Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Internist (Berl) ; 62(11): 1180-1190, 2021 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-34648044

RESUMEN

Digital health solutions, applications of artificial intelligence (AI) and new technologies, such as cardiac magnetic resonance imaging and cardiac human genetics are currently being validated in cardiac healthcare pathways. They show promising approaches for improving existing healthcare structures in the future by strengthening the focus on predictive, preventive and personalized medicine. In addition, the accompanying use of digital health applications will become increasingly more important in the future healthcare, especially in patients with chronic diseases. In this article, the authors describe a case of chronic heart failure (HF) as an example to provide an overview of how digitalized healthcare can be efficiently designed across sectors and disciplines in the future. Moreover, the importance of a self-determined patient management for the treatment process itself is underlined. Since HF is frequently accompanied by various comorbidities during the course of the disease that are often recognized only after a delay, the necessity for a timely simultaneous and preventive treatment of multiple comorbidities in cardiovascular diseases is emphasized. Against this background the currently separately applied disease management programs (DMP) are critically questioned. The development of a holistic DMP encompassing all indications for the treatment of chronic diseases may pave the way to a more efficient medical care system.


Asunto(s)
Inteligencia Artificial , Insuficiencia Cardíaca , Atención a la Salud , Predicción , Corazón , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Humanos
3.
Biochem Biophys Res Commun ; 424(2): 315-20, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22749993

RESUMEN

Kir2.x channels form the molecular basis of cardiac I(K1) current and play a major role in cardiac electrophysiology. However, there is a substantial lack of selective Kir2 antagonists. We found the ß(3)-adrenoceptor antagonist SR59230A to be an inhibitor of Kir2.x channels. Therefore, we characterized the effects of SR59230A on Kir2.x and other relevant cardiac potassium channels. Cloned channels were expressed in the Xenopus oocyte expression system and measured with the double-microelectrode voltage clamp technique. SR59230A inhibited homomeric Kir2.1 channels with an IC(50) of 33µM. Homomeric Kir2.2 and Kir2.3 channels and Kir2.x heteromers were also inhibited by SR59230A with similar potency. In contrast, no relevant inhibitory effects of SR59230A were found in cardiac Kv1.5, Kv4.3 and KvLQT1/minK channels. In hERG channels, SR59230A only induced a weak inhibition at a high concentration. These findings establish SR59230A as a novel inhibitor of Kir2.1-2.3 channels with a favorable profile with respect to additional effects on other cardiac repolarizing potassium channels.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 3/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Propanolaminas/farmacología , Receptores Adrenérgicos beta 3/metabolismo , Animales , Oocitos , Xenopus laevis
4.
Biochem Biophys Res Commun ; 408(2): 218-24, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21458413

RESUMEN

Inherited cardiac arrhythmias are caused by genetic defects in ion channels and associated proteins. Mutations in these channels often do not affect their biophysical properties, but rather interfere with their trafficking to the cell membrane. Accordingly, strategies that could reroute the mutated channels to the membrane should be sufficient to restore the electrical properties of the affected cells, thereby suppressing the underlying arrhythmia. We identified here both, embryonic and adult zebrafish breakdance (bre) as a valuable model for human Long-QT syndrome. Electrocardiograms of adult homozygous bre mutants exhibit significant QT prolongation caused by delayed repolarization of the ventricle. We further show that the bre mutation (zERG(I59S)) disrupts ERG protein trafficking, thereby reducing the amount of active potassium channels on the cell membrane. Interestingly, improvement of channel trafficking by cisapride or dimethylsulfoxid is sufficient to reconstitute ERG channels on the cell membrane in a manner that suffices to suppress the Long-QT induced arrhythmia in breakdance mutant zebrafish. In summary, we show for the first time that therapeutic intervention can cure protein trafficking defects and the associated cardiac arrhythmia in vivo.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Síndrome de QT Prolongado/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Modelos Animales de Enfermedad , Canales de Potasio Éter-A-Go-Go/genética , Células HEK293 , Frecuencia Cardíaca/genética , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Mutación , Transporte de Proteínas/genética , Disfunción Ventricular/genética , Disfunción Ventricular/fisiopatología , Pez Cebra , Proteínas de Pez Cebra/genética
5.
Herzschrittmacherther Elektrophysiol ; 32(2): 227-235, 2021 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-33982176

RESUMEN

The possibility of outpatient implantation of defibrillators, devices for cardiac resynchronization, and event recorders (collectively called cardiac implantable electronic devices, CIEDs) is becoming increasingly important. In Germany, only a few options for outpatient implantation are currently realized. Furthermore, there is a lack of uniform, recognized, and binding quality criteria. This article provides insight into the current contract constellations for outpatient surgery and defines a first, holistic quality concept for outpatient implantations of CIEDs. The present works aims to initiate a discourse in the specialist society in order to define a coordinated, binding quality concept. Then, this should serve as the basis for future outpatient implantation services, enabling comparability and to contribute long-term evidence.


Asunto(s)
Terapia de Resincronización Cardíaca , Desfibriladores Implantables , Marcapaso Artificial , Dispositivos de Terapia de Resincronización Cardíaca , Alemania , Humanos , Pacientes Ambulatorios
6.
J Mol Cell Cardiol ; 48(1): 230-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19540844

RESUMEN

Hereditary long QT syndrome (LQTS) is a cardiovascular disorder characterized by prolongation of the QT interval on the surface ECG and a high risk for arrhythmia-related sudden death. Mutations in a cardiac voltage-gated potassium channel, KCNQ1, account for the most common form of LQTS, LQTS1. The objective of this study was the characterization of a novel KCNQ1 mutation linked to LQTS. Electrophysiological properties and clinical features were determined and compared to characteristics of a different mutation at the same position. Single-strand conformation polymorphism analysis followed by direct sequencing was performed to screen LQTS genes for mutations. A novel missense mutation in the KCNQ1 gene, KCNQ1 P320H, was identified in the index patient presenting with recurrent syncope and aborted sudden death triggered by physical stress and swimming. Electrophysiological analyses of KCNQ1 P320H and the previously reported KCNQ1 P320A mutation indicate that both channels are non-functional and suppress wild type I(Ks) in a dominant-negative fashion. Based on homology modeling of the KCNQ1 channel pore region, we speculate that the proline residue at position 320 limits flexibility of the outer pore and is required to maintain the functional architecture of the selectivity filter/pore helix arrangement. Our observations on the KCNQ1 P320H mutation are consistent with previous studies indicating that pore mutations in potassium channel alpha-subunits are associated with more severe electrophysiological and clinical phenotypes than mutations in other regions of these proteins. This study emphasizes the significance of mutation screening for diagnosis, risk-assessment, and mutation-site specific management in LQTS patients.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Síndrome de Romano-Ward/genética , Adulto , Análisis Mutacional de ADN , Electrofisiología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Canal de Potasio KCNQ1/genética , Masculino , Mutación , Linaje , Canales de Potasio con Entrada de Voltaje/genética
7.
Circulation ; 117(7): 866-75, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18250272

RESUMEN

BACKGROUND: Genetic predisposition is believed to be responsible for most clinically significant arrhythmias; however, suitable genetic animal models to study disease mechanisms and evaluate new treatment strategies are largely lacking. METHODS AND RESULTS: In search of suitable arrhythmia models, we isolated the zebrafish mutation reggae (reg), which displays clinical features of the malignant human short-QT syndrome such as accelerated cardiac repolarization accompanied by cardiac fibrillation. By positional cloning, we identified the reg mutation that resides within the voltage sensor of the zebrafish ether-à-go-go-related gene (zERG) potassium channel. The mutation causes premature zERG channel activation and defective inactivation, which results in shortened action potential duration and accelerated cardiac repolarization. Genetic and pharmacological inhibition of zERG rescues recessive reg mutant embryos, which confirms the gain-of-function effect of the reg mutation on zERG channel function in vivo. Accordingly, QT intervals in ECGs from heterozygous and homozygous reg mutant adult zebrafish are considerably shorter than in wild-type zebrafish. CONCLUSIONS: With its molecular and pathophysiological concordance to the human arrhythmia syndrome, zebrafish reg represents the first animal model for human short-QT syndrome.


Asunto(s)
Arritmias Cardíacas/genética , Modelos Animales de Enfermedad , Canales de Potasio Éter-A-Go-Go/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Potenciales de Acción/genética , Sustitución de Aminoácidos , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/embriología , Arritmias Cardíacas/fisiopatología , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Canales de Potasio Éter-A-Go-Go/deficiencia , Canales de Potasio Éter-A-Go-Go/genética , Genotipo , Corazón/embriología , Activación del Canal Iónico/genética , Mutación Missense , Contracción Miocárdica , Oocitos , Técnicas de Placa-Clamp , Potasio/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Bloqueo Sinoatrial/tratamiento farmacológico , Bloqueo Sinoatrial/genética , Bloqueo Sinoatrial/fisiopatología , Síndrome , Terfenadina/uso terapéutico , Xenopus laevis , Pez Cebra/embriología , Pez Cebra/fisiología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Biochem Biophys Res Commun ; 381(2): 159-64, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19232322

RESUMEN

The zebrafish is increasingly recognized as an animal model for the analysis of hERG-related diseases. However, functional properties of the zebrafish orthologue of hERG have not been analyzed yet. We heterologously expressed cloned ERG channels in Xenopus oocytes and analyzed biophysical properties using the voltage clamp technique. zERG channels conduct rapidly activating and inactivating potassium currents. However, compared to hERG, the half-maximal activation voltage of zERG current is shifted towards more positive potentials and the half maximal steady-state inactivation voltage is shifted towards more negative potentials. zERG channel activation is delayed and channel deactivation is accelerated significantly. However, time course of zERG conducted current under action potential clamp is highly similar to the human orthologue. In summary, we show that ERG channels in zebrafish exhibit biophysical properties similar to the human orthologue. Considering the conserved channel function, the zebrafish represents a valuable model to investigate human ERG channel related diseases.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Potenciales de la Membrana , Oocitos , Xenopus , Pez Cebra/genética , Proteínas de Pez Cebra/agonistas , Proteínas de Pez Cebra/genética
9.
J Mol Cell Cardiol ; 44(1): 84-94, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18035370

RESUMEN

Inhibition of I(K1) currents by adrenergic alpha(1) receptors has been observed in cardiomyocytes and has been linked to arrhythmogenesis in an animal model. Both PKC-dependent and PKC-independent pathways have been implied in this regulation. The underlying molecular mechanisms, however, have not been elucidated to date. The molecular basis of native I(K1) current is mainly formed by Kir2.1 (KCNJ2), Kir2.2 (KCNJ12) and Kir2.3 (KCNJ4) channels that are differentially regulated by protein kinases. We therefore sought to investigate the role of those different Kir2.x channel subunits in this regulation and to identify the major signalling pathways involved. Adrenergic alpha(1A) receptors (the predominant cardiac isoform) were co-expressed with cloned Kir2.1, Kir2.2 and Kir2.3 channels in Xenopus oocytes and electrophysiological experiments were performed using two-microelectrode voltage clamp. Native I(K1) currents were measured with the whole-cell patch clamp technique in isolated rat ventricular cardiomyocytes. Activation of co-expressed adrenergic alpha(1A) receptors by phenylephrine induced differential effects in Kir2.x channels. No effect was noticed in Kir2.1 channels. However, a marked inhibitory effect was observed in Kir2.2 channels. This regulation was not attenuated by inhibitors of PKC, CamKII and PKA (chelerythrine, KN-93, KT-5720), and mutated Kir2.2 channels lacking functional phosphorylation sites for PKC and PKA exhibited the same effect as Kir2.2 wild-type channels. By contrast, the regulation could be suppressed by the general tyrosine kinase inhibitor genistein and by the src tyrosine kinase inhibitor PP2 indicating an essential role of src kinases. This finding was validated in rat ventricular cardiomyocytes where co-application of PP2 strongly attenuated the inhibitory regulation of I(K1) current by adrenergic alpha(1) receptors. The inactive analogue PP3 was tested as negative control for PP2 and did not reproduce the effects of PP2. In Kir2.3 channels, a marked inhibitory effect of alpha(1A) receptor activation was observed. This regulation could be attenuated by inhibition of PKC with chelerythrine or with Ro-32-0432, but not by tyrosine kinase inhibition with genistein. In summary, on the molecular level the inhibitory regulation of I(K1) currents by adrenergic alpha(1A) receptors is probably based on effects on Kir2.2 and Kir2.3 channels. Kir2.2 is regulated via src tyrosine kinase pathways independent of protein kinase C, whereas Kir2.3 is inhibited by protein kinase C-dependent pathways. Src tyrosine kinase pathways are essential for the inhibition of native I(K1) current by adrenergic alpha(1) receptors. This regulation may contribute to arrhythmogenesis under adrenergic stimulation.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio de Rectificación Interna/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ratas , Xenopus
10.
Biochem Biophys Res Commun ; 377(3): 981-6, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18976636

RESUMEN

The renal inward rectifier potassium channel Kir7.1 has been proposed to be functionally important for tubular K(+) recycling and secretion. This study investigated the regulation of Kir7.1 by PKA and PKC. Cloned human Kir7.1 channels were expressed heterologously in Xenopus oocytes. After pharmacological PKC activation, Kir7.1 currents were strongly inhibited. Co-application of PKC inhibitors attenuated this effect. Inactivation of PKC consensus sites also strongly attenuated the effect with a single site ((201)S) being essential for almost the total PKC sensitivity. In contrast, PKA activation induced an increase of Kir7.1 currents. This effect was absent in mutant Kir7.1 channels lacking PKA consensus site (287)S. In summary, this study demonstrates the dual regulation of Kir7.1 channel function by PKA and PKC. Structurally, these regulations depend on two key residues in the C-terminal channel domain ((Ser)201 for PKC and (Ser)287 for PKA).


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Riñón/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Proteína Quinasa C/metabolismo , Animales , Clonación Molecular , Activación Enzimática , Humanos , Riñón/enzimología , Datos de Secuencia Molecular , Oocitos , Fosforilación , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/genética , Estructura Terciaria de Proteína , Serina/genética , Serina/metabolismo , Transfección , Xenopus
11.
Eur J Pharmacol ; 579(1-3): 98-103, 2008 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-18054910

RESUMEN

The antihypertensive drug doxazosin has been associated with an increased risk for congestive heart failure and cardiomyocyte apoptosis. Human ether-a-go-go-related gene (hERG) K(+) channels, previously shown to be blocked by doxazosin at therapeutically relevant concentrations, represent plasma membrane receptors for the antihypertensive drug. To elucidate the molecular basis for doxazosin-associated pro-apoptotic effects, cell death was studied in human embryonic kidney cells using three independent apoptosis assays. Doxazosin specifically induced apoptosis in hERG-expressing HEK cells, while untransfected control groups were insensitive to treatment with the antihypertensive agent. An unexpected biological mechanism has emerged: binding of doxazosin to its novel membrane receptor, hERG, triggers apoptosis, possibly representing a broader pathophysiological mechanism in drug-induced heart failure.


Asunto(s)
Antihipertensivos/farmacología , Apoptosis/efectos de los fármacos , Doxazosina/farmacología , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Antagonistas Adrenérgicos alfa/farmacología , Línea Celular , Electrofisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Coloración y Etiquetado
12.
Naunyn Schmiedebergs Arch Pharmacol ; 378(1): 73-83, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18458880

RESUMEN

The antidepressant mianserin exhibits a tetracyclic structure that is different from typical tricyclic antidepressants (TCA) and that of selective serotonin reuptake inhibitors. In comparison to the older TCA, mianserin has been shown to have a superior risk profile regarding proarrhythmic effects, both in vitro and in vivo. However, the underlying molecular electrophysiological basis has not been elucidated to date. Therefore, we studied the effects of mianserin on cardiac hERG potassium channels, the predominant target of drug-induced proarrhythmia. HERG channels were expressed in the Xenopus oocyte expression system and in human embryonic kidney (HEK) cells and currents were measured with two-microelectrode voltage-clamp and whole-cell patch-clamp, respectively. Mianserin inhibited hERG currents in a dose-dependent manner with an IC(50) of 3.2 micromol/l in HEK cells. Onset of blockade was slow and the inhibitory effect was not reversible upon wash-out of the drug. In hERG channel mutants, Y652A and F656A, lacking aromatic residues in the S6 domain, the effect of mianserin was significantly reduced in comparison to the wild type. Mianserin inhibited hERG currents in the open and inactivated state, but not in the closed states. HERG inactivation kinetics were significantly altered by mianserin without marked effects on channel activation kinetics. The inhibitory effect was not frequency dependent. In conclusion, mianserin is a low-affinity hERG-blocking agent. However, taken together with the lack of APD-prolongation shown in other studies, mianserin seems to have a good safety profile. Lack of consistent QT prolonging effects of mianserin in previous studies may therefore be linked to additional effects such as inhibition of other cardiac ion channels. However, as demonstrated by clinical case reports, mianserin can induce proarrhythmic effects in susceptible patients. Therefore, in patients with complex co-medication (i.e., additional hERG-blocking agents) and in patients with risk factors for acquired long QT syndrome as well as in cases of overdose, adequate monitoring should be recommended.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Mianserina/toxicidad , Animales , Antidepresivos de Segunda Generación/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Humanos , Concentración 50 Inhibidora , Riñón/citología , Mianserina/administración & dosificación , Oocitos/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Pruebas de Toxicidad , Xenopus
13.
Biochem Biophys Res Commun ; 364(3): 429-35, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17961513

RESUMEN

The catechin EGCG is the main flavonoid compound of green tea and has received enormous pharmacological attention because of its putative beneficial health effects. This study investigated for the first time the effect of EGCG on hERG channels, the main pharmacological target of drugs that cause acquired long QT syndrome. Cloned hERG channels were expressed in Xenopus oocytes and in HEK293 cells. Heterologous hERG currents were inhibited by EGCG with an IC50 of 6.0 micromol/l in HEK293 cells and an IC50 of 20.5 micromol/l in Xenopus laevis oocytes. Onset of effect was slow and only little recovery from inhibition was observed upon washout. In X. laevis oocytes EGCG inhibited hERG channels in the open and inactivated states, but not in the closed states. The half-maximal activation voltage of hERG currents was shifted by EGCG towards more positive potentials. In conclusion, EGCG is a low-affinity inhibitor of hERG sharing major electrophysiological features with pharmaceutical hERG antagonists.


Asunto(s)
Catequina/análogos & derivados , Canales de Potasio Éter-A-Go-Go/fisiología , Activación del Canal Iónico/fisiología , Riñón/fisiología , Oocitos/fisiología , Potasio/metabolismo , Té/química , Animales , Catequina/administración & dosificación , Células Cultivadas , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Riñón/efectos de los fármacos , Oocitos/efectos de los fármacos , Xenopus laevis
14.
Naunyn Schmiedebergs Arch Pharmacol ; 375(5): 311-22, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17534603

RESUMEN

beta(3)-adrenoceptors have recently been shown to induce a complex modulation of intracellular signaling pathways including cyclic guanine monophosphate, cyclic adenosine monophosphate, nitric oxide, and protein kinases A and C. They are expressed in a broad variety of tissues including the myocardium, vascular smooth muscle, and endothelium. In those tissues, resting membrane potential is controlled mainly by inwardly rectifying potassium channels of the Kir2 family namely, Kir2.1 in the vascular smooth muscle, Kir2.1-2.3 in the myocardium, and Kir2.1-2.2 in the endothelium. In the present study, we investigated the possible modulation of Kir2 channel function by beta(3)-adrenoceptors in an expression system. Human-cloned beta(3)-adrenoceptors and Kir2.1 (KCNJ2), Kir2.2 (KCNJ12), and Kir2.3 (KCNJ4) channels were coexpressed in Xenopus oocytes, and currents were measured with double-microelectrode voltage clamp. Activation of beta(3)-adrenoceptors with isoproterenol resulted in markedly increased currents in Kir2.1 and in Kir2.2 potassium channels with EC50 values of 27 and 18 nM, respectively. In contrast, Kir2.3 currents were not modulated. Coapplication of specific inhibitors of protein kinase A (KT-5720) and calmodulin kinase II (KN-93) had no effects on the observed regulation in Kir2.1. However, coapplication of protein kinase C (PKC) inhibitors staurosporine and chelerythrine suppressed the observed effect. In Kir2.2, coapplication of KT-5720 reduced the effect of beta(3)-adrenoceptor activation. No differences in current increase after application of isoproterenol were observed between mutant Kir2.2 potassium channels lacking all functional PKC phosphorylation sites and Kir2.2 wild-type channels. In heteromeric Kir2.x channels, all types of heteromers were activated. The effect was most pronounced in Kir2.1/Kir2.2 and in Kir2.2/Kir2.3 channels. In summary, homomeric and heteromeric Kir2.x channels are activated by beta(3)-adrenoceptors via different protein kinase-dependent pathways: Kir2.1 subunits are modulated by PKC, whereas Kir2.2 is modulated by protein kinase A. In heteromeric composition, a marked activation of currents can be observed particularly with involvement of Kir2.2 subunits. This regulation may contribute to the hyperpolarizing effects of beta(3)-adrenoceptors in tissues that exhibit modulation by Kir2 channel function.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Proteína Quinasa C/fisiología , Receptores Adrenérgicos beta 3/fisiología , Animales , Femenino , Oocitos/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio , Canales de Potasio de Rectificación Interna/genética , Transducción de Señal , Xenopus laevis
15.
Naunyn Schmiedebergs Arch Pharmacol ; 376(4): 275-84, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17965852

RESUMEN

The anticholinergic antiparkinson drug orphenadrine is an antagonist at central and peripheral muscarinic receptors. Orphenadrine intake has recently been linked to QT prolongation and Torsade-de-Pointes tachycardia. So far, inhibitory effects on I (Kr) or cloned HERG channels have not been examined. HERG channels were heterologously expressed in a HEK 293 cell line and in Xenopus oocytes and HERG current was measured using the whole cell patch clamp and the double electrode voltage clamp technique. Orphenadrine inhibits cloned HERG channels in a concentration dependent manner, yielding an IC(50) of 0.85 microM in HEK cells. Onset of block is fast and reversible upon washout. Orphenadrine does not alter the half-maximal activation voltage of HERG channels. There is no shift of the half-maximal steady-state-inactivation voltage. Time constants of direct channel inactivation are not altered significantly and there is no use-dependence of block. HERG blockade is attenuated significantly in mutant channels lacking either of the aromatic pore residues Y652 and F656. In conclusion, we show that the anticholinergic agent orphenadrine is an antagonist at HERG channels. These results provide a novel molecular basis for the reported proarrhythmic side effects of orphenadrine.


Asunto(s)
Antiparkinsonianos/farmacología , Antagonistas Colinérgicos/farmacología , Canales de Potasio Éter-A-Go-Go/fisiología , Orfenadrina/farmacología , Animales , Línea Celular , Clonación Molecular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Humanos , Mutación , Oocitos/efectos de los fármacos , Oocitos/fisiología , Xenopus laevis
16.
J Mol Med (Berl) ; 84(1): 46-56, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16258766

RESUMEN

To elucidate the ionic mechanism of endothelin-1 (ET-1)-induced focal ventricular tachyarrhythmias, the regulation of I(K1) and its main molecular correlates, Kir2.1, Kir2.2 and Kir2.3 channels, by ET-1 was investigated. Native I(K1) in human atrial cardiomyocytes was studied with whole-cell patch clamp. Human endothelin receptors were coexpressed with human Kir2.1, Kir2.2 and Kir2.3 channels in Xenopus oocytes. Currents were measured with a two-microelectrode voltage clamp. In human cardiomyocytes, ET-1 induced a marked inhibition of I(K1) that could be suppressed by the protein kinase C (PKC) inhibitor staurosporine. To investigate the molecular mechanisms underlying this regulation, we studied the coupling of ET(A) receptors to homomeric and heteromeric Kir2.1, Kir2.2 and Kir2.3 channels in the Xenopus oocyte expression system. ET(A) receptors coupled functionally to Kir2.2 and Kir2.3 channels but not to Kir2.1 channels. In Kir2.2 channels lacking functional PKC phosphorylation sites, the inhibitory effect was abolished. The inhibition of Kir2.3 currents could be suppressed by the PKC inhibitors staurosporine and chelerythrine. The coupling of ET(A) receptors to heteromeric Kir2.1/Kir2.2 and Kir2.2/Kir2.3 channels resulted in a strong inhibition of currents comparable with the effect observed in Kir2.2 homomers. Surprisingly, in heteromeric Kir2.1/Kir2.3 channels, no effect was observed. ET-1 inhibits human cardiac I(K1) current via a PKC-mediated phosphorylation of Kir2.2 channel subunits and additional regulatory effects on Kir2.3 channels. This mechanism may contribute to the intrinsic arrhythmogenic potential of ET-1.


Asunto(s)
Endotelina-1/fisiología , Miocitos Cardíacos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Taquicardia/metabolismo , Anciano , Alcaloides/metabolismo , Animales , Benzofenantridinas/metabolismo , Endotelina-1/genética , Endotelina-1/farmacología , Inhibidores Enzimáticos/metabolismo , Atrios Cardíacos/citología , Humanos , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Oocitos/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptor de Endotelina A/metabolismo , Estaurosporina/metabolismo , Xenopus laevis
17.
Nutr Metab Cardiovasc Dis ; 17(9): 666-75, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16926094

RESUMEN

BACKGROUND AND AIMS: Hesperetin belongs to the flavonoid subgroup classified as citrus flavonoids and is the main flavonoid in oranges. A high dietary intake of flavonoids has been associated with a significant reduction in cardiovascular mortality. HERG potassium channels play a major role in cardiac repolarisation and represent the most important pharmacologic target of both antiarrhythmic and proarrhythmic drugs. METHODS AND RESULTS: We used the two-microelectrode voltage-clamp technique to analyse inhibitory effects of hesperetin on hERG potassium channels heterologously expressed in Xenopus oocytes. Hesperetin blocked hERG potassium channels in a concentration dependent manner. Onset of block was fast and completely reversible upon wash-out. There was no significant effect of hesperetin on channel kinetics. Affinity of hesperetin to mutant F656A hERG channel was significantly decreased compared to WT hERG, indicating a binding site in the channel pore cavity. In contrast, affinity of hesperetin to Y652A hERG was not different from the affinity to WT hERG. CONCLUSION: We found an antagonist of cardiac hERG channels that modulates hERG currents by accessing the aromatic pore binding site, particularly amino acid phe-656. Regarding high hesperetin concentrations found in oranges and the increasing consumption of oranges and orange juice in Europe, potential effects of hesperetin on cardiac electrophysiology in vivo deserve further investigation.


Asunto(s)
Aminoácidos Aromáticos/metabolismo , Enfermedades Cardiovasculares/prevención & control , Citrus sinensis/química , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Hesperidina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Aminoácidos Aromáticos/genética , Animales , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Hesperidina/química , Hesperidina/metabolismo , Mutación/fisiología , Oocitos/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/metabolismo , Xenopus
18.
J Biol Methods ; 4(3): e78, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-31453232

RESUMEN

Ventricular tachyarrythmia occurring in ischemic heart disease, dilated/hypertrophic cardiomyopathies or rare monogenic mutations of cardiac ion channels or associated proteins belong to the most frequent causes of sudden cardiac death (SCD). In further decades, next generation sequencing and bioinformatic analysis will become the gold standard of SCD risk stratification. At the moment, Sanger-sequencing is still obligatory in genetic diagnosis. A multiplex polymerase chain reaction (PCR) assay detecting eight SCD mutations in one reaction-tube was developed. To test the general validity of the assay, it was used with 12 patients, who had one or two of the eight mutations (LMNA, p.V256V; SCN5A, p.R1583C; RYR2, p.G1885E; MYH7, V606M; DSG2, p.T335A; KCNJ8, p.S422L; MYBPC, p.E441K; TNNT2, A38V). Thereafter, we tested the multiplex assay in a real diagnostic environment within a high risk family of several past SCD cases. This method allows efficient discrimination of multiple mutations by allele-specific PCR with standard PCR conditions. It relies on obtaining a PCR product specific to the mutation or wildtype-using primers that have the 3'end base complementary to the DNA template site, i.e. a specific primer only permits amplification to take place when its 3'terminal nucleotide matches with its target sequence. The PCR products are further analyzed by length, with Tape Station®(Agilent Technologies, Germany), a high-fidelity capillary chromatography test. The novel multiplex PCR assay strategy could be a good additional test used for SCD risk stratification. Advantages of the test are high velocity and ease of implementation, low price and flexibility of application within cardiomyopathy families for screening purposes.

19.
Naunyn Schmiedebergs Arch Pharmacol ; 390(5): 493-503, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28331977

RESUMEN

The cardiac IK1 current stabilizes the resting membrane potential of cardiomyocytes. Protein kinase A (PKA) induces an inhibition of IK1 current which strongly promotes focal arrhythmogenesis. The molecular mechanisms underlying this regulation have only partially been elucidated yet. Furthermore, the role of A-kinase anchoring proteins (AKAPs) in this regulation has not been examined to date. The objective of this project was to elucidate the molecular mechanisms underlying the inhibition of IK1 by PKA and to identify novel molecular targets for antiarrhythmic therapy downstream ß-adrenoreceptors. Patch clamp and voltage clamp experiments were used to record currents and co-immunoprecipitation, and co-localization experiments were performed to show spatial and functional coupling. Activation of PKA inhibited IK1 current in rat cardiomyocytes. This regulation was markedly attenuated by disrupting PKA-binding to AKAPs with the peptide inhibitor AKAP-IS. We observed functional and spatial coupling of the plasma membrane-associated AKAP15 and AKAP79 to Kir2.1 and Kir2.2 channel subunits, but not to Kir2.3 channels. In contrast, AKAPyotiao had no functional effect on the PKA regulation of Kir channels. AKAP15 and AKAP79 co-immunoprecipitated with and co-localized to Kir2.1 and Kir2.2 channel subunits in ventricular cardiomyocytes. In this study, we provide evidence for coupling of cardiac Kir2.1 and Kir2.2 subunits with the plasma membrane-bound AKAPs 15 and 79. Cardiac membrane-associated AKAPs are a functionally essential part of the regulatory cascade determining IK1 current function and may be novel molecular targets for antiarrhythmic therapy downstream from ß-adrenoreceptors.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Membrana Celular/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Miocitos Cardíacos/enzimología , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas de Anclaje a la Quinasa A/antagonistas & inhibidores , Proteínas de Anclaje a la Quinasa A/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Cricetulus , Activación Enzimática , Activadores de Enzimas/farmacología , Células HEK293 , Humanos , Inmunoprecipitación , Activación del Canal Iónico , Potenciales de la Membrana , Microinyecciones , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Péptidos/farmacología , Canales de Potasio de Rectificación Interna/genética , Unión Proteica , Ratas , Transfección , Xenopus
20.
Circulation ; 111(7): 835-8, 2005 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-15710766

RESUMEN

BACKGROUND: A high intake of dietary flavonoids, which are abundant in fruits, vegetables, tea, and wine, is known to reduce cardiovascular mortality. The effects of flavonoids on cardiac electrophysiology, which theoretically may have both antiarrhythmic and proarrhythmic consequences, have not been studied systematically to date. METHODS AND RESULTS: We screened a broad spectrum of flavonoids for their inhibitory activity on HERG channels by using heterologous expression in Xenopus oocytes. At a concentration of 1 mmol/L, 10 compounds caused a significant inhibition of HERG currents, whereas 11 other flavonoids had no effect. The IC50 value for HERG block by naringenin, the most potent inhibitor, was 102.3 micromol/L in Xenopus oocytes and 36.5 micromol/L in HEK cells. To demonstrate the physiological relevance of these findings, we studied the effects of pink grapefruit juice, which contains large amounts of naringenin glycosides (>1000 micromol/L), in human volunteers. In 10 persons, we observed a peak QTc prolongation of 12.5+/-4.2 ms 5 hours after oral ingestion of 1 L of grapefruit juice. This effect was significant (P=0.02). CONCLUSIONS: We found a significant QTc prolongation by grapefruit juice in healthy volunteers, probably caused by block of HERG channels by flavonoids. These findings reveal new perspectives on the potential for dietary modification of cardiac electrophysiology.


Asunto(s)
Proteínas de Transporte de Catión/antagonistas & inhibidores , Citrus paradisi/química , Electrocardiografía , Flavonoides/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Adulto , Animales , Antiarrítmicos , Bebidas , Canal de Potasio ERG1 , Electrofisiología , Canales de Potasio Éter-A-Go-Go , Femenino , Flavanonas/farmacología , Flavonoides/administración & dosificación , Humanos , Concentración 50 Inhibidora , Masculino , Oocitos , Transducción Genética , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA