Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Clin Invest ; 91(2): 693-701, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8381826

RESUMEN

We demonstrated recently that isoproterenol enhanced the cardiac voltage-dependent sodium currents (INa) in rabbit ventricular myocytes through dual G-protein regulatory pathways. In this study, we tested the hypothesis that isoproterenol reverses the sodium channel blocking effects of class I antiarrhythmic drugs through modulation of INa. The experiments were performed in rabbit ventricular myocytes using whole-cell patch-clamp techniques. Reversal of lidocaine suppression of INa by isoproterenol (1 microM) was significant at various concentrations of lidocaine (20, 65, and 100 microM, P < 0.05). The effects of isoproterenol were voltage dependent, showing reversal of INa suppression by lidocaine at normal and hyperpolarized potentials (negative to -80 mV) but not at depolarized potentials. Isoproterenol enhanced sodium channel availability but did not alter the steady state activation or inactivation of INa nor did it improve sodium channel recovery in the presence of lidocaine. The physiological significance of the single cell INa findings were corroborated by measurements of conduction velocities using an epicardial mapping system in isolated rabbit hearts. Lidocaine (10 microM) significantly suppressed epicardial impulse conduction in both longitudinal (theta L, 0.430 +/- 0.024 vs. 0.585 +/- 0.001 m/s at baseline, n = 7, P < 0.001) and transverse (theta T, 0.206 +/- 0.012 vs. 0.257 +/- 0.014 m/s at baseline, n = 8, P < 0.001) directions. Isoproterenol (0.05 microM) significantly reversed the lidocaine effects with theta L of 0.503 +/- 0.027 m/s and theta T of 0.234 +/- 0.015 m/s (P = 0.014 and 0.004 compared with the respective lidocaine measurements). These results suggest that enhancement of INa is an important mechanism by which isoproterenol reverses the effects of class I antiarrhythmic drugs.


Asunto(s)
Corazón/efectos de los fármacos , Isoproterenol/farmacología , Lidocaína/antagonistas & inhibidores , Canales de Sodio/efectos de los fármacos , Animales , Células Cultivadas , Corazón/fisiología , Sistema de Conducción Cardíaco/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Conejos
2.
J Gen Physiol ; 109(2): 169-80, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9041446

RESUMEN

Dihydropyridines (DHPs) are well known for their effects on L-typed voltage-dependent Ca2+ channels, However, these drugs also affect other voltage-dependent ion channels, including Shaker K+ channels. We examined the effects of DHPs on the Shaker K+ channels expressed in Xenopus oocytes. Intracellular applications of DHPs quickly and reversibly induced apparent inactivation in the Shaker K+ mutant channels with disrupted N- and C-type inactivation. We found that DHPs interact with the open state of the channel as evidenced by the decreased mean open time. The DHPs effects are voltage-dependent, becoming more effective with hyperpolarization. A model which involves binding of two DHP molecules to the channel is consistent with the results obtained in our experiments.


Asunto(s)
Dihidropiridinas/farmacología , Oocitos/metabolismo , Canales de Potasio/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Estimulación Eléctrica , Electrofisiología , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Nifedipino/farmacología , Técnicas de Placa-Clamp , Canales de Potasio/genética , Xenopus
3.
J Gen Physiol ; 88(6): 777-98, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2432158

RESUMEN

Individual myocytes were isolated from bullfrog atrium by enzymatic and mechanical dispersion, and a one-microelectrode voltage clamp was used to record the slow outward K+ currents. In normal [K+]o (2.5 mM), the slow outward current tails reverse between -95 and -100 mV. This finding, and the observed 51-mV shift of Erev/10-fold change in [K+]o, strongly suggest that the "delayed rectifier" in bullfrog atrial cells is a K+ current. This current, IK, plays an important role in initiating repolarization, and it is distinct from the quasi-instantaneous, inwardly rectifying background current, IK. In atrial cells, IK does not exhibit inactivation, and very long depolarizing clamp steps (20 s) can be applied without producing extracellular K+ accumulation. The possibility of [K+]o accumulation contributing to these slow outward current changes was assessed by (a) comparing reversal potentials measured after short (2 s) and very long (15 s) activating prepulses, and (b) studying the kinetics of IK at various holding potentials and after systematically altering [K+]o. In the absence of [K+]o accumulation, the steady state activation curve (n infinity) and fully activated current-voltage (I-V) relation can be obtained directly. The threshold of the n infinity curve is near -50 mV, and it approaches a maximum at +20 mV; the half-activation point is approximately -16 mV. The fully activated I-V curve of IK is approximately linear in the range -40 to +30 mV. Semilog plots of the current tails show that each tail is a single-exponential function, which suggests that only one Hodgkin-Huxley conductance underlies this slow outward current. Quantitative analysis of the time course of onset of IK and of the corresponding envelope of tails demonstrate that the activation variable, n, must be raised to the second power to fit the sigmoid onset accurately. The voltage dependence of the kinetics of IK was studied by recording and curve-fitting activating and deactivating (tail) currents. The resulting 1/tau n curve is U-shaped and somewhat asymmetric; IK exhibits strong voltage dependence in the diastolic range of potentials. Changes in the [Ca2+]o in the superfusing Ringer's, and/or addition of La3+ to block the transmembrane Ca2+ current, show that the time course and magnitude of IK are not significantly modulated by transmembrane Ca2+ movements, i.e., by ICa. These experimentally measured voltage- and time-dependent descriptors of IK strongly suggest an important functional role for IK in atrial tissue: it initiates repolarization and can be an important determinant of rate-induced changes in action potential duration.


Asunto(s)
Función Atrial , Canales Iónicos/fisiología , Potasio/metabolismo , Animales , Bufonidae , Estimulación Eléctrica , Electrofisiología , Cinética , Matemática , Microelectrodos , Factores de Tiempo
4.
Endocrinology ; 131(2): 979-81, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1379170

RESUMEN

FSH has been shown to stimulate the uptake of calcium in cultured rat Sertoli cells, resulting in an increase in cytosolic calcium concentrations. One possibility which has been put forth is that the FSH receptor per se may act as a calcium channel. This is all the more tantalizing given the proposed structure of this receptor which, like all other G protein-coupled receptors, is thought to have the putative transmembrane helices forming a bundle-like structure in the plasma membrane. To test whether the FSH receptor could function as a calcium channel, we performed whole-cell voltage clamp experiments on 293 and 293F(wt1) cells, which are a clonal line of 293 cells expressing high levels of rat FSH receptors. The 293 cells, which do not express FSH receptors, were found to lack any detectable inward calcium currents, and therefore, serve as an excellent model for transfecting with potential calcium conducting FSH receptors. When the 293F(wt1) cells were then tested, no inward calcium currents could be detected in either control or FSH-stimulated cells. These results suggest that the FSH receptor itself is not a calcium channel and, therefore, FSH must be stimulating endogenous calcium channels in rat Sertoli cell plasma membranes.


Asunto(s)
Canales de Calcio/fisiología , Receptores de HFE/fisiología , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Bucladesina/farmacología , Línea Celular Transformada , Colforsina/farmacología , AMP Cíclico/fisiología , ADN/genética , Electrofisiología , Embrión de Mamíferos , Hormona Folículo Estimulante/farmacología , Humanos , Potenciales de la Membrana , Receptores de HFE/genética , Tetraetilamonio , Compuestos de Tetraetilamonio/farmacología , Transfección
5.
J Investig Med ; 43(5): 451-8, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8528756

RESUMEN

BACKGROUND: The patch-clamp technique was used to study a large conductance, calcium-activated potassium channel (IK(Ca) in coronary arterial smooth muscle cells from rabbits. The properties of this channel are similar to those of IK(Ca) found in many types of vascular tissue. A brief single channel characterization of IK(Ca) in this tissue type has been completed for this study. METHODS: The effects of S-nitrosothiol compounds on IK(Ca) were studied in cell-attached patches. RESULTS: The probability of opening for IK(Ca) increased from 0.008 +/- 0.004 to 0.780 +/- 0.07 following application of S-nitroso-L-cysteine. S-nitroso-N-acetylpenicillamine (SNAP) also increased the probability of opening for IK(Ca) from 0.022 +/- 0.01 to 0.601 +/- 0.05. The probability of opening for IK(Ca) also increased from 0.026 +/- 0.01 to 0.809 +/- 0.02 following application of membrane-permeable analogs of cyclic guanosine monophosphate (cGMP) to the bath of cell-attached patches, suggesting that IK(Ca) in coronary artery smooth muscle cells is regulated by a cGMP-dependent mechanism. Rp-8-pCPT-cGMP, a protein kinase G inhibitor, blocked the effect of SNAP, an S-nitrosothiol compound. CONCLUSIONS: These findings suggest that one of the effects of nitrosothiol compounds is the activation of IK(Ca) through a cGMP-dependent mechanism in coronary artery smooth muscle cells.


Asunto(s)
Calcio/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , GMP Cíclico/farmacología , Cisteína/análogos & derivados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Penicilamina/análogos & derivados , Canales de Potasio/efectos de los fármacos , S-Nitrosotioles , Vasodilatadores/farmacología , Animales , Cisteína/farmacología , Técnicas In Vitro , Penicilamina/farmacología , Conejos , S-Nitroso-N-Acetilpenicilamina
6.
Am J Physiol ; 264(4 Pt 2): H1146-53, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8476092

RESUMEN

Cell-attached patches from rabbit coronary artery single smooth muscle cells contained two distinct potassium channel types, namely a large conductance calcium-activated potassium channel and a smaller voltage-activated potassium channel representing the delayed rectifier (IK). When a physiological potassium ion gradient was used, the average slope conductance of single IK channels was 7.26 pS. The time course of activation measured from ensemble averages was well fit by a single exponential raised to the power of 2 and was voltage dependent. Experiments were then performed with potassium (140 mM) on both sides of the membrane to resolve single IK channel currents during deactivation. Ensemble averages of this activity were well described by a two-component exponential, and the time constants were voltage dependent. Mean open times were significantly shorter during deactivation than during activation. Closed time distributions typically had two components. These kinetic characteristics were used in testing various state models for voltage-dependent potassium channels.


Asunto(s)
Arterias/fisiología , Vasos Coronarios/fisiología , Músculo Liso Vascular/citología , Canales de Potasio/fisiología , Animales , Arterias/citología , Arterias/ultraestructura , Vasos Coronarios/citología , Vasos Coronarios/ultraestructura , Potenciales de la Membrana/fisiología , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/ultraestructura , Canales de Potasio/ultraestructura , Conejos
7.
J Physiol ; 368: 265-92, 1985 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2416914

RESUMEN

Spontaneously active single cells have been obtained from the sinus venosus region of the bull-frog, Rana catesbeiana, using an enzymic dispersion procedure involving serial applications of trypsin, collagenase and elastase in nominally 0 Ca2+ Ringer solution. These cells have normal action potentials and fire spontaneously at a rate very similar to the intact sinus venosus. A single suction micro-electrode technique (Hamill, Marty, Neher, Sakmann & Sigworth, 1981; Hume & Giles, 1983) has been used to record the spontaneous diastolic depolarizations or pace-maker activity as well as the regenerative action potentials in these cells. This electrophysiological activity is completely insensitive to tetrodotoxin (TTX; 3 X 10(-6) M) and is very similar to that recorded from an in vitro sinus venosus preparation. The present experiments were aimed at identifying the transmembrane potassium currents, and analysing their role(s) in the development of the pace-maker potential and the repolarization of the action potential. Depolarizing voltage-clamp steps from the normal maximum diastolic potential (-75 mV) elicit a time- and voltage-dependent activation of an outward current. The reversal potential of this current in normal Ringer solution [( K+]0 2.5 mM) is near -95 mV; and it shifts by 51 mV per tenfold increase in [K+]0, which strongly suggests that this current is carried by K+. We therefore labelled it IK. The reversal potential of IK did not shift in the positive direction following very long (20 s) depolarizing clamp steps to +20 mV, indicating that 'extracellular' accumulation of [K+]0 does not produce any significant artifacts. The fully activated instantaneous current-voltage (I-V) relationship for IK is approximately linear over the range of potentials -130 to -30 mV. Thus, the ion transfer mechanism of IK may be described as a simple ohmic conductance in this range of potentials. Positive relative to -30 mV, however, the I-V exhibits significant inward rectification. A Hodgkin-Huxley analysis of the kinetics of IK, including a demonstration that the envelope of tails quantitatively matches the time course of the onset of IK during a prolonged depolarizing clamp step has been completed. The steady-state activation variable (n infinity) of IK spans the voltage range approximately -40 to +10 mV. It is well-fitted by a Boltzmann distribution function with half-activation at -20 mV. The time course of decay of IK is a single exponential. However, the activation or onset of IK shows clear sigmoidicity in the range of potentials from the activation threshold (-40 mV) to 0 mV.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Canales Iónicos/fisiología , Potasio/fisiología , Nodo Sinoatrial/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Técnicas In Vitro , Cinética , Microscopía Electrónica , Miocardio/ultraestructura , Rana catesbeiana , Nodo Sinoatrial/citología , Tetrodotoxina/farmacología
8.
Proc Natl Acad Sci U S A ; 82(22): 7796-800, 1985 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3877934

RESUMEN

An enzymatic dispersion procedure has been developed to obtain viable, spontaneously active single myocytes from cardiac pacemaker tissue: the bullfrog (Rana catesbeiana) sinus venosus. Recordings of time- and voltage-dependent Ca2+ and K+ currents have been made by using a single suction-microelectrode technique. The results show that two time- and voltage-dependent currents interact to modulate the slope of the pacemaker potential. These are: (i) the decay of a delayed rectifier K+ current and (ii) the activation of a Ca2+ current. In addition, the data strongly suggest that cardiac pacemaker tissue does not have an inwardly rectifying background K+ current.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Animales , Calcio/metabolismo , Diástole , Técnicas In Vitro , Potenciales de la Membrana , Potasio/metabolismo , Rana catesbeiana
9.
Circ Res ; 72(3): 517-25, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8431982

RESUMEN

We have recently shown that beta-adrenergic agonists enhance the cardiac sodium current (INa) in rabbits through dual G-protein regulatory pathways. To determine if muscarinic cholinergic receptor stimulation can also modulate INa, we studied the effects of acetylcholine (ACh) and carbachol on INa in enzymatically dispersed rabbit ventricular myocytes. Whole-cell patch-clamp experiments done at room temperature using 20 mM [Na+]o showed that 100 nM isoproterenol increased INa and accelerated current decay as previously described. ACh (1 microM) or carbachol (1 microM) significantly reversed the stimulatory isoproterenol effects at test potentials throughout the INa activation range and at holding potentials negative to -80 mV. This effect was completely inhibited by atropine (1 microM) and was confirmed by studying single-channel INa from cell-attached patches. When INa was stimulated by forskolin (1 microM), carbachol (1 microM) significantly reversed the effect. The muscarinic-mediated inhibition of INa was inhibited by pertussis toxin (0.1 or 1.0 microgram/ml) incubation (12-15 hours), suggesting that the effect was inhibitory G-protein dependent. Further investigation of the ACh inhibitory mechanism revealed that ACh alone had no effect on INa and that when cells were dialyzed with cAMP (5 microM), ACh failed to inhibit INa. Furthermore, cGMP failed to inhibit the effect of isoproterenol on INa. These data suggest that ACh acts at or proximal to adenylate cyclase stimulation. Thus, rabbit cardiac Na+ channels are regulated by muscarinic agonists in a fashion similar to cardiac Ca2+ channels.


Asunto(s)
Acetilcolina/farmacología , Corazón/efectos de los fármacos , Isoproterenol/antagonistas & inhibidores , Receptores Muscarínicos/fisiología , Sodio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Carbacol/farmacología , Proteínas de Unión al GTP , Potenciales de la Membrana/efectos de los fármacos , Conejos , Receptores Muscarínicos/efectos de los fármacos
10.
J Physiol ; 439: 751-68, 1991 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1910087

RESUMEN

1. Voltage- and time-dependent outward currents were recorded from relaxed enzymatically isolated smooth muscle cells from the rabbit left descending coronary artery using a single pipette voltage clamp technique. The calcium-activated potassium current was blocked by inclusion of EGTA in the pipette solution and CdCl2 in the extracellular bath. 2. Outward currents were elicited with depolarizing voltage steps to potentials positive to -20 mV. Long (5 s) voltage steps revealed slow inactivation of the current with a time constant of nearly 3 s at +60 mV. Potassium was identified as the predominant charge carrier by reversal potential measurements in potassium substitution experiments. 3. The results of kinetic analyses compared favourably with the Hodgkin-Huxley model for a delayed rectifier with some deviations. The sigmoid current onset was best fitted by raising the activation variable (n) to the second power. Deactivation tail currents were consistently found to be comprised of two exponential components. The kinetics of activation and deactivation were strongly voltage-dependent from -80 to +60 mV. 4. Envelope of tails experiments showed that the scaled tail current amplitudes followed the kinetic behaviour of current activation. The contribution of each of the two exponential tail components was also measured in these experiments. They did not reveal kinetically separable currents, nor were they differentially altered by 4-aminopyridine (4-AP), tetraethylammonium (TEA), or elevated [K+]o. 5. The steady-state voltage-dependence curves for both activation and inactivation were well fitted by a Boltzmann distribution with V1/2 = -5.60 mV and k = -8.66 mV for n infinity act and V1/2 = -24.20 mV and k = 5.16 mV for n infinity act. Super-imposition of the two curves revealed a 'window' of voltage where channels are available for activation without completely inactivating. 6. Neither of the commonly used potassium channel blockers, TEA or 4-AP, were particularly effective blockers of IK, reducing current by only 50-70% at an extracellular concentration of 10 mM. TEA block was mildly voltage-dependent and was more effective in reducing current towards the end of a 500 ms depolarization. 4-AP, on the other hand, demonstrated considerable voltage-dependence and preferentially reduced early currents. 7. Outward currents recorded from guinea-pig and human coronary artery myocytes under the same conditions as in the rabbit cell experiments displayed similar characteristics.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Músculo Liso Vascular/fisiología , Canales de Potasio/fisiología , 4-Aminopiridina/farmacología , Animales , Calcio/fisiología , Vasos Coronarios , Ácido Egtácico/farmacología , Cinética , Potenciales de la Membrana/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Conejos , Tetraetilamonio , Compuestos de Tetraetilamonio/farmacología
11.
Circ Res ; 70(1): 199-207, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1309315

RESUMEN

Voltage-dependent sodium channels from a variety of tissues are known to be phosphorylated by the cAMP-dependent protein kinase, protein kinase A. However, the functional significance of sodium channel phosphorylation is not clearly understood. Using whole-cell voltage-clamp techniques, we show that sodium currents (INas) in rabbit cardiac myocytes are enhanced by isoproterenol (ISO). This enhancement of INa by ISO 1) is holding potential dependent, 2) can be mimicked by forskolin and dibutyryl cAMP, and 3) is accompanied by an increase in the rate of Na+ channel inactivation. In single-channel, inside-out patch experiments, the catalytic subunit of protein kinase A also enhances INa and increases the rate of inactivation, suggesting that cardiac Na+ channel phosphorylation may be physiologically important. Addition of the protein kinase A inhibitor to the pipette solution in whole-cell experiments blocks the stimulatory effect of forskolin without blocking the effect of ISO, suggesting that ISO also enhances INa through a cAMP-independent pathway. To determine if ISO may stimulate INa through a direct G protein pathway, single channels were recorded in the presence of the Gs-activating GTP analogue, GTP gamma S, and the stimulatory G protein subunit, Gs alpha. Both of these agents enhanced INa without affecting the rate of Na+ channel inactivation. These results suggest that ISO enhances rabbit cardiac INa through a dual (direct and indirect) G protein regulatory pathway.


Asunto(s)
Corazón/efectos de los fármacos , Isoproterenol/farmacología , Miocardio/metabolismo , Canales de Sodio/efectos de los fármacos , Animales , Bucladesina/farmacología , Colforsina/farmacología , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Técnicas In Vitro , Miocardio/citología , Fosforilación , Conejos , Canales de Sodio/metabolismo , Estimulación Química
12.
Circ Res ; 75(4): 742-50, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7923619

RESUMEN

Whole-cell patch-clamp techniques were used to study enzymatically dispersed epicardial coronary artery smooth muscle cells. Depolarizing voltage pulses of 500-millisecond duration from -60 mV (118 mmol/L CsCl, 22 mmol/L tetraethylammonium chloride, and 5 mmol/L EGTA pipette solution) elicited inward L-type calcium currents (ICa). When EGTA was omitted from the pipette solution, an outward current was superimposed on the calcium current, and repolarizing voltage steps produced an inward tail current (IT). The amplitude of these inward currents was proportional to the ICa amplitude from -30 to +50 mV. The time course of decay of the current was well fit by a single exponential equation. The time constant (tau) of this equation did not change with the size of IT but was clearly voltage dependent (shorter at more negative potentials). Changing the chloride reversal potential from -1.3 to -39.7 mV by anion substitution using methanesulfonate as the chloride replacement in the pipette solution shifted the zero current level of IT from 0.9 +/- 0.56 to -33.1 +/- 0.85 mV. The tail current was blocked by nifedipine (10(-6) mol/L) and by isosmolar calcium substitution with barium in the bath solution and was enhanced by the dihydropyridine agonist Bay K 8644 (10(-6) mol/L). IT was also blocked by the chloride channel blockers DIDS (10(-4) mol/L) and niflumic acid (10(-5) mol/L). Caffeine (10(-2) mol/L), which releases intracellular calcium stores, caused an inward current at holding potentials (-60 mV), which was inhibited by DIDS. Caffeine also inhibited subsequent attempts to elicit IT by depolarizing pulses (88% reduction in IT).(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Calcio/fisiología , Cloruros/fisiología , Músculo Liso Vascular/citología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Canales de Calcio/fisiología , Canales de Cloruro/metabolismo , Canales de Cloruro/fisiología , Cloruros/metabolismo , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Conductividad Eléctrica , Estimulación Eléctrica , Humanos , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Modelos Biológicos , Conejos
13.
Proc R Soc Lond B Biol Sci ; 223(1232): 355-78, 1985 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-2858102

RESUMEN

Leading or primary pacemaker cells located within the rabbit sino-atrial node have been identified by using electrophysiological and pharmacological techniques. Stable intracellular recordings lasting 20-30 min from cells within the s.a. node reveal three distinct patterns of spontaneous intracellular responses: (i) leading or primary pacing; (ii) follower or subsidiary pacing; and (iii) 'anomalous' pacemaker discharge. Our main objective was to measure the first detectable effect, or effects, of acetylcholine on the spontaneous intracellular electrical activity in mammalian primary pacemaker cells. Trains of brief 'field' stimuli were applied to evoke transmitter release from endogenous nerve varicosities. Systematic variations in the amplitude and duration of each stimulus, and in the train length; in conjunction with application of beta blockers (l-pindolol (10(-6) M); l-propranolol, (2 X 10(-7) M)) yielded small and transient, but very consistent negative chronotropic effects. These electrophysiological changes were blocked by atropine (1 X 10(-7) M) and were mimicked by bath application of low doses of acetylcholine (10(-7)-10(-6) M) or muscarine chloride (10(-8)-10(-7) M). In primary cells the first, or threshold effect of vagal excitation is a decrease in the slope of the pacemaker potential, without a detectable (less than 2 m V) hyperpolarization or change in action potential duration. A reduction in the dV/dtmax of the initial depolarization is also quite consistently observed. Application of longer stimulus trains yield the classical hyperpolarizing response, which is often assumed to be the major electrophysiological correlate of the negative chronotropic effect. These data provide a detailed electrophysiological description of the 'physiological' effects of the vagus nerve excitation on primary or leading pacemaker cells of the mammalian s.-a. node. A plausible explanation for the absence of hyperpolarization is suggested; and a working hypothesis is presented for the changes in ionic current or currents, that underlie this negative chronotropic effect.


Asunto(s)
Acetilcolina/farmacología , Nodo Sinoatrial/efectos de los fármacos , Animales , Electrofisiología , Femenino , Masculino , Conejos , Nodo Sinoatrial/citología , Nodo Sinoatrial/fisiología , Nervio Vago/fisiología
14.
Fed Proc ; 42(13): 2994-7, 1983 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6311634

RESUMEN

A transmembrane flux of Ca2+ has been demonstrated in many nerve and muscle cells. In cardiac muscle, Ca2+ channels in the sarcolemma transfer sufficient Ca2+ to trigger and partially control tension development. This time- and voltage-dependent Ca2+ current is also important in the development of the pacemaker potential, or diastolic depolarization. In addition, transmitter release from autonomic nerve varicosities in the myocardium exhibits a strong dependence on external calcium concentration [( Ca2+]o). Agents that selectively alter either pre- or postsynaptic Ca2+ channels are therefore of considerable interest. Our results illustrate two distinct effects of Cd2+ in cardiac muscle. Data from conventional electrophysiological recordings from primary pacemaker cells within the rabbit sinoatrial node indicate that Cd2+ (10(-6)-10(-5) M) may selectively inhibit acetylcholine release. Voltage clamp measurements of transmembrane Ca2+ currents in single isolated bullfrog atrial cells show that Cd2+ (10(-4)-10(-3) M) is also a very potent inhibitor of postsynaptic Ca2+ channels; these effects of Cd2+ mimic those seen after [Ca2+]o removal.


Asunto(s)
Cadmio/toxicidad , Corazón/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Cloruro de Cadmio , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Atrios Cardíacos/efectos de los fármacos , Técnicas In Vitro , Canales Iónicos/efectos de los fármacos , Miocardio/metabolismo , Conejos , Rana catesbeiana , Nodo Sinoatrial/efectos de los fármacos
15.
J Physiol ; 415: 233-49, 1989 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2640462

RESUMEN

1. The effects of isoprenaline (ISO) on the calcium current (ICa) and delayed rectifier K+ current (IK) were examined using a tight-seal whole-cell voltage-clamp technique in single cells from bull-frog atrium to examine the ionic mechanism(s) of catecholamine-induced action potential shape changes. 2. The effects of ISO on the action potential were dose-dependent. Very low doses (5 x 10(-9) M) prolonged the action potential. Higher doses (10(-6) M) of ISO increased the plateau height, but shortened the action potential by accelerating the early repolarization phase. 3. ISO increased IK and ICa in a dose-dependent fashion. Both of these effects were blocked by a beta-receptor antagonist, propranolol (3 x 10(-7) M). In contrast IK1, the inwardly rectifying K+ current, was not changed significantly by ISO. 4. The ISO-induced increase in IK was observed in the presence of CdCl2 (3 x 10(-4) M), indicating that this effect is not due to a Ca2(+)-activated potassium current. 5. The reversal potential of IK in normal Ringer solution (-83 +/- 2 mV) was not significantly changed by ISO. Thus, stimulation of the Na(+)-K+ pump and a consequent hyperpolarizing shift in EK are not responsible for the increase in IK. 6. In the presence of ISO (10(-6) M) the steady-state activation curve (n infinity) for IK was consistently shifted to more negative values (by approximately 10 mV). The activation and deactivation kinetics of IK were also changed by ISO: activation was accelerated, deactivation was slowed. These ISO-induced changes in IK result in an increase in IK at voltages corresponding to the plateau of the action potential. 7. ISO (10(-6) M) increased ICa dramatically, approximately 6-fold at 0 mV. At the same time, the time constant of ICa inactivation decreased significantly (34 +/- 4 ms control; 23 +/- 4 ms ISO). 8. These results confirm that low doses of sympathetic agonists acting via beta-receptors increase ICa. Relatively high doses of beta-receptor agonists increase both ICa and IK, but these two effects appear to be generated by different biophysical mechanisms. 9. These dose-dependent changes in ICa and IK can explain the observed ISO-induced changes in action potential shape. At doses of approximately 10(-8) M ICa is increased, resulting in a more depolarized plateau and small lengthening of the action potential.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Corazón/fisiología , Isoproterenol/farmacología , Canales de Potasio/fisiología , Potasio/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Función Atrial , Calcio/fisiología , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Cinética , Rana catesbeiana
16.
J Physiol ; 427: 657-80, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1698983

RESUMEN

1. Calcium inward currents were recorded from relaxed enzymatically isolated smooth muscle cells from the rabbit epicardial left descending coronary artery using a single-pipette voltage-clamp technique. Outward K+ currents were blocked with CsCl-tetraethylammonium-filled pipette solutions. 2. Relaxed coronary smooth muscle cells had a maximum diameter of 8.6 +/- 0.6 microns and a cell length of 96.7 +/- 3.3 microns when bathed in 2.5 mM [Ca2+]o. The average resting membrane potential at room temperature was -32 +/- 10 mV. The mean cell capacitance was 18.5 +/- 1.7 pF and the input resistance was 3.79 +/- 0.58 G omega. 3. Depolarizing voltage-clamp steps from a holding potential of -80 mV elicited a single time- and voltage-dependent inward current which was dependent upon extracellular [Ca2+]. In 2.5 mM [Ca2+]o, the inward current was activated at a potential of -40 mV and peaked at +10 mV. This current was inhibited by 0.5 mM-CdCl2 and 1 microM-nifedipine and was enhanced with 1 microM-Bay K 8644. No detectable low-threshold, rapidly inactivating T-type calcium current was observed. 4. The apparent reversal potential of this inward current in 2.5 mM [Ca2+]o was +70 mV and shifted by 33.0 mV per tenfold increase in [Ca2+]o. This channel was also more permeable to barium and strontium ions than to calcium ions. 5. Single calcium channel recordings with 110 mM-Ba2+ as the charge carrier revealed a mean slope conductance of 20.7 +/- 0.8 pS. 6. This calcium current (ICa) exhibited a strong voltage-dependent inactivation process. However, the steady-state inactivation curve (f infinity) displayed a slight nonmonotonic, U-shaped dependence upon membrane potential. The potential at which half of the channels were inactivated was -27.9 mV with a slope factor of 6.9 mV. The steady-state activation curve (d infinity) was also well-described by a Boltzmann distribution with a half-activation potential at -4.4 mV and a slope factor of -63 mV. ICa was fully activated at approximately +20 mV. 7. The rate of inactivation was dependent upon the species of ion carrying the current. Both Sr2+ and Ba2+ decreased the rate as well as the degree of inactivation. The tau f (fitted time constant of inactivation) curve displayed a U-shaped relationship in 2.5 mM [Ca2+]o. The reactivation process was voltage dependent and could be described by a single exponential. 8. The current amplitude and the inactivation kinetics were temperature dependent.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Canales de Calcio/fisiología , Músculo Liso Vascular/fisiología , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Animales , Bario/fisiología , Canales de Calcio/efectos de los fármacos , Separación Celular , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Electrofisiología , Calor , Potenciales de la Membrana/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Níquel/farmacología , Conejos
17.
J Physiol ; 403: 239-66, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2855341

RESUMEN

1. Voltage clamp studies on single cells from bull-frog atrium have been carried out to study the ion transfer characteristics of the calcium current, ICa. In agreement with the preliminary results of Hume & Giles (1983), a TTX-resistant, 'second transient inward current' was recorded consistently. Its average peak size at 0 mV in 2.5 mM [Ca2+]o Ringer solution was approximately -200 pA, and it was blocked by Cd2+ and La3+ but not by tetrodotoxin (TTX, 3 x 10(-6) M). 2. The peak size of this current increases by approximately 4 times when [Ca2+]o is raised from 1.25 to 7.5 mM, indicating that Ca2+ is a major charge carrier. 3. A well-defined reversal potential, Erev, for ICa can be recorded in normal Ringer solution and also when Ba2+ or Sr2+ serve as the charge carriers. When [Ca2+]o is changed the shifts in Erev follow the predictions of a Nernstian Ca2+ electrode. However, all Erev values are well below those predicted from the thermodynamic Nernstian ECa values (see Campbell, Giles, Hume, Noble & Shibata, 1988a). 4. The Ca2+ current exhibits voltage-dependent inactivation, whether the direction of net current flow is inward or outward; however, the rate of inactivation is affected by the species of cation carrying the current. Inactivation is reduced substantially in Ba2+ Ringer solution. 5. Magnesium (5 mM) is not a significant carrier or blocker of ICa in normal [Ca2+]o Ringer solution; however, 5 mM [Mg2+]o can block the current carried by either Sr2+ or Ba2+. In the absence of Mg2+, equimolar substitutions of Sr2+ or Ba2+ for Ca2+ result in larger currents than those carried by Ca2+ in the normal Ringer solution. 6. Sodium appears not to be a significant charge carrier in the presence of normal [Ca2+]o. However, after free [Ca2+]o has been reduced to extremely low levels (less than 10(-6) M) Na+ can carry a significant fraction of 'ICa'. Thus, it appears that the high selectivity of ICa for Ca2+ ions depends upon the presence of Ca2+. 7. 'Slow tails' are frequently recorded after repolarizing clamp steps back to the holding potential. These 'slow tails' are prominent in normal [Na+]o, [Ca2+]o and [Sr2+]o Ringer solution; however, they are markedly reduced in [Ba2+]o, in Na+-free and Ca2+-free Ringer solutions. Experimental and theoretical work suggests these slow tails may be generated by an electrogenic Na+-Ca2+ exchanger (see Campbell, Giles, Robinson & Shibata, 1988b).(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Canales de Calcio/fisiología , Calcio/fisiología , Corazón/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Cationes Bivalentes/farmacología , Técnicas In Vitro , Lantano/farmacología , Magnesio/farmacología , Rana catesbeiana , Sodio/farmacología , Tetrodotoxina/farmacología
18.
J Membr Biol ; 89(2): 131-8, 1986.
Artículo en Inglés | MEDLINE | ID: mdl-3486295

RESUMEN

Single electrode whole cell voltage-clamp experiments and frequency domain analyses have been used to study and compare the K+ currents in enzymatically dispersed single cells from the atrium and the sinus venosus (pacemaker region) of the bullfrog heart. Admittance measurements made near the "resting' or zero-current potential yield data from which the equivalent circuit of each cell type may be obtained. Data from both atrial and pacemaker cells are well-fitted by a model consisting only of parallel resistance-capacitative elements, as predicted from their micro-anatomy. Neither of these amphibian cardiac cells contain a transverse tubule system (TT) and both have very little sarcoplasmic reticulum (SR). These results complement and extend two earlier investigations: (i) Moore, Schmid and Isenberg (J. Membrane Biol. 81:29-40, 1984) have reported that in guinea pig ventricle cells (which do contain an internal membrane system consisting of transverse tubules and a substantial SR) the SR may be electrically coupled to the sarcolemma; (ii) Shibata and Giles (Biophys. J. 45:136a, 1984) have shown that although bullfrog atrial cells have an inwardly rectifying background K+ current, IK1, pacemaker cells from the immediately adjacent sinus venosus do not. Data from admittance measurements also provide evidence that a TTX-insensitive inward Ca2+ current is activated in the pacemaker range of potentials.


Asunto(s)
Corazón/fisiología , Potenciales de Acción , Animales , Función Atrial , Bario/farmacología , Estimulación Eléctrica , Atrios Cardíacos/efectos de los fármacos , Técnicas In Vitro , Potenciales de la Membrana , Microelectrodos , Rana catesbeiana
19.
J Physiol ; 518 ( Pt 2): 371-84, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10381586

RESUMEN

1. Modulation of cardiac sodium currents (INa) by the G protein stimulatory alpha subunit (Gsalpha) was studied using patch-clamp techniques on freshly dissociated rat ventricular myocytes. 2. Whole-cell recordings showed that stimulation of beta-adrenergic receptors with 10 microM isoprenaline (isoproterenol, ISO) enhanced INa by 68.4 +/- 9.6 % (mean +/- s.e.m.; n = 7, P < 0.05 vs. baseline). With the addition of 22 microgram ml-1 protein kinase A inhibitor (PKI) to the pipette solution, 10 microM ISO enhanced INa by 30.5 +/- 7.0 % (n = 7, P < 0.05 vs. baseline). With the pipette solution containing both PKI and 20 microgram ml-1 anti-Gsalpha IgG or 20 microgram ml-1 anti-Gsalpha IgG alone, 10 microM ISO produced no change in INa. 3. The effect of Gsalpha on INa was not due to changes in the steady-state activation or inactivation curves, the time course of current decay, the development of inactivation, or the recovery from inactivation. 4. Whole-cell INa was increased by 45.2 +/- 5.3% (n = 13, P < 0.05 vs. control) with pipette solution containing 1 microM Gsalpha27-42 peptide (amino acids 27-42 of rat brain Gsalpha) without altering the properties of Na+ channel kinetics. Furthermore, application of 1 nM Gsalpha27-42 to Na+ channels in inside-out macropatches increased the ensemble-averaged INa by 32.5 +/- 6.8 % (n = 8, P < 0.05 vs. baseline). The increase in INa was reversible upon Gsalpha27-42 peptide washout. Single channel experiments showed that the Gsalpha27-42 peptide did not alter the Na+ single channel current amplitude, the mean open time or the mean closed time, but increased the number of functional channels (N) in the patch. 5. Application of selected short amino acid segments (Gsalpha27-36, Gsalpha33-42 and Gsalpha30-39) of the 16 amino acid Gsalpha peptide (Gsalpha27-42 peptide) showed that only the C-terminal segment of this peptide (Gsalpha33-42) significantly increased INa in a dose-dependent fashion. These results show that cardiac INa is regulated by Gsalpha via a mechanism independent of PKA that results in an increase in the number of functional Na+ channels. In addition, a 10 residue domain (amino acids 33-42) near the N-terminus of Gsalpha is important in modulating cardiac Na+ channels.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go , Proteínas de Unión al GTP/metabolismo , Corazón/efectos de los fármacos , Miocardio/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Canales de Sodio/metabolismo , Agonistas Adrenérgicos beta/farmacología , Algoritmos , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Electrofisiología , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Isoproterenol/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Ratas , Canales de Sodio/efectos de los fármacos
20.
Am J Physiol ; 257(6 Pt 2): H1773-81, 1989 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2557769

RESUMEN

Conventional microelectrode recordings combined with enzymatic cell dispersion methods and a single microelectrode voltage-clamp technique were used to record transmembrane action potentials and ionic currents in isolated single myocytes and in excised segments of human right atrium. Recordings of the outward current(s), which is responsible for the resting potential and early repolarization of the action potential in human right atrium, consistently showed that this tissue has 1) a relatively small inwardly rectifying background potassium current (IK1) which generates the resting potential in mammalian ventricular tissue and Purkinje fibers, and 2) a large time- and voltage-dependent, but Ca2(+)-independent, transient outward current. A somewhat similar K+ current was originally described in neurons and recently has also been identified in a variety of mammalian cardiac tissues. As expected from previous work, this transient outward current in human atrium is blocked by 4-aminopyridine (4-AP; 0.5 mM) and exhibits time- and voltage-dependent inactivation and reactivation. Measurements of action potential shape changes and phasic tension as a function of stimulus frequency, or after 4-AP application, show that in human atrium this current can produce pronounced changes in both the early repolarization of the action potential and force generation.


Asunto(s)
Corazón/fisiología , 4-Aminopiridina/farmacología , Potenciales de Acción/efectos de los fármacos , Anciano , Función Atrial , Cadmio/farmacología , Cloruro de Cadmio , Conductividad Eléctrica , Electrofisiología/métodos , Atrios Cardíacos/efectos de los fármacos , Humanos , Técnicas In Vitro , Cinética , Potenciales de la Membrana , Persona de Mediana Edad , Tetrodotoxina/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda