the kinetics and voltage-dependence of the inactivated state. METHODS AND RESULTS: We modeled the cardiac ventricular action potential (AP) using parameters and techniques described by Luo and Rudy as our control. On this background, we modified only the properties of the voltage-gated Na(+) channel according to our patch-clamp analysis of D1790G channels. Our results indicate that D1790G-induced changes in Na(+) channel activity prolong APs in a steeply heart rate-dependent manner not directly due to changes in Na(+) entry through mutant channels but instead to alterations in the balance of net plateau currents by modulation of calcium-sensitive exchange and ion channel currents. CONCLUSIONS: We conclude that the D1790G mutation of the Na(+) channel alpha-subunit can prolong the cardiac ventricular AP despite the absence of mutation-induced sustained Na(+) channel current. This prolongation is calcium-dependent, is enhanced at slow heart rates, and at sufficiently slow heart rate triggers arrhythmogenic early afterdepolarizations.


Asunto(s)
Síndrome de QT Prolongado/genética , Mutación Puntual , Canales de Sodio/genética , Canales de Sodio/fisiología , Función Ventricular/fisiología , Potenciales de Acción , Calcio/metabolismo , Línea Celular , Membrana Celular/fisiología , Frecuencia Cardíaca , Humanos , Cinética , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Proteínas Recombinantes/metabolismo , Transfección
10.
Circulation ; 102(8): 921-5, 2000 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-10952963

RESUMEN

BACKGROUND: Multiple mutations of SCN5A, the gene that encodes the human Na(+) channel alpha-subunit, are linked to 1 form of the congenital long-QT syndrome (LQT-3). D1790G (DG), an LQT-3 mutation of the C-terminal region of the Na(+) channel alpha-subunit, alters steady-state inactivation of expressed channels but does not promote sustained Na(+) channel activity. Recently, flecainide, but not lidocaine, has been found to correct the disease phenotype, delayed ventricular repolarization, in DG carriers. METHODS AND RESULTS: To understand the molecular basis of this difference, we studied both drugs using wild-type (WT) and mutant Na(+) channels expressed in HEK 293 cells. The DG mutation conferred a higher sensitivity to lidocaine (EC(50), WT=894 and DG=205 micromol/L) but not flecainide tonic block in a concentration range that is not clinically relevant. In contrast, in a concentration range that is therapeutically relevant, DG channels are blocked selectively by flecainide (EC(50), WT=11.0 and DG=1.7 micromol/L), but not lidocaine (EC(50), WT=318.0 and DG=176 micromol/L) during repetitive stimulation. CONCLUSIONS: These results (1) demonstrate that the DG mutation confers a unique pharmacological response on expressed channels; (2) suggest that flecainide use-dependent block of DG channels underlies its therapeutic effects in carriers of this gene mutation; and (3) suggest a role of the Na(+) channel alpha-subunit C-terminus in the flecainide/channel interaction.


Asunto(s)
Antiarrítmicos/farmacología , Síndrome de QT Prolongado/genética , Bloqueadores de los Canales de Sodio , Canales de Sodio/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Flecainida/farmacología , Ligamiento Genético , Humanos , Cinética , Lidocaína/farmacología , Síndrome de QT Prolongado/tratamiento farmacológico , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.5 , Mutación Puntual , Especificidad por Sustrato
11.
Circulation ; 101(14): 1698-706, 2000 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-10758053

RESUMEN

BACKGROUND: Mutations in the cardiac sodium channel gene (SCN5A) can cause one variant of the congenital long-QT syndrome. The effects of some of these mutations on the alpha-subunit channel properties can be blocked by type Ib antiarrhythmic drugs. Recently, we have described a new SCN5A mutation (D1790G) that affects the channel properties in a manner suggesting that sodium blockers of the Ib type will be ineffective in carriers of this mutation. Hence, the ECG effects of flecainide-acetate, a type Ic sodium blocker, were evaluated in carriers of this mutation. METHODS AND RESULTS: Eight asymptomatic mutation carriers and 5 control subjects were studied. Intravenous lidocaine was tested first in only 2 mutation carriers and had no significant effect on any ECG parameter. Flecainide significantly shortened all heart rate-corrected repolarization duration parameters only in carriers and not in control subjects: QT(c) shortened by 9.5% (from 517+/-45 to 468+/-36 ms, P=0.011), and the S-offset to T-onset interval shortened by 64.7% (from 187+/-88 to 66+/-50 ms, P=0.0092). Flecainide also normalized the marked baseline repolarization dispersion in most mutation carriers. These effects among carriers were maintained during long-term (9 to 17 months) outpatient flecainide therapy with no adverse effects. CONCLUSIONS: This report is the first to describe SCN5A mutation carriers who significantly responded to flecainide therapy yet did not respond to lidocaine. These results have important implications for long-QT allele-specific therapeutic strategies.


Asunto(s)
Antiarrítmicos/uso terapéutico , Flecainida/uso terapéutico , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/genética , Mutación , Canales de Sodio/genética , Estudios de Casos y Controles , Electrocardiografía , Femenino , Corazón/efectos de los fármacos , Corazón/fisiopatología , Heterocigoto , Humanos , Inyecciones Intravenosas , Lidocaína/uso terapéutico , Síndrome de QT Prolongado/fisiopatología , Masculino , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje
12.
Circulation ; 103(9): 1303-10, 2001 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-11238277

RESUMEN

BACKGROUND: Sodium channels isolated from mammalian brain are composed of alpha-, beta(1)-, and beta(2)-subunits. The composition of sodium channels in cardiac muscle, however, has not been defined, and disagreement exists over which beta-subunits are expressed in the myocytes. Some investigators have demonstrated beta(1) expression in heart. Others have not detected any auxiliary subunits. On the basis of Northern blot analysis of total RNA, beta(2) expression has been thought to be exclusive to neurons and absent from cardiac muscle. METHODS AND RESULTS: The goal of this study was to define the subunit composition of cardiac sodium channels in myocytes. We show that cardiac sodium channels are composed of alpha-, beta(1)-, and beta(2)-subunits. Nav1.5 and Nav1.1 are expressed in myocytes and are associated with beta(1)- and beta(2)-subunits. Immunocytochemical localization of Nav1.1, beta(1), and beta(2) in adult heart sections showed that these subunits are expressed at the Z lines, as shown previously for Nav1.5. Coexpression of Nav1.5 with beta(2) in transfected cells resulted in no detectable changes in sodium current. CONCLUSIONS: Cardiac sodium channels are composed of alpha- (Nav1.1 or Nav1.5), beta(1)-, and beta(2)-subunits. Although beta(1)-subunits modulate cardiac sodium channel current, beta(2)-subunit function in heart may be limited to cell adhesion.


Asunto(s)
Miocardio/metabolismo , Canales de Sodio/fisiología , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos , Encéfalo/metabolismo , Línea Celular , Electrofisiología , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Miocardio/citología , Subunidades de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Sodio/genética , Canales de Sodio/inmunología
13.
J Gen Physiol ; 89(4): 629-44, 1987 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2438373

RESUMEN

We have measured the density of negative surface charges near the voltage sensor for inactivation gating of (L-type) Ca channels in intact calf Purkinje fibers and in isolated myocytes from guinea pig and rat ventricles. Divalent cation-induced changes in the half-maximal voltage for inactivation were determined and were well described by curves predicted by surface potential theory. We measured shifts in inactivation induced by Ca, Sr, and Ba in the single cells, and by Sr in the Purkinje fibers. All of the data were consistent with an estimated negative surface charge density of 1 electronic charge per 250 A2. In addition, the data suggest that Ca, but neither Ba nor Sr, binds to the negative charges with an association constant on the order of 1 M-1. We find that divalent ion-induced changes in surface potential can account for most of the antagonism between these ions and Ca channel block by 1,4-dihydropyridines.


Asunto(s)
Calcio/fisiología , Corazón/fisiología , Canales Iónicos/fisiología , Nifedipino/análogos & derivados , Animales , Bario/farmacología , Bovinos , Conductividad Eléctrica , Cobayas , Canales Iónicos/efectos de los fármacos , Potenciales de la Membrana , Nifedipino/antagonistas & inhibidores , Nifedipino/farmacología , Nisoldipino , Ramos Subendocárdicos/fisiología , Ratas , Sarcolema/fisiología , Estroncio/farmacología
14.
J Gen Physiol ; 93(6): 1109-27, 1989 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2549176

RESUMEN

We have investigated the modulation of L-type calcium channel currents in isolated ventricular cells by the dihydropyridine derivative amlodipine, a weak base with a pKa of 8.6. Under conditions that favor neutral drug molecules, amlodipine block resembles other, previously described, neutral dihydropyridine derivatives: block is more pronounced at depolarized voltages, repetitive pulsing is not needed to promote block, and recovery is complete at hyperpolarized voltages. When the drug is ionized, depolarized voltages still enhance block, however, the time course is slow and speeded by repetitive pulses that open channels. Recovery from block by ionized drug molecules is very slow and incomplete, but can be rapidly modified by changes in external hydrogen ion concentration. We conclude from these observations that the degree of ionization of the drug molecule can affect access to the dihydropyridine receptor and that external protons can interact with the drug-receptor complex even if channels are blocked and closed. These observations place limitations on the location of this receptor in the ventricular cell membrane.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Miocardio/citología , Nifedipino/análogos & derivados , Receptores Nicotínicos/efectos de los fármacos , Amlodipino , Animales , Canales de Calcio , Cobayas , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Nifedipino/farmacología
15.
J Gen Physiol ; 91(5): 641-57, 1988 May.
Artículo en Inglés | MEDLINE | ID: mdl-2458428

RESUMEN

We have investigated the effects of H ions on (L-type) Ca channel current in isolated ventricular cells. We find that the current amplitude is enhanced in solutions that are alkaline relative to pH 7.4 and reduced in solutions acidic to this pH. We measured pH0-induced shifts in channel gating and analyzed our results in terms of surface potential theory. The shifts are well described by changes in surface potential caused by the binding of H ions to negative charges on the cell surface. The theory predicts a pK of 5.8 for this binding. Gating shifts alone cannot explain all of our observations on modulation of current amplitude. Our results suggest that an additional mechanism contributes to modification of the current amplitude.


Asunto(s)
Calcio/metabolismo , Canales Iónicos/metabolismo , Miocardio/metabolismo , Protones , Animales , Conductividad Eléctrica , Femenino , Cobayas , Concentración de Iones de Hidrógeno , Masculino , Potenciales de la Membrana , Miocardio/citología , Ratas , Ratas Endogámicas
16.
J Gen Physiol ; 84(5): 705-26, 1984 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6096480

RESUMEN

We have studied the influence of divalent cations on Ca channel current in the calf cardiac Purkinje fiber to determine whether this current inactivates by voltage- or Ca-mediated mechanisms, or by a combination of the two. We measured the reversal (or zero current) potential of the current when Ba, Sr, or Ca were the permeant divalent cations and determined that depletion of charge carrier does not account for time-dependent relaxation of Ca channel current in these preparations. Inactivation of Ca channel current persists when Ba or Sr replaces Ca as the permeant divalent cation, but the voltage dependence of the rate of inactivation is markedly changed. This effect cannot be explained by changes in external surface charge. Instead, we interpret the results as evidence that inactivation is both voltage and Ca dependent. Inactivation of Sr or Ba currents reflects a voltage-dependent process. When Ca is the divalent charge carrier, an additional effect is observed: the rate of inactivation is increased as Ca enters during depolarizing pulses, perhaps because of an additional Ca-dependent mechanism.


Asunto(s)
Calcio/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Canales Iónicos/fisiología , Ramos Subendocárdicos/metabolismo , Animales , Bario/metabolismo , Fenómenos Biomecánicos , Calcio/fisiología , Bovinos , Conductividad Eléctrica , Electrofisiología , Homeostasis , Iontoforesis , Cinética , Compuestos de Amonio Cuaternario/farmacología , Estroncio/metabolismo , Propiedades de Superficie , Factores de Tiempo
17.
J Gen Physiol ; 102(4): 693-712, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8270910

RESUMEN

We have investigated the interactions between extracellular divalent cations and the ATP-sensitive potassium channel in single guinea pig ventricular cells and found that, under whole-cell patch clamp recording conditions, extracellularly applied Co2+, Cd2+, and Zn2+ block current through the ATP-sensitive K channel (IKATP). The respective Kd's for block of IKATP by Cd2+ and Zn2+ are 28 and 0.46 microM. The Kd for Co2+ is > 200 microM. Extracellular Ca2+ and Mg2+ appear to have no effect at concentrations up to 1 and 2 mM, respectively. Block of IKATP by extracellular cations is not voltage dependent, and both onset and recovery from block occur within seconds. Single-channel experiments using the inside-out patch configuration show that internally applied Cd2+ and Zn2+ are not effective blockers of IKATP. Experiments in the outside-out patch configuration confirm that the divalent cations interact directly with IKATP channel activity. Our study also shows that this block of IKATP is dependent on intracellular ATP concentrations. Under whole-cell conditions, when cells are dialyzed with [ATP]pipette = 0, the degree of cation block is reduced. This dependence on intracellular ATP was confirmed at the single-channel level by experiments in excised, inside-out patch configurations. Our results show that some, but not all, divalent cations inhibit current through IKATP channels by binding to sites that are not within the transmembrane electric field, but are on the extracellular membrane surface. The interdependence of internal ATP and external divalent cation binding is consistent with an allosteric interaction between two binding sites and is highly suggestive of a modulatory mechanism involving conformational change of the channel protein.


Asunto(s)
Adenosina Trifosfato/farmacología , Adenosina Trifosfato/fisiología , Cationes Bivalentes/farmacología , Membranas Intracelulares/metabolismo , Miocardio/metabolismo , Bloqueadores de los Canales de Potasio , Regulación Alostérica , Animales , Sitios de Unión , Cadmio/farmacología , Guanidinas/farmacología , Cobayas , Concentración Osmolar , Pinacidilo , Canales de Potasio/efectos de los fármacos , Zinc/farmacología
18.
J Gen Physiol ; 66(2): 169-92, 1975 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1176946

RESUMEN

We studied the influence of Mn, La, and D600 on action potentials and plateau currents in cardiac Purkinje fibers. The Ca antagonists each abolished the second inward current, but they failed to act selectively. Voltage clamp experiments revealed two additional effects: decrease of slow outward current (iotachi) activation, and increase of net outward time-independent plateau current. These effects occurred at inhibitor concentrations used in earlier studies, and were essential to the reconstruction of observed Ca antagonist effects on electrical activity. The inhibitory influence of Mn, La, and D600 on iotachi suggested that iotachi activation might depend upon prior Ca entry. This hypothesis was not supported, however, when [Ca]omicron was varied: elevating [Ca]omicron enhanced Ca entry, but iotachi was nevertheless depressed. Thus, the results suggested instead that Ca antagonists and Ca ions have rather similar effects on iotachi, possibly mediated by changes in membrane surface charge.


Asunto(s)
Calcio/antagonistas & inhibidores , Sistema de Conducción Cardíaco/fisiología , Ramos Subendocárdicos/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/farmacología , Bovinos , Galopamilo/farmacología , Técnicas In Vitro , Lantano/farmacología , Manganeso/farmacología , Ramos Subendocárdicos/efectos de los fármacos
19.
J Gen Physiol ; 67(5): 599-617, 1976 May.
Artículo en Inglés | MEDLINE | ID: mdl-1271045

RESUMEN

It is well known that cardiac action potentials are shortened by increasing the external calcium concentration (Cao). The shortening is puzzling since Ca ions are thought to carry inward current during the plateau. We therefore studied the effects of Cao on action potentials and membrane currents in short Purkinje fiber preparations. Two factors favor the earlier repolarization. First, calcium-rich solutions generally raise the plateau voltage; in turn, the higher plateau level accelerates time- and voltage-dependent current changes which trigger repolarization. Increases in plateau height imposed by depolarizing current consistently produced shortening of the action potential. The second factor in the action of Ca ions involves iK1, the background K current (inward rectifier). Raising Cao enhances iK1 and thus favors faster repolarization. The Ca-sensitive current change was identified as an increase in iK1 by virtue of its dependence on membrane potential and Ko. A possible third factor was considered and ruled out: unlike epinephrine, calcium-rich solutions do not enhance slow outward plateau current, ikappa. These results are surprising in showing that calcium ions and epinephrine act quite differently on repolarizing currents, even though they share similar effects on the height and duration of the action potential.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Calcio/farmacología , Sistema de Conducción Cardíaco/efectos de los fármacos , Ramos Subendocárdicos/efectos de los fármacos , Animales , Bovinos , Epinefrina/farmacología , Ramos Subendocárdicos/metabolismo , Factores de Tiempo
20.
J Gen Physiol ; 117(1): 43-52, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134230

RESUMEN

I(Ks), a slowly activating delayed rectifier K(+) current through channels formed by the assembly of two subunits KCNQ1 (KvLQT1) and KCNE1 (minK), contributes to the control of the cardiac action potential duration. Coassembly of the two subunits is essential in producing the characteristic and physiologically critical kinetics of assembled channels, but it is not yet clear where or how these subunits interact. Previous investigations of external access to the KCNE1 protein in assembled I(Ks) channels relied on occlusion of the pore by extracellular application of TEA(+), despite the very low TEA(+) sensitivity (estimated EC(50) > 100 mM) of channels encoded by coassembly of wild-type KCNQ1 with the wild type (WT) or a series of cysteine-mutated KCNE1 constructs. We have engineered a high affinity TEA(+) binding site into the h-KCNQ1 channel by either a single (V319Y) or double (K318I, V319Y) mutation, and retested it for pore-delimited access to specific sites on coassembled KCNE1 subunits. Coexpression of either KCNQ1 construct with WT KCNE1 in Chinese hamster ovary cells does not alter the TEA(+) sensitivity of the homomeric channels (IC(50) approximately 0.4 mM [TEA(+)](out)), providing evidence that KCNE1 coassembly does not markedly alter the structure of the outer pore of the KCNQ1 channel. Coexpression of a cysteine-substituted KCNE1 (F54C) with V319Y significantly increases the sensitivity of channels to external Cd(2+), but neither the extent of nor the kinetics of the onset of (or the recovery from) Cd(2+) block was affected by [TEA(+)](o) at 10x the IC(50) for channel block. These data strongly suggest that access of Cd(2+) to the cysteine-mutated site on KCNE1 is independent of pore occlusion caused by TEA(+) binding to the outer region of the KCNE1/V319Y pore, and that KCNE1 does not reside within the pore region of the assembled channels.


Asunto(s)
Corazón/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/farmacología , Canales de Potasio/fisiología , Potenciales de Acción , Animales , Sitios de Unión , Células CHO , Cadmio/metabolismo , Técnicas de Cultivo de Célula , Cricetinae , Cisteína/metabolismo , Humanos , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Mutación Puntual , Canales de Potasio/química , Canales de Potasio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA