Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Basic Res Cardiol ; 110(2): 5, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25600224

RESUMEN

The sustained component of the K(+) outward current in human atrial myocytes is believed to be due to the slowly inactivating ultra-rapid potassium current I Kur and not to the fast inactivating transient outward current Ito. Here we provide evidence for contribution of Ito to this late current due to the effects of dipeptidyl peptidase-like protein (DPP) 10 (DPP10a) interacting with Kv4.3 channels. We studied the late current component of Ito in human atrial myocytes and CHO cells co-expressing Kv4.3 or Kv4.3/KChIP2 (control) and DPP proteins using voltage-clamp technique and a pharmacological approach. A voltage dependent and slowly inactivating late current (43% of peak amplitude) could be observed in atrial myocytes. We found a similar current in CHO cells expressing Kv4.3/KChIP2 + DPP10a, but not in cells co-expressing Kv4.3 + DPP or Kv4.3/KChIP2 + DPP6-S. Assuming that DPP10a influences atrial Ito, we detected DPP10 expression of three alternatively spliced mRNAs, DPP10 protein and colocalization of Kv4.3 and DPP10 proteins in human atrial myocytes. DPP10a did not affect properties of expressed Kv1.5 excluding a contribution to the sustained IKur in atrial cells. To test for the contribution of Kv4-based Ito on sustained K(+) outward currents in human atrial myocytes, we used 4-AP to block IKur, in combination with Heteropoda toxin 2 to block Kv4 channels. We could clearly separate an Ito fraction of about 19% contributing to the late current in atrial myocytes. Thus, the interaction of DPP10a, expressed in human atrium, with Kv4.3 channels generates a sustained current component of Ito, which may affect late repolarization phase of atrial action potentials.


Asunto(s)
Potenciales de Acción/fisiología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Células CHO , Cricetulus , Femenino , Técnica del Anticuerpo Fluorescente , Atrios Cardíacos/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
2.
J Mol Cell Cardiol ; 56: 8-18, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23291429

RESUMEN

In human atrial myocytes the transient outward current I(to) develops a conspicuous faster inactivation with increasing temperatures. Since ß-subunits are known to modulate I(to) current kinetics, we hypothesized that the temperature sensitivity of I(to) is not only determined by the property of the ion-passing α-subunit Kv4.3 but also by its interaction with accessory ß-subunits. We therefore studied the influence of the transmembrane ß-subunits KCNE1, KCNE2 and DPP6 on Kv4.3/KChIP2 channels in CHO cells at room temperature and at physiological temperature. Exposure to 37°C caused a significant acceleration of the channel kinetics, whereas current densities and voltage dependences remained unaltered at 37°C compared to 23°C. However, Kv4.3/KChIP2 channels without transmembrane ß-subunits showed the strongest temperature sensitivity with considerably increased rates of activation and inactivation at 37°C. KCNE2 significantly slowed the current kinetics at 37°C compared to Kv4.3/KChIP2 channels, whereas KCNE1 did not influence the channel properties at both temperatures. Interestingly, the accelerating effects of DPP6 on current kinetics described at 23°C were diminished at physiological temperature, thus at 37°C current kinetics became remarkably similar for channel complexes Kv4.3/KChIP2 with and without DPP6 isoforms. A Markov state model was developed on the basis of experimental measurements to simulate the influence of ß-subunits on Kv4.3 channel complex at both temperatures. In conclusion, the remarkably fast kinetics of the native I(to) at 37°C could be reproduced by co-expressing Kv4.3, KChIP2, KCNE2 and DPP6 in CHO cells, whereas the high temperature sensitivity of human I(to) could be not mimicked.


Asunto(s)
Subunidades de Proteína/fisiología , Canales de Potasio Shal/metabolismo , Potenciales de Acción , Animales , Células CHO , Cricetinae , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Humanos , Activación del Canal Iónico , Cinética , Cadenas de Markov , Modelos Biológicos , Proteínas del Tejido Nervioso/fisiología , Técnicas de Placa-Clamp , Canales de Potasio/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Estabilidad Proteica , Termodinámica
3.
Br J Pharmacol ; 154(4): 774-86, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18536731

RESUMEN

BACKGROUND AND PURPOSE: The human cardiac transient outward potassium current (Ito) is believed to be composed of the pore-forming Kv4.3 alpha-subunit, coassembled with modulatory beta-subunits as KChIP2, MiRP1 and DPP6 proteins. beta-Subunits can alter the pharmacological response of Ito; therefore, we analysed the effects of flecainide on Kv4.3/KChIP2 channels coassembled with MiRP1 and/or DPP6 beta-subunits. EXPERIMENTAL APPROACH: Currents were recorded in Chinese hamster ovary cells stably expressing K(V)4.3/KChIP2 channels, and transiently transfected with either MiRP1, DPP6 or both, using the whole-cell patch-clamp technique. KEY RESULTS: In control conditions, Kv4.3/KChIP2/MiRP1 channels exhibited the slowest activation and inactivation kinetics and showed an 'overshoot' in the time course of recovery from inactivation. The midpoint values (Vh) of the activation and inactivation curves for Kv4.3/KChIP2/DPP6 and Kv4.3/KChIP2/MiRP1/DPP6 channels were approximately 10 mV more negative than Vh values for Kv4.3/KChIP2 and Kv4.3/KChIP2/MiRP1 channels. Flecainide (0.1-100 microM) produced a similar concentration-dependent blockade of total integrated current flow (IC50 approximately 10 microM) in all the channel complexes. However, the IC50 values for peak current amplitude and inactivated channel block were significantly different. Flecainide shifted the Vh values of both the activation and inactivation curves to more negative potentials and apparently accelerated inactivation kinetics in all channels. Moreover, flecainide slowed recovery from inactivation in all the channel complexes and suppressed the 'overshoot' in Kv4.3/KChIP2/MiRP1 channels. CONCLUSIONS AND IMPLICATIONS: Flecainide directly binds to the Kv4.3 alpha-subunit when the channels are in the open and inactivated state and the presence of the beta-subunits modulates the blockade by altering the gating function.


Asunto(s)
Antiarrítmicos/farmacología , Flecainida/farmacología , Subunidades de Proteína , Canales de Potasio Shal/efectos de los fármacos , Animales , Antiarrítmicos/administración & dosificación , Células CHO , Cricetinae , Cricetulus , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Relación Dosis-Respuesta a Droga , Femenino , Flecainida/administración & dosificación , Humanos , Concentración 50 Inhibidora , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Péptido Hidrolasas/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio Shal/metabolismo , Transfección
4.
Br J Pharmacol ; 154(8): 1619-30, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18536759

RESUMEN

BACKGROUND AND PURPOSE: This study was designed to establish the pathology-specific inhibitory effects of the IKur/Ito/IK,ACh blocker AVE0118 on atrium-selective channels and its corresponding effects on action potential shape and effective refractory period in patients with chronic AF (cAF). EXPERIMENTAL APPROACH: Outward K+-currents of right atrial myocytes and action potentials of atrial trabeculae were measured with whole-cell voltage clamp and microelectrode techniques, respectively. Outward currents were dissected by curve fitting. KEY RESULTS: Four components of outward K+-currents and AF-specific alterations in their properties were identified. Ito was smaller in cAF than in SR, and AVE0118 (10 microM) apparently accelerated its inactivation in both groups without reducing its amplitude. Amplitudes of rapidly and slowly inactivating components of IKur were lower in cAF than in SR. The former was abolished by AVE0118 in both groups, the latter was partially blocked in SR, but not in cAF, even though its inactivation was apparently accelerated in cAF. The large non-inactivating current component was similar in magnitude in both groups, but decreased by AVE0118 only in SR. AVE0118 strongly suppressed AF-related constitutively active IK,ACh and prolonged atrial action potential and effective refractory period exclusively in cAF. CONCLUSIONS AND IMPLICATIONS: In atrial myocytes of cAF patients, we detected reduced function of distinct IKur components that possessed decreased component-specific sensitivity to AVE0118 most likely as a consequence of AF-induced electrical remodelling. Inhibition of profibrillatory constitutively active IK,ACh may lead to pathology-specific efficacy of AVE0118 that is likely to contribute to its ability to convert AF into SR.


Asunto(s)
Fibrilación Atrial/tratamiento farmacológico , Compuestos de Bifenilo/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Anciano , Fibrilación Atrial/fisiopatología , Enfermedad Crónica , Electrofisiología , Femenino , Atrios Cardíacos/citología , Atrios Cardíacos/patología , Humanos , Técnicas In Vitro , Masculino , Microelectrodos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo
5.
Br J Pharmacol ; 152(6): 835-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17876301

RESUMEN

The cardiac ultrarapid outward current I(Kur), encoded by KCNA5, is of special pharmacological interest, because it is considered to be atrium-specific. I(Kur) has therefore become a target in the therapy of atrial tachyarrhythmias. However, the concept of atrium specificity is only valid if a functional I(Kur) current is in fact absent from the ventricle. However, new work has detected a I(Kur)-like current in canine ventricular myocytes, sensitive to 4-aminopyridine and suppressed by the I(Kur) blocker DPO-1, findings that support the existence of a functional ventricular I(Kur). These indications are, however, indirect and more effort is needed to clarify unequivocally the putative role of an expectedly small I(Kur) component in the ventricle.


Asunto(s)
Corazón/efectos de los fármacos , Corazón/fisiología , Canal de Potasio Kv1.5/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Perros , Ventrículos Cardíacos/efectos de los fármacos , Canal de Potasio Kv1.5/genética , Especificidad de Órganos , Función Ventricular
6.
Herzschrittmacherther Elektrophysiol ; 17(2): 64-72, 2006 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-16786464

RESUMEN

During the last ten years we have made substantial progress in our understanding of the underlying mechanisms of atrial fibrillation. The high rate associated alterations in electrical and structural properties of the atria, referred to as atrial remodeling, promote the progression of atrial fibrillation. The development of new therapeutic approaches addresses three different directions: (i) prevention of atrial remodeling, especially of structural remodeling; (ii) increase of long-term efficacy of currently used drugs and improvement of their side-effect profile; and (iii) design of atria- and pathology-specific antiarrhythmic drugs without concomitant proarrhythmic effects in the ventricles. The current review outlines the pathophysiology of atrial fibrillation and focuses on electrical remodeling. The properties of new antiarrhythmic drugs for atrial fibrillation are discussed in detail.


Asunto(s)
Antiarrítmicos/administración & dosificación , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/fisiopatología , Bloqueadores de los Canales de Calcio/administración & dosificación , Canales de Calcio/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Animales , Sistema de Conducción Cardíaco/efectos de los fármacos , Humanos , Activación del Canal Iónico/efectos de los fármacos , Modelos Cardiovasculares , Guías de Práctica Clínica como Asunto , Pautas de la Práctica en Medicina , Resultado del Tratamiento
7.
Circulation ; 112(24): 3697-706, 2005 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-16330682

RESUMEN

BACKGROUND: The molecular mechanism of increased background inward rectifier current (IK1) in atrial fibrillation (AF) is not fully understood. We tested whether constitutively active acetylcholine (ACh)-activated I(K,ACh) contributes to enhanced basal conductance in chronic AF (cAF). METHODS AND RESULTS: Whole-cell and single-channel currents were measured with standard voltage-clamp techniques in atrial myocytes from patients with sinus rhythm (SR) and cAF. The selective I(K,ACh) blocker tertiapin was used for inhibition of I(K,ACh). Whole-cell basal current was larger in cAF than in SR, whereas carbachol (CCh)-activated I(K,ACh) was lower in cAF than in SR. Tertiapin (0.1 to 100 nmol/L) reduced I(K,ACh) in a concentration-dependent manner with greater potency in cAF than in SR (-logIC50: 9.1 versus 8.2; P<0.05). Basal current contained a tertiapin-sensitive component that was larger in cAF than in SR (tertiapin [10 nmol/L]-sensitive current at -100 mV: cAF, -6.7+/-1.2 pA/pF, n=16/5 [myocytes/patients] versus SR, -1.7+/-0.5 pA/pF, n=24/8), suggesting contribution of constitutively active I(K,ACh) to basal current. In single-channel recordings, constitutively active I(K,ACh) was prominent in cAF but not in SR (channel open probability: cAF, 5.4+/-0.7%, n=19/9 versus SR, 0.1+/-0.05%, n=16/9; P<0.05). Moreover, IK1 channel open probability was higher in cAF than in SR (13.4+/-0.4%, n=19/9 versus 11.4+/-0.7%, n=16/9; P<0.05) without changes in other channel characteristics. CONCLUSIONS: Our results demonstrate that larger basal inward rectifier K+ current in cAF consists of increased IK1 activity and constitutively active I(K,ACh). Blockade of I(K,ACh) may represent a new therapeutic target in AF.


Asunto(s)
Fibrilación Atrial/etiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Subunidades beta de la Proteína de Unión al GTP/genética , Acetilcolina/farmacología , Anciano , Apéndice Atrial/citología , Venenos de Abeja/farmacología , Carbacol/farmacología , Enfermedad Crónica , Electrofisiología , Femenino , Subunidades beta de la Proteína de Unión al GTP/fisiología , Genotipo , Humanos , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Polimorfismo de Nucleótido Simple , Potasio/metabolismo
8.
Circulation ; 110(17): 2651-7, 2004 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-15492323

RESUMEN

BACKGROUND: Although downregulation of L-type Ca2+ current (I(Ca,L)) in chronic atrial fibrillation (AF) is an important determinant of electrical remodeling, the molecular mechanisms are not fully understood. Here, we tested whether reduced I(Ca,L) in AF is associated with alterations in phosphorylation-dependent channel regulation. METHODS AND RESULTS: We used whole-cell voltage-clamp technique and biochemical assays to study regulation and expression of I(Ca,L) in myocytes and atrial tissue from 148 patients with sinus rhythm (SR) and chronic AF. Basal I(Ca,L) at +10 mV was smaller in AF than in SR (-3.8+/-0.3 pA/pF, n=138/37 [myocytes/patients] and -7.6+/-0.4 pA/pF, n=276/86, respectively; P<0.001), though protein levels of the pore-forming alpha1c and regulatory beta2a channel subunits were not different. In both groups, norepinephrine (0.01 to 10 micromol/L) increased I(Ca,L) with a similar maximum effect and comparable potency. Selective blockers of kinases revealed that basal I(Ca,L) was enhanced by Ca2+/calmodulin-dependent protein kinase II in SR but not in AF. Norepinephrine-activated I(Ca,L) was larger with protein kinase C block in SR only, suggesting decreased channel phosphorylation in AF. The type 1 and type 2A phosphatase inhibitor okadaic acid increased basal I(Ca,L) more effectively in AF than in SR, which was compatible with increased type 2A phosphatase but not type 1 phosphatase protein expression and higher phosphatase activity in AF. CONCLUSIONS: In AF, increased protein phosphatase activity contributes to impaired basal I(Ca,L). We propose that protein phosphatases may be potential therapeutic targets for AF treatment.


Asunto(s)
Fibrilación Atrial/enzimología , Fibrilación Atrial/fisiopatología , Canales de Calcio Tipo L/metabolismo , Regulación hacia Abajo , Fosfoproteínas Fosfatasas/metabolismo , Anciano , Enfermedad Crónica , Conductividad Eléctrica , Activación Enzimática , Femenino , Humanos , Isoproterenol/farmacología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/fisiología , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Proteínas Quinasas/metabolismo
9.
Circulation ; 104(21): 2551-7, 2001 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11714649

RESUMEN

BACKGROUND: Clinical and experimental evidence suggest that the parasympathetic nervous system is involved in the pathogenesis of atrial fibrillation (AF). However, it is unclear whether changes in G-protein-coupled inward rectifying K(+) current (I(K,ACh)) contribute to chronic AF. METHODS AND RESULTS: In the present study, we used electrophysiological recordings and competitive reverse-transcription polymerase chain reaction to study changes in I(K,ACh) and the level of the I(K,ACh) GIRK4 subunit in isolated human atrial myocytes and the atrial tissue of 39 patients with sinus rhythm and 24 patients with chronic AF. The density of I(K,ACh) was approximately 50% smaller in myocytes from patients with AF compared with those in sinus rhythm, and this was accompanied by decreased levels of GIRK4 mRNA. The current density of the inward rectifying K(+) current (I(K1)) was 2-fold larger during AF than in sinus rhythm, in correspondence with an increase in Kir2.1 mRNA. The larger I(K1) in AF is consistent with more negative membrane potentials in right atrial trabeculae from AF patients. Moreover, action potential duration was reduced in AF, and the action potential shortening produced by muscarinic receptor stimulation was attenuated, indicating that the changes of I(K1) and I(K,ACh) were functionally relevant. CONCLUSIONS: Chronic human AF induces transcriptionally mediated upregulation of I(K1) but downregulation of I(K,ACh) and attenuates the muscarinic receptor-mediated shortening of atrial action potentials. This suggests that atrial myocytes adapt to a chronically high rate by downregulating I(K,ACh) to counteract the shortening of the atrial effective refractory period due to electrical remodeling.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Canales de Potasio de Rectificación Interna/fisiología , Canales de Potasio/biosíntesis , Receptores Muscarínicos/fisiología , Acetilcolina/farmacología , Potenciales de Acción/efectos de los fármacos , Anciano , Fibrilación Atrial/metabolismo , Carbacol/farmacología , Células Cultivadas , Enfermedad Crónica , Regulación hacia Abajo , Conductividad Eléctrica , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Atrios Cardíacos/citología , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Masculino , Agonistas Muscarínicos/farmacología , Miocardio/citología , Canales de Potasio/genética , ARN Mensajero/biosíntesis
10.
J Cardiovasc Pharmacol ; 38(3): 438-49, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11486248

RESUMEN

The thiadiazinone derivatives EMD 60417, EMD 66430, and EMD 66398 were developed as class III antiarrhythmic agents. Their chemical structure is closely related to that of their calcium-sensitizing congener [+]-EMD 60263, and EMD 66398 possesses the methylsulfonylaminobenzoyl moiety present in the prototypical IKr blocker E-4031. We compared the electrophysiologic effects of these compounds with standard drugs (almokalant, E-4031, quinidine) in cardiac myocytes from guinea-pig ventricle and human atrium by whole-cell patch-clamp technique. The test compounds' class III action, which is related to impairment of K+ channel function, was confirmed by action potential measurements. EMD 60417, EMD 66430, EMD 66398, and almokalant (1 microM each) reversibly prolonged the action potential duration in guinea-pig myocytes. In the same cells, the rapidly activating component IKr of the delayed rectifier K+ current, which has been defined by its sensitivity to E-4031, was reduced by EMD 60417, EMD 66430, EMD 66398, and almokalant. Inhibition of IKr was concentration-dependent as determined by attenuation of tail currents. The slowly activating component IKs of the delayed rectifier K+ current was not affected. The inward rectifier K+ current IK1 was not influenced at potentials close to the reversal potential. Transient and sustained outward K+ currents (Ito, Iso) measured in human atrial myocytes were not altered by any EMD compound. L-type Ca2+ current was hardly affected at concentrations of 1-10 microM, but sodium current was decreased. Action potential prolongation by EMD 60417, EMD 66430, and EMD 66398 is due to block of IKr. INa is inhibited at higher concentrations by EMD 66430 and EMD 60417. EMD 66398 is more potent and selective for IKr than EMD 60417 and EMD 66430, and thus resembles E-4031 in structure and function.


Asunto(s)
Antiarrítmicos/farmacología , Corazón/efectos de los fármacos , Miocardio/metabolismo , Tiadiazinas/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Antiarrítmicos/química , Canales de Calcio/metabolismo , Electrofisiología , Cobayas , Corazón/fisiología , Humanos , Técnicas In Vitro , Estructura Molecular , Miocardio/citología , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Canales de Sodio/metabolismo , Tiadiazinas/química
11.
J Mol Cell Cardiol ; 33(8): 1515-25, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11448139

RESUMEN

Autoantibodies against the beta1-adrenoceptor (beta1-AAB) from patients with dilated cardiomyopathy (DCM) increase the beating frequency of cultured neonatal rat cardiomyocytes. This effect is accompanied by only a small increase in cAMP production. Here we have investigated whether beta1-AAB affect electrophysiological properties and cell shortening of isolated cardiomyocytes by interacting with the beta1-adrenoceptor. Beta1-AAB were obtained during immunoadsorption of patients with DCM and were used for experiments in isolated myocytes cultured from neonatal rat hearts, or freshly isolated from adult rat ventricles or from human right atria. The unselective beta -adrenoceptor agonist (-)-isoprenaline was studied for comparison. Immunoglobulin G (IgG) antibodies increased the spontaneous beating frequency of neonatal rat cardiomyocytes to a lesser degree than (-)-isoprenaline, but both effects were maximum and stable after 2 min. In rat ventricular and human atrial myocytes, IgG increased action potential duration (APD) in a concentration-dependent manner with larger effects on late than on early repolarization phases. Similar effects were obtained with purified beta1-AAB, whereas flow through of the chromatography column was ineffective. (-)-isoprenaline prolonged APD to the same extent during plateau and late phase of repolarization. beta1-AAB increased L-Type Ca2+ current in correspondence with the prolongation of APD. The effects of beta1-AAB and (-)-isoprenaline on APD were strongly attenuated after preincubation of the myocytes with the selective beta1-adrenoceptor antagonist (-)-bisoprolol. In addition, beta1-AAB increased cell shortening in ventricular myocytes from adult rat hearts. Beta1-AAB enhancing the beating frequency of cultured cardiomyocytes, increase L-Type Ca2+ current, APD and contractility in freshly isolated cardiomyocytes mediated via beta1-adrenoceptors. These effects may contribute to beta1-adrenoceptor-mediated cardiotoxicity in heart failure.


Asunto(s)
Potenciales de Acción , Autoanticuerpos , Cardiomiopatías/etiología , Contracción Miocárdica , Miocardio/metabolismo , Receptores Adrenérgicos beta 1/fisiología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Cardiomiopatías/inmunología , Cardiomiopatías/fisiopatología , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Humanos , Miocardio/citología , Ratas , Receptores Adrenérgicos beta 1/inmunología , Factores de Tiempo
12.
Eur J Pharmacol ; 418(1-2): 7-14, 2001 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11334859

RESUMEN

Azelastine is used for symptomatic relief of allergic rhinitis and asthma bronchiale. In vitro studies in smooth muscle cells from guinea pig trachea and ileum demonstrate that the drug blocks L-type Ca(2+) current (I(Ca, L)). However, for safety reasons, it is important to know whether azelastine also affects cardiac I(Ca, L) in therapeutically relevant concentrations. We have therefore studied the effects of azelastine on I(Ca, L) in guinea pig ventricular myocytes using standard whole-cell patch-clamp technique. Force of contraction and action potentials from isolated papillary muscles of the same species were also investigated at physiological temperature (36 degrees C). Azelastine (30 microM) significantly reduced force of contraction, shortened action potential duration, and depressed maximum upstroke velocity. I(Ca, L) was elicited by 200-ms-long clamp steps from -100 to 0 mV (one pulse every 3 s). Azelastine blocked I(Ca, L) reversibly and concentration-dependently with an IC(50) of 20.2+/-1.3 microM and a Hill coefficient of 1.1. At 10 microM, azelastine shifted steady-state inactivation by 5 mV (n=7) to more negative potentials. The time course of I(Ca, L) inactivation could be described by a double exponential function. Azelastine (10 microM) significantly shortened the slow inactivation time constant (tau(s)) from 54.2+/-2.8 ms under control conditions to 38.7+/-2.9 ms (n=16) in the presence of drug. Azelastine also reduced low-voltage-activated Ca(2+) currents with a similar IC(50) value (24 microM, at -35 mV). Since the therapeutic plasma concentrations are in the order of 10-100 nM, we conclude that azelastine does indeed affect also cardiac I(Ca, L), but the concentrations required are at least two orders of magnitude larger than those obtained during drug therapy.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Contracción Miocárdica/efectos de los fármacos , Músculos Papilares/efectos de los fármacos , Músculos Papilares/metabolismo , Ftalazinas/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Conductividad Eléctrica , Cobayas , Técnicas In Vitro , Masculino , Músculos Papilares/citología , Técnicas de Placa-Clamp
13.
Circulation ; 102(6): 692-7, 2000 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-10931811

RESUMEN

BACKGROUND: A C825T polymorphism was recently identified in the human gene encoding for the beta(3)-subunit of heterotrimeric G proteins. The 825T allele is associated with a splice variant of Gbeta(3) and enhanced signal transduction. We hypothesized that patients carrying the 825T allele exhibit the modified Gbeta(3) phenotype. The resulting enhancement of signal transduction should be detectable in the Gbetagamma-dimer-mediated acetylcholine-stimulated K(+) current (I(K,ACh)). METHODS AND RESULTS: Seventy patients undergoing cardiac surgery were genotyped for the C825T polymorphism. In right atrial myocytes from these patients, the inward rectifier K(+) currents (I(K1), I(K,ACh)) were studied with the whole-cell patch-clamp technique. Background current I(K1) was measured with depolarizing ramp pulses and quantified as inward current at -100 mV; mean amplitudes were (pA/pF) 4.98+/-0.49 (n=30/93 patients/cells) in patients with CC genotype, 4.25+/-0.36 (n=31/121 patients/cells) with TC, and 7. 46+/-1.14 (n=9/32 patients/cells; P<0.05) with TT. Conversely, mean I(K,ACh), which is maximally activated by carbachol (2 micromol/L), was reduced in patients with TT genotype (pA/pF, 4.30+/-1.33, n=9/27 patients/cells; P<0.05) compared with the other 2 groups (6.56+/-0. 54, n=30/80 and 6.16+/-0.45, n=31/117 patients/cells, for CC and TC genotype, respectively). Essentially similar results were obtained with adenosine (1 mmol/L). CONCLUSIONS: We found an association between the Gbeta(3) 825T allele and amplitude of human atrial I(K1) and I(K,ACh). Increased background current density in TT carriers could shorten action potential duration and may be due to I(K,ACh) being constitutively active in this genotype.


Asunto(s)
Alelos , Proteínas de Unión al GTP/genética , Corazón/fisiopatología , Canales de Potasio de Rectificación Interna , Canales de Potasio/fisiología , Anciano , Bloqueadores de los Canales de Calcio/uso terapéutico , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Relación Dosis-Respuesta a Droga , Conductividad Eléctrica , Femenino , Genotipo , Corazón/efectos de los fármacos , Atrios Cardíacos , Cardiopatías/tratamiento farmacológico , Cardiopatías/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , Polimorfismo Genético/genética
14.
Circ Res ; 86(6): 628-35, 2000 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-10746997

RESUMEN

Under Na(+)-free conditions, low-voltage-activated Ca(2+) currents in cardiomyocytes from various species have been described either as Ni(2+)-sensitive T-type Ca(2+) current (I(Ca(T))) or as tetrodotoxin (TTX)-sensitive Ca(2+) current (I(Ca(TTX))). So far, coexistence of the 2 currents within the same type of myocyte has never been reported. We describe experimental conditions under which I(Ca(T)) and I(Ca(TTX)) can be separated and studied in the same cell. Rat and guinea pig ventricular myocytes were investigated with the whole-cell voltage-clamp technique in Na(+)-free solutions. Whereas rat myocytes lack I(Ca(T)) and exhibit I(Ca(TTX)) only, guinea pig myocytes possess both of these low-voltage-activated Ca(2+) currents, which are separated pharmacologically by superfusion with TTX or Ni(2+). I(Ca(T)) and I(Ca(TTX)) were of similar amplitude but significantly differed in their electrophysiological properties: I(Ca(TTX)) activated at more negative potentials than did I(Ca(T)), the potential for half-maximum steady-state inactivation was more negative, and current deactivation and recovery from inactivation were faster. I(Ca(TTX)) but not I(Ca(T)) increased after membrane rupture ("run-up"). Isolation of I(Ca(TTX)) by application of the bivalent cation Ni(2+) is critical because of possible shifts in voltage dependence. Therefore, we investigated whether the T-type Ca(2+) channel blocker mibefradil (10 micromol/L) is a suitable tool for the study of I(Ca(TTX)). However, mibefradil not only blocked I(Ca(T)) by 85+/-2% but also decreased I(Ca(TTX)) by 48+/-8%. We conclude that under Na(+)-free conditions I(Ca(T)) and I(Ca(TTX)) coexist in guinea pig ventricular myocytes and that both currents are sensitive to mibefradil. Future investigations of I(Ca(T)) will have to consider the TTX-sensitive current component to avoid possible interference.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/metabolismo , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Mibefradil/farmacología , Miocardio/metabolismo , Tetrodotoxina/farmacología , Animales , Membrana Celular/fisiología , Cobayas , Ventrículos Cardíacos , Homeostasis , Cinética , Masculino , Miocardio/citología , Ratas , Ratas Wistar , Rotura
15.
Naunyn Schmiedebergs Arch Pharmacol ; 360(4): 445-56, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10551282

RESUMEN

Putative beta4-adrenoceptors mediate cardiostimulation and arrhythmias in mammalian heart. Both beta1- and putative beta4-adrenoceptors mediate arrhythmias but through different mechanisms. To elucidate further the mechanisms of cardiostimulation and arrhythmias we measured Ca2+ transients and L-type Ca2+ currents in mouse ventricular myocytes. We used (-)-CGP 12177, an antagonist of beta1- and beta2-adrenoceptors with agonist properties at the putative beta4-adrenoceptor, and (-)-isoprenaline as an agonist for beta1- and beta2-adrenoceptors. (-)-CGP 12177 increased Ca2+ transients in electrically stimulated cells loaded with Indo-1. The maximum increase of Ca2+ transients caused by (-)-CGP 12177 amounted to approximately one-third of that caused by maximally effective (-)-isoprenaline concentrations. Both (-)-CGP 12177 and (-)-isoprenaline caused concentration-dependent arrhythmic Ca2+ transients. The arrhythmias appeared at paced Ca2+ transients and between paced Ca2+ transients. The arrhythmic potency of (-)-CGP 12177 (-logEC50=9.4) was approximately 40 times greater than that of (-)-isoprenaline (-logEC50=7.8). L-type Ca2+ current was measured in the whole cell configuration of the patch clamp technique. In the presence of both 3-isobutyl 1-methylxanthine (6 micromol/l) and (-)-propranolol (500 nmol/l), (-)-CGP 12177 (100 nmol/l) increased significantly L-type Ca2+ current by 19% of the effect of (-)-isoprenaline. The (-)-CGP 12177-evoked increase of Ca2+ transients contrasts with the smaller effects on L-type Ca2+ current, suggesting that activation of the putative beta4-adrenoceptor causes a more efficient Ca2+-induced Ca2+ release than activation of the beta1-adrenoceptor. Beta4-Adrenoceptors mediate arrhythmias with smaller Ca2+ transients and smaller increases of L-type Ca2+ current than beta1-adrenoceptors, in line with different but still unknown mechanisms as previously suggested for the intact heart.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Propanolaminas/farmacología , Receptores Adrenérgicos beta/efectos de los fármacos , 1-Metil-3-Isobutilxantina/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica , Ventrículos Cardíacos , Técnicas In Vitro , Isoproterenol/farmacología , Masculino , Ratones , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Propranolol/farmacología , Receptores Adrenérgicos beta/fisiología , Receptores Adrenérgicos beta 1/efectos de los fármacos , Receptores Adrenérgicos beta 1/fisiología , Factores de Tiempo
16.
Cardiovasc Res ; 42(1): 173-82, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10435008

RESUMEN

OBJECTIVES: The reported increase in basal activity of hearts from transgenic mice (TG4) overexpressing the human beta 2-adrenoceptor (beta 2-AR) was explained by spontaneously active beta 2-ARs that stimulate the beta-adrenergic cascade in the absence of an agonist. In order to examine altered myocardial function on a cellular level, we have investigated L-type calcium current (ICa,L) and cell shortening in ventricular myocytes from TG4 hearts. Myocytes from littermates (LM) and wild type animals (WT) served as controls. METHODS: Cardiac beta-AR density was measured by [125I]-iodocyanopindolol binding to ventricular membranes. ICa,L was assessed by standard whole-cell voltage clamp technique. Contractility was measured as cell shortening in ventricular myocytes and as force of contraction in electrically stimulated left atria. RESULTS: Overexpression of beta 2-ARs was confirmed by an almost 400-fold increase in beta-AR density. The beta 1:beta 2-AR ratio in WT mice was 71:29. Myocytes from TG4 and LM mice were similar in size as judged by membrane capacitance and two dimensional cell area. ICa,L amplitude was significantly lower in TG4 than in LM myocytes (with 2 mM [Ca2+]o -4.82 +/- 0.48 vs. -6.56 +/- 0.38 pA/pF, respectively). In TG4 myocytes, the ICa,L response to isoproterenol (1 microM) was almost abolished. Cell shortening was not different in physiological [Ca2+]o, but smaller in maximum [Ca2+]o when comparing TG4 to control myocytes. Basal force of contraction in left atria did not differ between TG4 and LM at any age investigated. In TG4 left atria the inotropic response to isoproterenol was also absent, whereas responses to high [Ca2+]o or dibutyryl-cAMP (1 mM) were present but reduced. The rate of spontaneous beating of right atria was elevated in TG4 mice. CONCLUSIONS: Since only spontaneous beating rate but neither basal ICa,L amplitude nor basal contractile activity were elevated, our data fail to reveal evidence for spontaneously active, stimulating beta 2-ARs in left atrium and ventricle. A contractile deficit unrelated to the beta-adrenoceptor pathway is evident in TG4 myocytes and left atria.


Asunto(s)
Canales de Calcio/metabolismo , Contracción Miocárdica , Miocardio/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos beta/farmacología , Análisis de Varianza , Animales , Bucladesina/farmacología , Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas , Estimulación Eléctrica , Expresión Génica , Atrios Cardíacos , Yodocianopindolol/farmacología , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Miocardio/citología , Técnicas de Placa-Clamp , Receptores Adrenérgicos beta 2/genética , Transducción de Señal , Estadísticas no Paramétricas , Estimulación Química
17.
Am J Physiol ; 277(1): H107-18, 1999 07.
Artículo en Inglés | MEDLINE | ID: mdl-10409188

RESUMEN

In rat ventricle, two Ca(2+)-insensitive components of K(+) current have been distinguished kinetically and pharmacologically, the transient, 4-aminopyridine (4-AP)-sensitive I(to) and the sustained, tetraethylammonium (TEA)-sensitive I(K). However, a much greater diversity of depolarization-activated K(+) channels has been reported on the level of mRNA and protein. In the search for electrophysiological evidence of further current components, the whole cell voltage-clamp technique was used to analyze steady-state inactivation of outward currents by conditioning potentials in a wide voltage range. Peak (I(peak)) and late (I(late)) currents during the test pulse were analyzed by Boltzmann curve fitting, producing three fractions each. Fractions a and b had different potentials of half-maximum inactivation (V(0.5)); the third residual fraction, r, did not inactivate. Fractions a for I(peak) and I(late) had similar relative amplitudes and V(0.5) values, whereas size and V(0.5) of fractions b differed significantly between I(peak) and I(late). Only b of I(peak) was transient, suggesting a relation with I(to), whereas a, b, and r of I(late) appeared to be three different sustained currents. Therefore, four individual outward current components were distinguished: I(to) (b of I(peak)), I(K) (a), the steady-state current I(ss) (r), and the novel current I(Kx) (b of I(late)). This was further supported by differential sensitivity to TEA, 4-AP, clofilium, quinidine, dendrotoxin, heteropodatoxin, and hanatoxin. With the exception of I(to), none of the currents exhibited a marked transmural gradient. Availability of I(K) was low at resting potential; nevertheless, I(K) contributed to action potential shortening in hyperpolarized subendocardial myocytes. In conclusion, on the basis of electrophysiological and pharmacological evidence, at least four components contribute to outward current in rat ventricular myocytes.


Asunto(s)
Potenciales de Acción , Ventrículos Cardíacos/citología , Función Ventricular , 4-Aminopiridina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Venenos Elapídicos/farmacología , Electrofisiología , Ventrículos Cardíacos/efectos de los fármacos , Masculino , Neurotoxinas/farmacología , Técnicas de Placa-Clamp , Péptidos/farmacología , Bloqueadores de los Canales de Potasio , Ratas , Ratas Wistar , Tetraetilamonio/farmacología
18.
J Cardiovasc Pharmacol ; 33(2): 301-8, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10028941

RESUMEN

The thiadiazinone enantiomers [+]-EMD 60263 and [-]-EMD 60264 ((+)-5-(1-(alpha-ethylimino-3,4-dimethoxybenzyl)-1,2,3,4-tetrah ydroquinoline-6-yl)-6-methyl-3,6-dihydro-2H-1,3,4-thiadiazine-2 -on) exhibit distinct stereoselectivity for Ca2+-sensitizing action ([+]-enantiomer) and phosphodiesterase inhibition ([-]-enantiomer). However, in isolated guinea pig papillary muscle, both compounds cause an action-potential prolongation that has been related to a nonselective depression of the delayed rectifier potassium current. Because [-]-EMD 60264 did not increase force of contraction despite phosphodiesterase inhibition, we postulated that one or several additional actions may oppose the anticipated positive inotropic effect. Therefore we investigated whether other membrane currents were also affected in voltage-clamped ventricular cardiomyocytes. Both [+]-EMD 60263 and [-]-EMD 60264 reduced sodium current as well as L-type calcium current in guinea pig ventricular myocytes, but steady-state inactivation or conductance curves of I(Na) and I(Ca) were not shifted along the voltage axis. Inward rectifier and transient outward current were studied in rat myocytes, but neither current was affected. We conclude that the positive inotropic action of [+]-EMD 60263 can be explained by prevalence of the Ca2+-sensitizing effect over its inhibitory actions on Na+ and Ca2+ current, whereas the negative inotropic effect of [-]-EMD 60264 may be caused by inhibition of I(Ca) predominating over PDE inhibition.


Asunto(s)
Ventrículos Cardíacos/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Tiadiazinas/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/metabolismo , Cobayas , Técnicas In Vitro , Membranas/efectos de los fármacos , Potasio/metabolismo , Ratas , Sodio/metabolismo , Estereoisomerismo
19.
Br J Pharmacol ; 125(4): 659-66, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9831899

RESUMEN

1. Tedisamil is a new antiarrhythmic drug with predominant class III action. The aim of the present study was to investigate the blocking pattern of the compound on the transient outward current (I(to)) in human subepicardial myocytes isolated from explanted left ventricles. Using the single electrode whole cell voltage clamp technique, I(to) was analysed after appropriate voltage inactivation of sodium current and block of calcium current. 2. Tedisamil reduced the amplitude of peak I(to), but did not affect the amplitude of non-inactivating outward current. The drug accelerated the apparent rate of I(to) inactivation. The reduction in time constant of I(to) inactivation depended on drug concentration, the apparent IC50 value was 4.4 microM. 3. Tedisamil affected I(to) amplitude in a use-dependent manner. After 2 min at -80 mV, maximum block of I(to) was reached after 4-5 clamp steps either at the frequency of 0.2 or 2 Hz, indicating that the block was not frequency-dependent in an experimentally relevant range. Recovery from block was very slow and proceeded with a time constant of 12.1+/-1.8 s. Also in the presence of drug, a fraction of channels recovered from inactivation with a similar time constant as in control myocytes (i.e. 81+/-40 ms and 51+/-8 ms, respectively, n.s.). 4. From the onset of fractional block of I(to) by tedisamil during the initial 60 ms of a clamp step, we calculated k1 = 9 x 10(6) mol(-1) s(-1) for the association rate constant, and k2 = 23 s(-1) for the dissociation rate constant. The resulting apparent KD was 2.6 microM and is similar to the IC50 value. 5. The effects of tedisamil on I(to) could be simulated by assuming a four state channel model where the drug binds to the channel in an open (activated) conformation. It is concluded that in human subepicardial myocytes tedisamil is an open channel blocker of I(to) and that this effect probably contributes to the antiarrhythmic potential of this drug.


Asunto(s)
Antiarrítmicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Ciclopropanos/farmacología , Bloqueo Cardíaco/inducido químicamente , Ventrículos Cardíacos/efectos de los fármacos , Miocardio/citología , Potenciales de Acción/efectos de los fármacos , Sitios de Unión , Calcio/farmacocinética , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Bloqueadores de los Canales de Potasio , Sodio/farmacocinética , Factores de Tiempo
20.
Basic Res Cardiol ; 93 Suppl 1: 60-5, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9833132

RESUMEN

Cardiac action potentials undergo characteristic changes in response to increasing pacing frequency, i.e., the resting potential becomes less negative (depolarization), the amplitude decreases, and the action potential duration (APD) shortens. The electrophysiological properties of the major cardiac inward and outward currents are discussed with respect to their possible contribution to rate-dependent changes in AP shape. Short diastolic intervals may not allow sufficient time for channel recovery. Incomplete recovery from inactivation will reduce both inward (INa, ICa) and outward (Ito) currents and hence lead to APD shortening or prolongation, respectively. Incomplete deactivation during diastole will increase current and, in the case of the delayed rectifier IKs, produces rate-dependent APD shortening. Heterogeneity in current density of myocytes from various regions within the ventricular wall complicates the direct translation of rate-dependent changes in APD into changes of the QT interval of the ECG. With increasing rates of stimulation, K+ accumulates within the tubular system. Membrane depolarization may be attributed to this frequency-dependent increase in [K+]o, whereas APD shortening is not mimicked by high [K+]o. Nevertheless, the various cardiac K+ channels differ in their sensitivity to extracellular [K+]o. Some of the frequency-dependent effects of drugs with class III action may be related to an influence of [K+]o on drug potency to block K+ channels.


Asunto(s)
Electrocardiografía , Frecuencia Cardíaca/fisiología , Potenciales de Acción/fisiología , Estimulación Eléctrica , Humanos , Iones , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Función Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...