Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118502, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31269418

RESUMEN

This work discusses active and passive electrical properties of transverse (T-)tubules in ventricular cardiomyocytes to understand the physiological roles of T-tubules. T-tubules are invaginations of the lateral membrane that provide a large surface for calcium-handling proteins to facilitate sarcomere shortening. Higher heart rates correlate with higher T-tubular densities in mammalian ventricular cardiomyocytes. We assess ion dynamics in T-tubules and the effects of sodium current in T-tubules on the extracellular potential, which leads to a partial reduction of the sodium current in deep segments of a T-tubule. We moreover reflect on the impact of T-tubules on macroscopic conduction velocity, integrating fundamental principles of action potential propagation and conduction. We also theoretically assess how the conduction velocity is affected by different T-tubular sodium current densities. Lastly, we critically assess literature on ion channel expression to determine whether action potentials can be initiated in T-tubules.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Sarcómeros/metabolismo , Potenciales de Acción/fisiología , Señalización del Calcio/genética , Fenómenos Electromagnéticos , Ventrículos Cardíacos/patología , Humanos , Miocitos Cardíacos/patología , Sarcolema/patología , Sarcómeros/patología , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patología , Sodio/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-27055825

RESUMEN

Ion channels are transmembrane proteins that selectively allow ions to flow across the plasma membrane and play key roles in diverse biological processes. A multitude of diseases, called channelopathies, such as epilepsies, muscle paralysis, pain syndromes, cardiac arrhythmias or hypoglycemia are due to ion channel mutations. A wide corpus of literature is available on ion channels, covering both their functions and their roles in disease. The research community needs to access this data in a user-friendly, yet systematic manner. However, extraction and integration of this increasing amount of data have been proven to be difficult because of the lack of a standardized vocabulary that describes the properties of ion channels at the molecular level. To address this, we have developed Ion Channel ElectroPhysiology Ontology (ICEPO), an ontology that allows one to annotate the electrophysiological parameters of the voltage-gated class of ion channels. This ontology is based on a three-state model of ion channel gating describing the three conformations/states that an ion channel can adopt: closed, open and inactivated. This ontology supports the capture of voltage-gated ion channel electrophysiological data from the literature in a structured manner and thus enables other applications such as querying and reasoning tools. Here, we present ICEPO (ICEPO ftp site:ftp://ftp.nextprot.org/pub/current_release/controlled_vocabularies/), as well as examples of its use.


Asunto(s)
Bases de Datos como Asunto , Electrofisiología , Ontología de Genes , Canales Iónicos/metabolismo , Humanos , Activación del Canal Iónico , Modelos Biológicos , Anotación de Secuencia Molecular , Mutación/genética
3.
Neuroscience ; 227: 370-80, 2012 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-23022218

RESUMEN

Neuronal hyperexcitability following peripheral nerve lesions may stem from altered activity of voltage-gated sodium channels (VGSCs), which gives rise to allodynia or hyperalgesia. In vitro, the ubiquitin ligase Nedd4-2 is a negative regulator of VGSC α-subunits (Na(v)), in particular Na(v)1.7, a key actor in nociceptor excitability. We therefore studied Nedd4-2 in rat nociceptors, its co-expression with Na(v)1.7 and Na(v)1.8, and its regulation in pathology. Adult rats were submitted to the spared nerve injury (SNI) model of neuropathic pain or injected with complete Freund's adjuvant (CFA), a model of inflammatory pain. L4 dorsal root ganglia (DRG) were analyzed in sham-operated animals, seven days after SNI and 48 h after CFA with immunofluorescence and Western blot. We observed Nedd4-2 expression in almost 50% of DRG neurons, mostly small and medium-sized. A preponderant localization is found in the non-peptidergic sub-population. Additionally, 55.7 ± 2.7% and 55.0 ± 3.6% of Nedd4-2-positive cells are co-labeled with Na(v)1.7 and Na(v)1.8 respectively. SNI significantly decreases the proportion of Nedd4-2-positive neurons from 45.9 ± 1.9% to 33.5 ± 0.7% (p<0.01) and the total Nedd4-2 protein to 44% ± 0.13% of its basal level (p<0.01, n=4 animals in each group, mean ± SEM). In contrast, no change in Nedd4-2 was found after peripheral inflammation induced by CFA. These results indicate that Nedd4-2 is present in nociceptive neurons, is downregulated after peripheral nerve injury, and might therefore contribute to the dysregulation of Na(v)s involved in the hyperexcitability associated with peripheral nerve injuries.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ganglios Espinales/patología , Neuralgia/patología , Neuronas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factor de Transcripción Activador 3/metabolismo , Análisis de Varianza , Animales , Recuento de Células , Modelos Animales de Enfermedad , Adyuvante de Freund/farmacología , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Técnicas In Vitro , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Ubiquitina-Proteína Ligasas Nedd4 , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transfección
4.
Br J Pharmacol ; 161(1): 79-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20718741

RESUMEN

BACKGROUND AND PURPOSE: Myocardial ischaemia is associated with perturbations of electrophysiological profile of cardiac myocytes. The persistent sodium current (I(Nap)) is one of the major contributors to ischaemic arrhythmias and appears as an attractive therapeutic target. We investigated the effects of F 15845, a new anti-anginal drug on I(Nap) and in integrative models of I(Nap)-induced arrhythmias. EXPERIMENTAL APPROACH: Sodium current was investigated using patch clamp technique on wild-type and DeltaKPQ-mutated hNav1.5 channels transfected in HEK293 cells. Effects of F 15845 on action potentials (APs) were studied by the glass microelectrode technique and its anti-arrhythmic activities were investigated in ischaemia- and aconitine-induced arrhythmias in the rat. KEY RESULTS: We demonstrated that F 15845 is a potent blocker of I(Nap) acting from the extracellular side of the channel. Blockade of I(Nap) was voltage dependent and characterized by an almost pure tonic block. F 15845 shortened AP from rabbit Purkinje fibres, confirming its lack of pro-arrhythmic activity, and prevented AP lengthening induced by the I(Nap) activator veratridine. F 15845 did not affect APs from rabbit atria and guinea pig papillary muscle where I(Nap) is not functional, confirming its inability to affect other cardiac ionic currents. F 15845 was effective at preventing fatal ventricular fibrillation and ventricular tachycardia during coronary ligation without modifying heart rate and blood pressure, and dose dependently increased the dose threshold of aconitine required to induce ventricular arrhythmias. CONCLUSIONS AND IMPLICATIONS: F 15845, a novel anti-anginal drug targeting I(Nap), demonstrates new anti-arrhythmic properties which may be of therapeutic benefit against ischaemia-induced arrhythmias.


Asunto(s)
Arritmias Cardíacas/prevención & control , Benzotiepinas/farmacología , Isquemia Miocárdica/complicaciones , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Aconitina , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/metabolismo , Línea Celular , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Humanos , Masculino , Técnicas de Placa-Clamp , Ramos Subendocárdicos/efectos de los fármacos , Ramos Subendocárdicos/fisiología , Conejos , Ratas , Ratas Sprague-Dawley , Porcinos , Veratridina/farmacología
5.
Can J Cardiol ; 25(3): 156-60, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19279983

RESUMEN

The aim of the present study was to identify the molecular mechanism behind ventricular tachycardia in a patient with Brugada syndrome. Arrhythmias in patients with Brugada syndrome often occur during sleep. However, a 28-year-old man with no previously documented arrhythmia or syncope who experienced shortness of breath and chest pain during agitation is described. An electrocardiogram revealed monomorphic ventricular tachycardia; after he was converted to nodal rhythm, he spontaneously went into sinus rhythm, and showed classic Brugada changes with coved ST elevation in leads V(1) to V(2). Mutation analysis of SCN5A revealed a novel mutation, 3480 deletion T frame shift mutation, resulting in premature truncation of the protein. Heterologous expression of this truncated protein in human embryonic kidney 293 cells showed a markedly reduced protein expression level. By performing whole-cell patch clamp experiments using human embryonic kidney 293 cells transfected with the mutated SCN5A, no current could be recorded. Hence, the results suggest that the patient suffered from haploinsufficiency of Na(v)1.5, and that this mutation was the cause of his Brugada syndrome.


Asunto(s)
Síndrome de Brugada/genética , Deleción Cromosómica , Mutación del Sistema de Lectura , Proteínas Musculares/genética , Canales de Sodio/genética , Taquicardia Ventricular/genética , Adulto , Electrocardiografía , Humanos , Masculino , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Linaje , Polimorfismo Conformacional Retorcido-Simple , Transfección
6.
Neurology ; 71(21): 1669-75, 2008 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-19015483

RESUMEN

BACKGROUND: Mutations in SCN4A may lead to myotonia. METHODS: Presentation of a large family with myotonia, including molecular studies and patch clamp experiments using human embryonic kidney 293 cells expressing wild-type and mutated channels. RESULTS: In a large family with historic data on seven generations and a clear phenotype, including myotonia at movement onset, with worsening by cold temperature, pregnancy, mental stress, and especially after rest after intense physical activity, but without weakness, the phenotype was linked with the muscle sodium channel gene (SCN4A) locus, in which a novel p.I141V mutation was found. This modification is located within the first transmembrane segment of domain I of the Na(v)1.4 alpha subunit, a region where no mutation has been reported so far. Patch clamp experiments revealed a mutation-induced hyperpolarizing shift (-12.9 mV) of the voltage dependence of activation, leading to a significant increase (approximately twofold) of the window current amplitude. In addition, the mutation shifted the voltage dependence of slow inactivation by -8.7 mV and accelerated the entry to this state. CONCLUSIONS: We propose that the gain-of-function alteration in activation leads to the observed myotonic phenotype, whereas the enhanced slow inactivation may prevent depolarization-induced paralysis.


Asunto(s)
Mutación , Miotonía/genética , Canales de Sodio/genética , Línea Celular , Análisis Mutacional de ADN/métodos , Salud de la Familia , Femenino , Humanos , Isoleucina/genética , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Miotonía/patología , Miotonía/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.4 , Subunidades de Proteína/genética , Transfección/métodos , Valina/genética
7.
Arch Mal Coeur Vaiss ; 100(9): 787-93, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18033008

RESUMEN

The main cardiac voltage-gated Na+ channel, Nav1.5, plays a key role in generation of the cardiac action potential (cardiac excitability) and propagation of the electrical impulse in the heart (cardiac conduction). During the past decade, numerous mutations in SCN5A, the gene, encoding Nav1.5, were found in patients with different pathologic cardiac phenotypes such as the congenital long QT syndrome type 3, Brugada syndrome, and progressive cardiac conduction defect (or Lenègre-Lev disease). These mutations define a sub-group of Nav1.5 / SCN5A-related cardiac channelopathies. Recent works have suggested that Nav1.5 is part of several multi-protein complexes located in different membrane compartments of the cardiac cells. In some instances, the genes of these regulatory proteins were also found to be mutated in patients with inherited forms of cardiac arrhythmias. The proteins that associate with Nav1.5, and form these complexes, can be classified as 1) anchoring/adaptor proteins, 2) enzymes interacting with and modifying the channel, and 3) proteins modulating the biophysical properties of Nav1.5 upon binding. The purpose of this short article is to review the proposed roles of these interactions. These recent observations indicate that the expression level, cellular localization, and activity of Nav1.5 are finely regulated by complex molecular mechanisms that we are only starting to elucidate.


Asunto(s)
Proteínas Musculares/fisiología , Canales de Sodio/fisiología , Animales , Fenómenos Biofísicos , Biofisica , Enzimas/fisiología , Humanos , Proteínas Musculares/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Canales de Sodio/genética
8.
Clin Pharmacol Ther ; 81(5): 719-28, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17329992

RESUMEN

Methadone inhibits the cardiac potassium channel hERG and can cause a prolonged QT interval. Methadone is chiral but its therapeutic activity is mainly due to (R)-methadone. Whole-cell patch-clamp experiments using cells expressing hERG showed that (S)-methadone blocked the hERG current 3.5-fold more potently than (R)-methadone (IC50s (half-maximal inhibitory concentrations) at 37 degrees C: 2 and 7 microM). As CYP2B6 slow metabolizer (SM) status results in a reduced ability to metabolize (S)-methadone, electrocardiograms, CYP2B6 genotypes, and (R)- and (S)-methadone plasma concentrations were obtained for 179 patients receiving (R,S)-methadone. The mean heart-rate-corrected QT (QTc) was higher in CYP2B6 SMs (*6/*6 genotype; 439+/-25 ms; n=11) than in extensive metabolizers (non *6/*6; 421+/-25 ms; n=168; P=0.017). CYP2B6 SM status was associated with an increased risk of prolonged QTc (odds ratio=4.5, 95% confidence interval=1.2-17.7; P=0.03). This study reports the first genetic factor implicated in methadone metabolism that may increase the risk of cardiac arrhythmias and sudden death. This risk could be reduced by the administration of (R)-methadone.


Asunto(s)
Analgésicos Opioides/farmacología , Hidrocarburo de Aril Hidroxilasas/metabolismo , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/genética , Metadona/farmacología , Oxidorreductasas N-Desmetilantes/metabolismo , Bloqueadores de los Canales de Potasio , Adulto , Alelos , Analgésicos Opioides/sangre , Analgésicos Opioides/química , Citocromo P-450 CYP2B6 , ADN Complementario/biosíntesis , ADN Complementario/genética , Canal de Potasio ERG1 , Electrocardiografía/efectos de los fármacos , Femenino , Genotipo , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Cinética , Síndrome de QT Prolongado/fisiopatología , Masculino , Metadona/sangre , Metadona/química , Persona de Mediana Edad , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estereoisomerismo
9.
J Biol Chem ; 276(33): 30623-30, 2001 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-11410597

RESUMEN

Defects of the SCN5A gene encoding the cardiac sodium channel alpha-subunit are associated with both the long QT-3 (LQT-3) subtype of long-QT syndrome and Brugada syndrome (BrS). One previously described SCN5A mutation (1795insD) in the C terminus results in a clinical phenotype combining QT prolongation and ST segment elevation, indicating a close interrelationship between the two disorders. Here we provide additional evidence that these two disorders are closely related. We report the analysis of two novel mutations on the same codon, Y1795C (LQT-3) and Y1795H (BrS), expressed in HEK 293 cells and characterized using whole-cell patch clamp procedures. We find marked and opposing effects on channel gating consistent with activity associated with the cellular basis of each clinical disorder. Y1795H speeds and Y1795C slows the onset of inactivation. The Y1795H, but not the Y1795C, mutation causes a marked negative shift in the voltage dependence of inactivation, and neither mutation affects the kinetics of the recovery from inactivation. Interestingly, both mutations increase the expression of sustained Na+ channel activity compared with wild type (WT) channels, although this effect is most pronounced for the Y1795C mutation, and both mutations promote entrance into an intermediate or a slowly developing inactivated state. These data confirm the key role of the C-terminal tail of the cardiac Na+ channel in the control of channel gating, illustrate how subtle changes in channel biophysics can have significant and distinct effects in human disease, and, additionally, provide further evidence of the close interrelationship between BrS and LQT-3 at the molecular level.


Asunto(s)
Bloqueo Cardíaco/genética , Síndrome de QT Prolongado/genética , Mutación , Canales de Sodio/genética , Canales de Sodio/fisiología , Humanos , Canal de Sodio Activado por Voltaje NAV1.5 , Fenotipo
10.
J Mol Cell Cardiol ; 33(5): 873-82, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11343411

RESUMEN

J. Kurokawa, H. Abriel and R. S. Kass. Molecular Basis of the Delayed Rectifier Current I(Ks)in Heart. Journal of Molecular and Cellular Cardiology (2001) 33, 873-882. Electrical activity underlies the control of the frequency, strength, and duration of contraction of the heart. During the cardiac cycle, a regular rhythmic pattern must be established in time-dependent changes in ionic conductances in order to ensure events that underlie normal cardiac function. This pattern must be tightly regulated by sympathetic nervous activity to ensure a physiologically relevant relationship between diastolic filling and ejection times with variable heart rate. The duration of the ventricular action potential is controlled in part by a slowly activated potassium channel current, I(Ks). The molecular identity of the subunits that comprise the channels conducting this current is important, not only for understanding the fundamental mechanisms that control electrical activity in healthy individuals, but also for understanding the molecular basis of at least one inherited human disease, LQTS-1. This brief review summarizes key points of information regarding the molecular determinants of the activity of these channels, their relationship to human disease, and what is known, and yet to be discovered, about the molecular determinants of the regulation of this channel by sympathetic nervous activity.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Animales , Arritmias Cardíacas/genética , Cadmio/metabolismo , Membrana Celular/metabolismo , Cobayas , Humanos , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Cinética , Síndrome de QT Prolongado/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Factores de Tiempo
11.
Circ Res ; 88(7): 740-5, 2001 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-11304498

RESUMEN

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


Asunto(s)
Arritmias Cardíacas/diagnóstico , Corazón/fisiopatología , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/genética , Canales de Sodio/genética , Adolescente , Sustitución de Aminoácidos , Arritmias Cardíacas/genética , Línea Celular , Secuencia Conservada , Análisis Mutacional de ADN , Electrocardiografía , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Fenotipo , Sodio/metabolismo , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología , Transfección
12.
Circulation ; 103(9): 1303-10, 2001 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-11238277

RESUMEN

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


Asunto(s)
Miocardio/metabolismo , Canales de Sodio/fisiología , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos , Encéfalo/metabolismo , Línea Celular , Electrofisiología , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Miocardio/citología , Subunidades de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Sodio/genética , Canales de Sodio/inmunología
13.
Circulation ; 102(8): 921-5, 2000 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-10952963

RESUMEN

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


Asunto(s)
Antiarrítmicos/farmacología , Síndrome de QT Prolongado/genética , Bloqueadores de los Canales de Sodio , Canales de Sodio/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Flecainida/farmacología , Ligamiento Genético , Humanos , Cinética , Lidocaína/farmacología , Síndrome de QT Prolongado/tratamiento farmacológico , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.5 , Mutación Puntual , Especificidad por Sustrato
14.
Circulation ; 102(5): 584-90, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10920073

RESUMEN

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


Asunto(s)
Síndrome de QT Prolongado/genética , Mutación Puntual , Canales de Sodio/genética , Canales de Sodio/fisiología , Función Ventricular/fisiología , Potenciales de Acción , Calcio/metabolismo , Línea Celular , Membrana Celular/fisiología , Frecuencia Cardíaca , Humanos , Cinética , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Proteínas Recombinantes/metabolismo , Transfección
16.
Kidney Int ; 57(3): 809-15, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10720933

RESUMEN

The epithelial Na+ channel (ENaC) is comprised of three subunits, alpha, beta and gamma, and plays an essential role in Na+ and fluid absorption in the kidney, colon and lung. We had identified proline-rich sequences at the C termini of alpha beta gamma ENaC, which include the sequence PPxY, the PY motif. This sequence in beta or gamma ENaC is deleted or mutated in Liddle's syndrome, a hereditary form of arterial hypertension. Our previous work demonstrated that these PY motifs bind to the WW domains of Nedd4, a ubiquitin protein ligase containing a C2 domain, three or four WW domains and a ubiquitin protein ligase Hect domain. Accordingly, we have recently demonstrated that Nedd4 regulates ENaC function by controlling the number of channels at the cell surface, that this regulation is impaired in ENaC bearing Liddle's syndrome mutations, and that ENaC stability and function are regulated by ubiquitination. The C2 domain is responsible for localizing Nedd4 to the plasma membrane in a Ca(2+)-dependent manner, and in polarized epithelial MDCK cells this localization is primarily apical. In accordance, electrophysiological characterization of ENaC expressed in MDCK cells revealed inhibition of channel activity by elevated intracellular Ca2+ levels. Thus, in response to Ca2+, Nedd4 may be mobilized to the apical membrane via its C2 domain, where it binds ENaC via Nedd4-WW:ENaC-PY motifs' interactions, leading to ubiquitination of the channel by the Nedd4-Hect domain and subsequent channel endocytosis and lysosomal degradation. This process may be at least partially impaired in Liddle's syndrome due to reduced Nedd4 binding, leading to increased retention of ENaC at the cell surface.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Ligasas/fisiología , Canales de Sodio/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinas/metabolismo , Animales , Calcio/fisiología , Proteínas de Unión al Calcio/química , Complejos de Clasificación Endosomal Requeridos para el Transporte , Canales Epiteliales de Sodio , Epitelio/metabolismo , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Ligasas/química , Ubiquitina-Proteína Ligasas Nedd4
17.
FEBS Lett ; 466(2-3): 377-80, 2000 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-10682864

RESUMEN

The cardiac voltage-gated Na+ channel H1, involved in the generation of cardiac action potential, contains a C-terminal PY motif (xPPxY). Since PY motifs are known ligands to WW domains, we investigated their role for H1 regulation and the possible involvement of the WW domain containing ubiquitin-protein ligase Nedd4, taking advantage of the Xenopus oocyte system. Mutation of the PY motif leads to higher peak currents when compared to wild-type channel. Moreover, co-expression of Nedd4 reduced the peak currents, whereas an enzymatically inactive Nedd4 mutant increased them, likely by competing with endogenous Nedd4. The effect of Nedd4 was not observed in the PY motif mutated channel or in the skeletal muscle voltage-gated Na+ channel, which lacks a PY motif. We conclude that H1 may be regulated by Nedd4 depending on WW-PY interaction, and on an active ubiquitination site.


Asunto(s)
Proteínas de Unión al Calcio/genética , Regulación de la Expresión Génica/fisiología , Ligasas , Miocardio/metabolismo , Canales de Sodio/genética , Ubiquitina-Proteína Ligasas , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte , Activación del Canal Iónico , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ubiquitina-Proteína Ligasas Nedd4 , Ratas , Homología de Secuencia de Aminoácido , Canales de Sodio/química , Canales de Sodio/metabolismo , Xenopus , Proteínas de Xenopus
18.
Biochim Biophys Acta ; 1418(1): 85-96, 1999 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-10209213

RESUMEN

The catalytic alpha subunit of the (Na,K)- and (H,K)-ATPases needs to be coexpressed with a beta subunit in order to produce cation transport activity. Although the isoform of the beta subunit is known to influence the functional characteristics of the Na,K pump, the role of the different domains of the beta subunit is not fully understood. We have studied the function of a Na,K pump resulting from the expression of a wild-type alpha subunit with a N-terminally truncated mutant of the beta subunit using the two-electrode voltage clamp and the cut-open oocyte techniques. While the maximal activity, measured as the K+-activated outward current, was not significantly altered, the beta N-terminal truncation induced an ouabain-sensitive conductance in the absence of extracellular K+. The voltage dependence of the ouabain-sensitive charge distribution indicated that in the Na/Na exchange conditions, the E1-E2 conformation equilibrium was shifted towards the E2 conformation, a change resulting from alteration of both the forward and the backward reaction rate. Removal of the intracellular domain of the beta subunit modifies several aspects of the whole enzyme function by a mechanism that must imply the state of the extracellular and/or transmembrane parts of the alpha/beta subunit complex.


Asunto(s)
Membrana Celular/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Potenciales de la Membrana , Oocitos/enzimología , Ouabaína , Técnicas de Placa-Clamp , ATPasa Intercambiadora de Sodio-Potasio/química , Xenopus
19.
J Physiol ; 516 ( Pt 1): 31-43, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10066920

RESUMEN

1. Regulation of the amiloride-sensitive epithelial sodium channel (ENaC) is essential for the control of body sodium homeostasis. The downregulation of the activity of this Na+ channel that occurs when the intracellular Na+ concentration ([Na+]i) is increased is known as feedback inhibition. Although intracellular Na+ is the trigger for this phenomenon, its cellular and molecular mediators are unknown. 2. We used the 'cut-open oocyte' technique to control the composition of the intracellular milieu of Xenopus oocytes expressing rat ENaCs to enable us to test several factors potentially involved in feedback inhibition. 3. The effects of perfusion of the intracellular space were demonstrated by an electromicrographic study and the time course of the intracellular solution exchange was established by observing the effect of intracellular pH: a decrease from pH 7.4 to 6.5 reduced the amiloride-sensitive current by about 40 % within 2 min. 4. Feedback inhibition was observed in non-perfused oocytes when Na+ entry induced a large increase in [Na+]i. Intracellular perfusion prevented feedback regulation even though the [Na+]i was allowed to increase to values above 50 mM. 5. No effects on the amiloride-sensitive current were observed after changes in the concentration of Na+ (from 1 to 50 mM), Ca2+ (from 10 to 1000 nM) or ATP (from nominally free to 1 or 5 mM) in the intracellular perfusate. 6. We conclude that feedback inhibition requires intracellular factors that can be removed by intracellular perfusion. Although a rise in [Na+]i may be the trigger for the feedback inhibition of the ENaC, this effect is not mediated by a direct effect of Na+, Ca2+ or ATP on the ENaC protein.


Asunto(s)
Amilorida/farmacología , Diuréticos/farmacología , Canales de Sodio/metabolismo , Adenosina Trifosfato/farmacología , Animales , Calcio/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Canales Epiteliales de Sodio , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/ultraestructura , Retroalimentación/fisiología , Femenino , Cinética , Microscopía Electrónica , Oocitos/metabolismo , Técnicas de Placa-Clamp , Ratas , Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Canales de Sodio/ultraestructura , Xenopus laevis
20.
J Clin Invest ; 103(5): 667-73, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10074483

RESUMEN

Liddle's syndrome is an inherited form of hypertension linked to mutations in the epithelial Na+ channel (ENaC). ENaC is composed of three subunits (alpha, beta, gamma), each containing a COOH-terminal PY motif (xPPxY). Mutations causing Liddle's syndrome alter or delete the PY motifs of beta- or gamma-ENaC. We recently demonstrated that the ubiquitin-protein ligase Nedd4 binds these PY motifs and that ENaC is regulated by ubiquitination. Here, we investigate, using the Xenopus oocyte system, whether Nedd4 affects ENaC function. Overexpression of wild-type Nedd4, together with ENaC, inhibited channel activity, whereas a catalytically inactive Nedd4 stimulated it, likely by acting as a competitive antagonist to endogenous Nedd4. These effects were dependant on the PY motifs, because no Nedd4-mediated changes in channel activity were observed in ENaC lacking them. The effect of Nedd4 on ENaC missing only one PY motif (of beta-ENaC), as originally described in patients with Liddle's syndrome, was intermediate. Changes were due entirely to alterations in ENaC numbers at the plasma membrane, as determined by surface binding and immunofluorescence. Our results demonstrate that Nedd4 is a negative regulator of ENaC and suggest that the loss of Nedd4 binding sites in ENaC observed in Liddle's syndrome may explain the increase in channel number at the cell surface, increased Na+ reabsorption by the distal nephron, and hence the hypertension.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , Ligasas , Canales de Sodio/metabolismo , Ubiquitina-Proteína Ligasas , Animales , Proteínas de Unión al Calcio/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte , Células Epiteliales/metabolismo , Mutación , Ubiquitina-Proteína Ligasas Nedd4 , Ratas , Canales de Sodio/genética , Síndrome , Xenopus , Proteínas de Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA