Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Gen Physiol Biophys ; 34(4): 383-92, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26001288

RESUMEN

Although several physiological, pathophysiological and regulatory properties of classical inward rectifier K+ current I(K1), G-protein coupled inwardly-rectifying K+ current I(K,ACh) and the small-conductance Ca2+ activated K+ current I(K,Ca) have been identified, quantitative biophysical details remain unclear. Both I(K1) and I(K,ACh) are implicated in atrial fibrillation (AF), and recently also I(K,Ca) has been speculated to be linked with the genesis and sustainability of AF. All these three currents have been shown to be involved in the electrical remodeling in the atria of patients suffering from AF, and it is therefore important to characterize their biophysical properties and compare their relative current contribution in atrial electrophysiology in both sinus rhythm (SR) and AF. The aim of this study is to investigate the contribution of the three potassium currents when subjected to voltage protocols adapted from atrial action potentials recorded in human tissue at 1 and 3 Hz. The current recordings were performed in the HEK-293 heterologous cell system expressing either I(K1), I(K,ACh) or I(K,Ca) to establish the individual contribution of each of these currents during the voltage changes of atrial action potential waveforms. I(K1) primarily contributes to the atrial electrophysiology at the latter part of repolarization and during the diastolic phase, while both I(K,Ca) under high [Ca2+]i and I(K,ACh) contribute relatively most during repolarization.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial/fisiopatología , Calcio/metabolismo , Potenciales de la Membrana , Canales de Potasio de Rectificación Interna/metabolismo , Potasio/metabolismo , Células HEK293 , Frecuencia Cardíaca , Humanos , Activación del Canal Iónico
2.
Am J Physiol Heart Circ Physiol ; 306(8): H1204-12, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24561865

RESUMEN

Brugada syndrome (BrS) is a rare inherited disease that can give rise to ventricular arrhythmia and ultimately sudden cardiac death. Numerous loss-of-function mutations in the cardiac sodium channel Nav1.5 have been associated with BrS. However, few mutations in the auxiliary Navß1-4 subunits have been linked to this disease. Here we investigated differences in expression and function between Navß1 and Navß1b and whether the H162P/Navß1b mutation found in a BrS patient is likely to be the underlying cause of disease. The impact of Navß subunits was investigated by patch-clamp electrophysiology, and the obtained in vitro values were used for subsequent in silico modeling. We found that Navß1b transcripts were expressed at higher levels than Navß1 transcripts in the human heart. Navß1 and Navß1b coexpressed with Nav1.5 induced a negative shift on steady state of activation and inactivation compared with Nav1.5 alone. Furthermore, Navß1b was found to increase the current level when coexpressed with Nav1.5, Navß1b/H162P mutated subunit peak current density was reduced by 48% (-645 ± 151 vs. -334 ± 71 pA/pF), V1/2 steady-state inactivation shifted by -6.7 mV (-70.3 ± 1.5 vs. -77.0 ± 2.8 mV), and time-dependent recovery from inactivation slowed by >50% compared with coexpression with Navß1b wild type. Computer simulations revealed that these electrophysiological changes resulted in a reduction in both action potential amplitude and maximum upstroke velocity. The experimental data thereby indicate that Navß1b/H162P results in reduced sodium channel activity functionally affecting the ventricular action potential. This result is an important replication to support the notion that BrS can be linked to the function of Navß1b and is associated with loss-of-function of the cardiac sodium channel.


Asunto(s)
Síndrome de Brugada/genética , Ventrículos Cardíacos/química , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo , Potenciales de Acción , Animales , Células CHO , Cricetulus , Electrofisiología , Predisposición Genética a la Enfermedad , Ventrículos Cardíacos/fisiopatología , Humanos , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Isoformas de Proteínas , ARN Mensajero/análisis , Canales de Sodio/metabolismo , Transfección
3.
Eur J Hum Genet ; 26(5): 660-668, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29396561

RESUMEN

Atrioventricular nodal reentry tachycardia (AVNRT) is the most common form of regular paroxysmal supraventricular tachycardia. This arrhythmia affects women twice as frequently as men, and is often diagnosed in patients <40 years of age. Familial clustering, early onset of symptoms and lack of structural anomaly indicate involvement of genetic factors in AVNRT pathophysiology. We hypothesized that AVNRT patients have a high prevalence of variants in genes that are highly expressed in the atrioventricular conduction axis of the heart and potentially involved in arrhythmic diseases. Next-generation sequencing of 67 genes was applied to the DNA profile of 298 AVNRT patients and 10 AVNRT family members using HaloPlex Target Enrichment System. In total, we identified 229 variants in 60 genes; 215 missenses, four frame shifts, four codon deletions, three missense and splice sites, two stop-gain variants, and one start-lost variant. Sixty-five of these were not present in the Exome Aggregation Consortium (ExAC) database. Furthermore, we report two AVNRT families with co-segregating variants. Seventy-five of 284 AVNRT patients (26.4%) and three family members to different AVNRT probands had one or more variants in genes affecting the sodium handling. Fifty-four out of 284 AVNRT patients (19.0%) had variants in genes affecting the calcium handling of the heart. We furthermore find a large proportion of variants in the HCN1-4 genes. We did not detect a significant enrichment of rare variants in the tested genes. This could be an indication that AVNRT might be an electrical arrhythmic disease with abnormal sodium and calcium handling.


Asunto(s)
Arritmias Cardíacas/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Canales Iónicos/genética , Taquicardia por Reentrada en el Nodo Atrioventricular/genética , Adulto , Anciano , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/fisiopatología , Electrocardiografía , Exoma/genética , Familia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Caracteres Sexuales , Taquicardia por Reentrada en el Nodo Atrioventricular/fisiopatología
4.
Nat Commun ; 8(1): 1289, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29097701

RESUMEN

Familial growth hormone deficiency provides an opportunity to identify new genetic causes of short stature. Here we combine linkage analysis with whole-genome resequencing in patients with growth hormone deficiency and maternally inherited gingival fibromatosis. We report that patients from three unrelated families harbor either of two missense mutations, c.347G>T p.(Arg116Leu) or c.1106C>T p.(Pro369Leu), in KCNQ1, a gene previously implicated in the long QT interval syndrome. Kcnq1 is expressed in hypothalamic GHRH neurons and pituitary somatotropes. Co-expressing KCNQ1 with the KCNE2 ß-subunit shows that both KCNQ1 mutants increase current levels in patch clamp analyses and are associated with reduced pituitary hormone secretion from AtT-20 cells. In conclusion, our results reveal a role for the KCNQ1 potassium channel in the regulation of human growth, and show that growth hormone deficiency associated with maternally inherited gingival fibromatosis is an allelic disorder with cardiac arrhythmia syndromes caused by KCNQ1 mutations.


Asunto(s)
Fibromatosis Gingival/genética , Hormona de Crecimiento Humana/deficiencia , Canal de Potasio KCNQ1/genética , Mutación Missense , Adolescente , Hormona Adrenocorticotrópica/metabolismo , Adulto , Alelos , Sustitución de Aminoácidos , Animales , Arritmias Cardíacas/genética , Niño , Preescolar , Femenino , Fibromatosis Gingival/metabolismo , Humanos , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/metabolismo , Masculino , Herencia Materna/genética , Ratones , Persona de Mediana Edad , Modelos Moleculares , Linaje , Mapas de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Adulto Joven
5.
Physiol Rep ; 4(8)2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27117805

RESUMEN

The inwardly rectifying potassium current (IK 1) conducted through Kir2.X channels contribute to repolarization of the cardiac action potential and to stabilization of the resting membrane potential in cardiomyocytes. Our aim was to investigate the effect of the recently discovered IK 1 inhibitor PA-6 on action potential repolarization and refractoriness in isolated rat hearts. Transiently transfected HEK-293 cells expressing IK 1 were voltage-clamped with ramp protocols. Langendorff-perfused heart experiments were performed on male Sprague-Dawley rats, effective refractory period, Wenckebach cycle length, and ventricular effective refractory period were determined following 200 nmol/L PA-6 perfusion. 200 nmol/L PA-6 resulted in a significant time-latency in drug effect on the IK 1 current expressed in HEK-293 cells, giving rise to a maximal effect at 20 min. In the Langendorff-perfused heart experiments, PA-6 prolonged the ventricular action potential duration at 90% repolarization (from 41.8 ± 6.5 msec to 72.6 ± 21.1 msec, 74% compared to baseline, P < 0.01, n = 6). In parallel, PA-6 significantly prolonged the ventricular effective refractory period compared to baseline (from 34.8 ± 4.6 msec to 58.1 ± 14.7 msec, 67%, P < 0.01, n = 6). PA-6 increased the short-term beat-to-beat variability and ventricular fibrillation was observed in two of six hearts. Neither atrial ERP nor duration of atrial fibrillation was altered following PA-6 application. The results show that pharmacological inhibition of cardiac IK 1 affects ventricular action potential repolarization and refractoriness and increases the risk of ventricular arrhythmia in isolated rat hearts.


Asunto(s)
Potenciales de Acción/fisiología , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Pentamidina/análogos & derivados , Canales de Potasio/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Células HEK293 , Humanos , Preparación de Corazón Aislado , Masculino , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transfección
6.
Channels (Austin) ; 9(3): 121-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26043299

RESUMEN

The voltage-gated Kv1.5 potassium channel, conducting the ultra-rapid rectifier K(+) current (IKur), is regulated through several pathways. Here we investigate if Kv1.5 surface expression is controlled by the 2 kinases PKC and AMPK, using Xenopus oocytes, MDCK cells and atrial derived HL-1 cells. By confocal microscopy combined with electrophysiology we demonstrate that PKC activation reduces Kv1.5 current, through a decrease in membrane expressed channels. AMPK activation was found to decrease the membrane expression in MDCK cells, but not in HL-1 cells and was furthermore shown to be dependent on co-expression of Nedd4-2 in Xenopus oocytes. These results indicate that Kv1.5 channels are regulated by both kinases, although through different molecular mechanisms in different cell systems.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Canal de Potasio Kv1.5/metabolismo , Proteína Quinasa C/metabolismo , Animales , Línea Celular Tumoral , Perros , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Femenino , Células de Riñón Canino Madin Darby , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Xenopus , Xenopus laevis
7.
PLoS One ; 9(2): e89629, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586922

RESUMEN

Angiotensin II (Ang II) type 1 (AT1) receptor is known to mediate a variety of physiological actions of Ang II including autophagy. However, the role of AT1 receptor in cardiomyocyte autophagy triggered by mechanical stress still remains elusive. The aim of this study was therefore to examine whether and how AT1 receptor participates in cardiomyocyte autophagy induced by mechanical stresses. A 48-hour mechanical stretch and a 4-week transverse aorta constriction (TAC) were imposed to cultured cardiomyocytes of neonatal rats and adult male C57B/L6 mice, respectively, to induce cardiomyocyte hypertrophy prior to the assessment of cardiomyocyte autophagy using LC3b-II. Losartan, an AT1 receptor blocker, but not PD123319, the AT2 inhibitor, was found to significantly reduce mechanical stretch-induced LC3b-II upregulation. Moreover, inhibition of p38MAP kinase attenuated not only mechanical stretch-induced cardiomyocyte hypertrophy but also autophagy. To the contrary, inhibition of ERK and JNK suppressed cardiac hypertrophy but not autophagy. Intriguingly, mechanical stretch-induced autophagy was significantly inhibited by Losartan in the absence of Ang II. Taken together, our results indicate that mechanical stress triggers cardiomyocyte autophagy through AT1 receptor-mediated activation of p38MAP kinase independently of Ang II.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II/fisiología , Autofagia , Miocitos Cardíacos/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Fenómenos Biomecánicos , Células COS , Cardiomegalia/enzimología , Cardiomegalia/patología , Células Cultivadas , Chlorocebus aethiops , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA