Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 321(1): C158-C175, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34038243

RESUMEN

In whole cell patch clamp recordings, it was discovered that normal human adrenal zona glomerulosa (AZG) cells express members of the three major families of K+ channels. Among these are a two-pore (K2P) leak-type and a G protein-coupled, inwardly rectifying (GIRK) channel, both inhibited by peptide hormones that stimulate aldosterone secretion. The K2P current displayed properties identifying it as TREK-1 (KCNK2). This outwardly rectifying current was activated by arachidonic acid and inhibited by angiotensin II (ANG II), adrenocorticotrophic hormone (ACTH), and forskolin. The activation and inhibition of TREK-1 was coupled to AZG cell hyperpolarization and depolarization, respectively. A second K2P channel, TASK-1 (KCNK3), was expressed at a lower density in AZG cells. Human AZG cells also express inwardly rectifying K+ current(s) (KIR) that include quasi-instantaneous and time-dependent components. This is the first report demonstrating the presence of KIR in whole cell recordings from AZG cells of any species. The time-dependent current was selectively inhibited by ANG II, and ACTH, identifying it as a G protein-coupled (GIRK) channel, most likely KIR3.4 (KCNJ5). The quasi-instantaneous KIR current was not inhibited by ANG II or ACTH and may be a separate non-GIRK current. Finally, AZG cells express a voltage-gated, rapidly inactivating K+ current whose properties identified as KV1.4 (KCNA4), a conclusion confirmed by Northern blot. These findings demonstrate that human AZG cells express K2P and GIRK channels whose inhibition by ANG II and ACTH is likely coupled to depolarization-dependent secretion. They further demonstrate that human AZG K+ channels differ fundamentally from the widely adopted rodent models for human aldosterone secretion.


Asunto(s)
Hormona Adrenocorticotrópica/farmacología , Angiotensina II/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canal de Potasio Kv1.4/genética , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Zona Glomerular/metabolismo , Adolescente , Adulto , Aldosterona/biosíntesis , Ácido Araquidónico/farmacología , Autopsia , Niño , Colforsina/farmacología , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Expresión Génica , Humanos , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.4/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Cultivo Primario de Células , Zona Glomerular/citología , Zona Glomerular/efectos de los fármacos
2.
Am J Physiol Heart Circ Physiol ; 321(2): H461-H474, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34270374

RESUMEN

An exaggerated exercise pressor reflex (EPR) causes excessive sympathoexcitation and exercise intolerance during physical activity in the chronic heart failure (CHF) state. Muscle afferent sensitization contributes to the genesis of the exaggerated EPR in CHF. However, the cellular mechanisms underlying muscle afferent sensitization in CHF remain unclear. Considering that voltage-gated potassium (Kv) channels critically regulate afferent neuronal excitability, we examined the potential role of Kv channels in mediating the sensitized EPR in male rats with CHF. Real-time reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting experiments demonstrate that both mRNA and protein expressions of multiple Kv channel isoforms (Kv1.4, Kv3.4, Kv4.2, and Kv4.3) were downregulated in lumbar dorsal root ganglions (DRGs) of CHF rats compared with sham rats. Immunofluorescence data demonstrate significant decreased Kv channel staining in both NF200-positive and IB4-positive lumbar DRG neurons in CHF rats compared with sham rats. Data from patch-clamp experiments demonstrate that the total Kv current, especially IA, was dramatically decreased in medium-sized IB4-negative muscle afferent neurons (a subpopulation containing mostly Aδ neurons) from CHF rats compared with sham rats, indicating a potential functional loss of Kv channels in muscle afferent Aδ neurons. In in vivo experiments, adenoviral overexpression of Kv4.3 in lumbar DRGs for 1 wk attenuated the exaggerated EPR induced by muscle static contraction and the mechanoreflex by passive stretch without affecting the blunted cardiovascular response to hindlimb arterial injection of capsaicin in CHF rats. These data suggest that Kv channel dysfunction in DRGs plays a critical role in mediating the exaggerated EPR and muscle afferent sensitization in CHF.NEW & NOTEWORTHY The primary finding of this manuscript is that voltage-gated potassium (Kv) channel dysfunction in DRGs plays a critical role in mediating the exaggerated EPR and muscle afferent sensitization in chronic heart failure (CHF). We propose that manipulation of Kv channels in DRG neurons could be considered as a potential new approach to reduce the exaggerated sympathoexcitation and to improve exercise intolerance in CHF, which can ultimately facilitate an improved quality of life and reduce mortality.


Asunto(s)
Tolerancia al Ejercicio/fisiología , Ganglios Espinales/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Neuronas Aferentes/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Reflejo Anormal , Vías Aferentes , Animales , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Insuficiencia Cardíaca/metabolismo , Canal de Potasio Kv1.4/metabolismo , Masculino , Músculo Esquelético/inervación , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Reflejo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Canales de Potasio Shaw/metabolismo
3.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-33498463

RESUMEN

DNA methylation is an epigenetic change to the genome that impacts gene activities without modification to the DNA sequence. Alteration in the methylation pattern is a naturally occurring event throughout the human life cycle which may result in the development of diseases such as cancer. In this study, we analyzed methylation data from The Cancer Genome Atlas, under the Lower-Grade Glioma (LGG) and Glioblastoma Multiforme (GBM) projects, to identify methylation markers that exhibit unique changes in DNA methylation pattern along with tumor grade progression, to predict patient survival. We found ten glioma grade-associated Cytosine-phosphate-Guanine (CpG) sites that targeted four genes (SMOC1, KCNA4, SLC25A21, and UPP1) and the methylation pattern is strongly associated with glioma specific molecular alterations, primarily isocitrate dehydrogenase (IDH) mutation and chromosome 1p/19q codeletion. The ten CpG sites collectively distinguished a cohort of diffuse glioma patients with remarkably poor survival probability. Our study highlights genes (KCNA4 and SLC25A21) that were not previously associated with gliomas to have contributed to the poorer patient outcome. These CpG sites can aid glioma tumor progression monitoring and serve as prognostic markers to identify patients diagnosed with less aggressive and malignant gliomas that exhibit similar survival probability to GBM patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Metilación de ADN , Glioma/genética , Neoplasias Encefálicas/patología , Transportadores de Ácidos Dicarboxílicos/genética , Glioma/patología , Humanos , Canal de Potasio Kv1.4/genética , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial/genética , Osteonectina/genética , Pronóstico , Uridina Fosforilasa/genética
4.
Mar Drugs ; 18(8)2020 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-32823677

RESUMEN

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Asunto(s)
Venenos de Moluscos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Animales , Caracol Conus , Activación del Canal Iónico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Canal de Potasio Kv1.6/antagonistas & inhibidores , Canal de Potasio Kv1.6/genética , Canal de Potasio Kv1.6/metabolismo , Potenciales de la Membrana , Oocitos , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/metabolismo , Xenopus laevis
5.
Int J Sports Med ; 40(5): 354-358, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30812034

RESUMEN

The present is an observational study following a genetic epidemiology model using a case-control design. We tested the hypothesis of an association between the prevalence of the genotypic and allelic frequencies distribution of the potassium voltage-gated channel of the shaker related subfamily member 4 gene (KCNA4) rs1323860 (C/T transition) and endurance performance level in Hispanic male marathon runners (MR). The subjects (n=1876) were adult Hispanic male MR. Fast-MR (cases; n=938) were finishers in the top 3rd percentile. Slow MR (controls; n=938) were finishers in the lowest 3rd percentile of their respective age. Genomic DNA was purified from a whole blood sample. Polymerase chain reaction was used to amplify a KCNA4 SNP which consists of a C/T (rs1323860) transition. The observed genotype frequencies, in both Cases and Controls, met Hardy-Weinberg equilibrium (X2, P≥0.05). Genotype and allele frequencies were statistically different (P<0.01) between cases and controls. Odds ratio revealed that the C allele was 1.33 times more likely prevalent in the cases than in the controls (95% CI; 1.17, 1.51; P<0.001). The magnitude of the statistical power for the present study was 0.86. In conclusion, the findings strongly suggest that KCNA4 gene rs1323860 (C/T transition) is auxiliary in the complex phenotype of endurance running performance level in Hispanic male marathon runners.


Asunto(s)
Rendimiento Atlético/fisiología , Canal de Potasio Kv1.4/genética , Resistencia Física , Carrera/fisiología , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Oportunidad Relativa , Fenotipo , Polimorfismo Genético
6.
Biochemistry ; 57(18): 2733-2743, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29616558

RESUMEN

Drugs do not act solely by canonical ligand-receptor binding interactions. Amphiphilic drugs partition into membranes, thereby perturbing bulk lipid bilayer properties and possibly altering the function of membrane proteins. Distinguishing membrane perturbation from more direct protein-ligand interactions is an ongoing challenge in chemical biology. Herein, we present one strategy for doing so, using dimeric 6-bromo-2-mercaptotryptamine (BrMT) and synthetic analogues. BrMT is a chemically unstable marine snail toxin that has unique effects on voltage-gated K+ channel proteins, making it an attractive medicinal chemistry lead. BrMT is amphiphilic and perturbs lipid bilayers, raising the question of whether its action against K+ channels is merely a manifestation of membrane perturbation. To determine whether medicinal chemistry approaches to improve BrMT might be viable, we synthesized BrMT and 11 analogues and determined their activities in parallel assays measuring K+ channel activity and lipid bilayer properties. Structure-activity relationships were determined for modulation of the Kv1.4 channel, bilayer partitioning, and bilayer perturbation. Neither membrane partitioning nor bilayer perturbation correlates with K+ channel modulation. We conclude that BrMT's membrane interactions are not critical for its inhibition of Kv1.4 activation. Further, we found that alkyl or ether linkages can replace the chemically labile disulfide bond in the BrMT pharmacophore, and we identified additional regions of the scaffold that are amenable to chemical modification. Our work demonstrates a strategy for determining if drugs act by specific interactions or bilayer-dependent mechanisms, and chemically stable modulators of Kv1 channels are reported.


Asunto(s)
Canal de Potasio Kv1.4/química , Membrana Dobles de Lípidos/química , Caracoles/química , Triptaminas/química , Secuencia de Aminoácidos , Animales , Humanos , Ligandos , Unión Proteica , Relación Estructura-Actividad , Xenopus laevis
7.
Biochem Biophys Res Commun ; 482(4): 1135-1140, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27916464

RESUMEN

The neurotoxic cone snail peptide µ-GIIIA specifically blocks skeletal muscle voltage-gated sodium (NaV1.4) channels. The related conopeptides µ-PIIIA and µ-SIIIA, however, exhibit a wider activity spectrum by also inhibiting the neuronal NaV channels NaV1.2 and NaV1.7. Here we demonstrate that those µ-conopeptides with a broader target range also antagonize select subtypes of voltage-gated potassium channels of the KV1 family: µ-PIIIA and µ-SIIIA inhibited KV1.1 and KV1.6 channels in the nanomolar range, while being inactive on subtypes KV1.2-1.5 and KV2.1. Construction and electrophysiological evaluation of chimeras between KV1.5 and KV1.6 revealed that these toxins block KV channels involving their pore regions; the subtype specificity is determined in part by the sequence close to the selectivity filter but predominantly by the so-called turret domain, i.e. the extracellular loop connecting the pore with transmembrane segment S5. Conopeptides µ-SIIIA and µ-PIIIA, thus, are not specific for NaV channels, and the known structure of some KV channel subtypes may provide access to structural insight into the molecular interaction between µ-conopeptides and their target channels.


Asunto(s)
Conotoxinas/química , Canal de Potasio Kv.1.1/antagonistas & inhibidores , Canal de Potasio Kv.1.2/antagonistas & inhibidores , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.6/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/química , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Electrofisiología , Células HEK293 , Humanos , Neuronas/metabolismo , Péptidos/química , Dominios Proteicos
8.
Kyobu Geka ; 70(2): 111-113, 2017 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-28174404

RESUMEN

Two 50s female patients with the taste disorder of sweet taste loss and stage IV a type B2 invasive thymoma underwent surgery at our hospital. One patient with myasthenia gravis (MG) developed postoperative myasthenic crisis and recovered by the treatment with plasma apheresis and steroid pulse therapy. Her taste disorder fully recovered together with her MG symptom. The taste disorder of the other patient without MG had persisted for 3 years after the surgery. The taste disorder of sweet taste loss was reported as one of non-motor symptoms caused by MG-related autoimmune mechanisms associated with thymoma, improving with the therapy for MG. Anti-Kv 1.4 antibody was reported to be positive in nearly half patients with the taste disorder and MG and is speculated to affect selectively the sweet taste receptor.


Asunto(s)
Trastornos del Gusto/etiología , Timoma/complicaciones , Neoplasias del Timo/complicaciones , Autoanticuerpos , Femenino , Humanos , Canal de Potasio Kv1.4/inmunología , Metilprednisolona/administración & dosificación , Persona de Mediana Edad , Miastenia Gravis/complicaciones , Miastenia Gravis/terapia , Quimioterapia por Pulso , Trastornos del Gusto/inmunología , Trastornos del Gusto/terapia , Toracoscopía , Timectomía/métodos , Timoma/diagnóstico por imagen , Timoma/terapia , Neoplasias del Timo/diagnóstico por imagen , Neoplasias del Timo/terapia
9.
Am J Physiol Cell Physiol ; 311(2): C255-68, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27281482

RESUMEN

Polyunsaturated fatty acids (PUFAs) modulate voltage-gated K(+) channel inactivation by an unknown site and mechanism. The effects of ω-6 and ω-3 PUFAs were investigated on the heterologously expressed Kv1.4 channel. PUFAs inhibited wild-type Kv1.4 during repetitive pulsing as a result of slowing of recovery from inactivation. In a mutant Kv1.4 channel lacking N-type inactivation, PUFAs reversibly enhanced C-type inactivation (Kd, 15-43 µM). C-type inactivation was affected by extracellular H(+) and K(+) as well as PUFAs and there was an interaction among the three: the effect of PUFAs was reversed during acidosis and abolished on raising K(+) Replacement of two positively charged residues in the extracellular pore (H508 and K532) abolished the effects of the PUFAs (and extracellular H(+) and K(+)) on C-type inactivation but had no effect on the lipoelectric modulation of voltage sensor activation, suggesting two separable interaction sites/mechanisms of action of PUFAs. Charge calculations suggest that the acidic head group of the PUFAs raises the pKa of H508 and this reduces the K(+) occupancy of the selectivity filter, stabilizing the C-type inactivated state.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Canal de Potasio Kv1.4/metabolismo , Animales , Hidrógeno/metabolismo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Xenopus laevis/metabolismo , Xenopus laevis/fisiología
10.
Biol Pharm Bull ; 39(6): 1069-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27251511

RESUMEN

Metergoline is an ergot-derived psychoactive drug that is a ligand for various serotonin and dopamine receptors. Little is known about the effect of metergoline on different types of receptors and ion channels. Potassium channels are the most diverse group of ion channels. Kv1.4, a shaker family K channel alpha subunit, is one of a family of voltage gated K channels that mediates transient and rapid inactivating A-type currents and N-type inactivation. We demonstrated previously that metergoline inhibited the activity of neuronal voltage-dependent Na(+) channels in Xenopus laevis oocytes (Acta Pharmacol. Sin., 35, 2014, Lee et al.). In this study, we sought to elucidate the regulatory effects underlying metergoline-induced human Kv1.4 channel inhibition. We used the two electrode voltage-clamp (TEVC) technique to investigate the effect of metergoline on human Kv1.4 channel currents in Xenopus laevis oocytes expressing human Kv1.4 alpha subunits. Interestingly, metergoline treatment also induced inhibition of peak currents in human Kv1.4 channels in a concentration-dependent manner. The IC50 of peak currents of hKv1.4 currents was 3.6±0.6 µM. These results indicate that metergoline might regulate the human Kv1.4 channel activity that is expressed in X. laevis oocytes. Further, this regulation of potassium currents by metergoline might be one of the pharmacological actions of metergoline-mediated psychoactivity.


Asunto(s)
Antidepresivos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Metergolina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Femenino , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Oocitos/fisiología , Xenopus laevis
11.
Proc Natl Acad Sci U S A ; 110(42): E4036-44, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082096

RESUMEN

Fine-tuned regulation of K(+) channel inactivation enables excitable cells to adjust action potential firing. Fast inactivation present in some K(+) channels is mediated by the distal N-terminal structure (ball) occluding the ion permeation pathway. Here we show that Kv1.4 K(+) channels are potently regulated by intracellular free heme; heme binds to the N-terminal inactivation domain and thereby impairs the inactivation process, thus enhancing the K(+) current with an apparent EC50 value of ∼20 nM. Functional studies on channel mutants and structural investigations on recombinant inactivation ball domain peptides encompassing the first 61 residues of Kv1.4 revealed a heme-responsive binding motif involving Cys13:His16 and a secondary histidine at position 35. Heme binding to the N-terminal inactivation domain induces a conformational constraint that prevents it from reaching its receptor site at the vestibule of the channel pore.


Asunto(s)
Hemo , Canal de Potasio Kv1.4 , Animales , Cristalografía por Rayos X , Hemo/química , Hemo/genética , Hemo/metabolismo , Transporte Iónico/fisiología , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Xenopus laevis
12.
Proc Natl Acad Sci U S A ; 110(41): 16657-62, 2013 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-24067659

RESUMEN

High-conductance Ca(2+)- and voltage-activated K(+) (Slo1 or BK) channels (KCNMA1) play key roles in many physiological processes. The structure of the Slo1 channel has two functional domains, a core consisting of four voltage sensors controlling an ion-conducting pore, and a larger tail that forms an intracellular gating ring thought to confer Ca(2+) and Mg(2+) sensitivity as well as sensitivity to a host of other intracellular factors. Although the modular structure of the Slo1 channel is known, the functional properties of the core and the allosteric interactions between core and tail are poorly understood because it has not been possible to study the core in the absence of the gating ring. To address these questions, we developed constructs that allow functional cores of Slo1 channels to be expressed by replacing the 827-amino acid gating ring with short tails of either 74 or 11 amino acids. Recorded currents from these constructs reveals that the gating ring is not required for either expression or gating of the core. Voltage activation is retained after the gating ring is replaced, but all Ca(2+)- and Mg(2+)-dependent gating is lost. Replacing the gating ring also right-shifts the conductance-voltage relation, decreases mean open-channel and burst duration by about sixfold, and reduces apparent mean single-channel conductance by about 30%. These results show that the gating ring is not required for voltage activation but is required for Ca(2+) and Mg(2+) activation. They also suggest possible actions of the unliganded (passive) gating ring or added short tails on the core.


Asunto(s)
Activación del Canal Iónico/fisiología , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Animales , Calcio/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.4/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Magnesio/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Oligonucleótidos/genética , Oocitos/metabolismo , Técnicas de Placa-Clamp , Péptidos/farmacología , Tetraetilamonio/farmacología , Xenopus
13.
Pak J Pharm Sci ; 29(5): 1513-1517, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27731805

RESUMEN

This research is to explore the effects of traditional Chinese medicine Ginseng-spikenard heart-nourishing capsule on the inactivation of c-type Kv1.4 channels (Kv1.4∆N) in Xenopus laevis oocytes with two-electrode voltageclamp technique. Defolliculated oocytes (stage V-VI) were injected with transcribed cRNAs of ferret Kv1.4δN channels. During recording, oocytes were continuously perfused with ND96 solution (control group) and solution prepared from Ginseng-spikenard heart-nourishing capsule (experimental group). Results found that, at the command potential of +50 mV, the current of experimental group was reduced to 48.33±4.0% of that in control group. The inactivation time constants in control and experimental groups were 2962.56±175.35 ms and 304.13±36.22ms, respectively (P<0.05, n=7). The recovery time of fKv1.4∆N channel after inactivation in control group and experimental groups was 987±68.39 ms and 1734.15±98.45 ms, respectively (P<0.05, n=5). Ginseng-spikenard heart-nourishing capsule can inhibit the Kv1.4δN channel, which may be one of the mechanisms of underlying antiarrhythmia.


Asunto(s)
Antiarrítmicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Animales , Femenino , Hurones , Técnicas de Transferencia de Gen , Cinética , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Potenciales de la Membrana , Oocitos , Xenopus laevis
14.
J Membr Biol ; 248(2): 187-96, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25416425

RESUMEN

Kv1.4 potassium channels are heavily glycosylated proteins involved in shaping action potentials and in neuronal excitability and plasticity. Kv1.4 N354Q, without an N-glycan, exhibited decreased protein stability and trafficking to the cell surface (Watanabe et al. in J Biol Chem 279:8879-8885, 2004). Here we investigated whether the composition of the N-glycan affected Kv1.4 cell surface expression. Kv1.4 proteins carrying N-glycans with different compositions were generated by adding glycosidase inhibitors or using N-glycosylation-deficient mutant cell lines. We found that oligomannose-type, hybrid-type, or incomplete complex-type N-glycans had a negative effect on surface protein expression of Kv1.4 compared with complex-type N-glycans. The decrease in surface protein level of Kv1.4 was mainly due to a reduction in total protein level, induced by altered N-glycan composition. Kv1.4 in CSTP-treated cells carried a unique oligomannose-type N-glycan that contains three glucose residues. This N-glycan had the most negative effect on cell surface expression of Kv1.4. It decreased Kv1.4 surface protein level by a combined mechanism of reducing total protein level and increasing ER-retention. Our data suggest that composition of the N-glycan plays an important role in protein stability and trafficking, and a sialylated complex-type N-glycan promoted high cell surface expression of Kv1.4.


Asunto(s)
Membrana Celular/metabolismo , Canal de Potasio Kv1.4/metabolismo , Animales , Células CHO , Cricetulus , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Expresión Génica , Glicósido Hidrolasas/antagonistas & inhibidores , Glicosilación/efectos de los fármacos , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/genética , Manosa/química , Manosa/metabolismo , Mutación , Polisacáridos/química , Polisacáridos/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Transporte de Proteínas
15.
BMC Neurosci ; 16: 30, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25940378

RESUMEN

BACKGROUND: High titers of lentiviral vectors are required for the efficient transduction of a gene of interest. During preparation of lentiviral the vectors, the protein of interest is inevitably expressed in the viral vector-producing cells. This expression may affect the production of the lentiviral vector. METHODS: We prepared lentiviral vectors expressing inwardly rectifying potassium channel (Lv-Kir2.1), its dominant-negative form (Lv-Kir-DN), and other K(+) channels, using the ubiquitously active ß-actin and neuron-specific synapsin I promoters. RESULTS: The titer of Lv-Kir-DN was higher than that of Lv-Kir2.1, suggesting a negative effect of induced K(+) currents on viral titer. We then blocked Kir2.1 currents with the selective blocker Ba(2+) during Lv-Kir2.1 production, and obtained about a 5-fold increase in the titer. Higher extracellular K(+) concentrations increased the titer of Lv-Kir2.1 about 9-fold. With a synapsin I promoter Ba(2+) increased the titer because of the moderate expression of Kir2.1 channel. Channel blockade also increased the titers of the lentivirus expressing Kv1.4 and TREK channels, but not HERG. The increase in titer correlated with the K(+) currents generated by the channels expressed. CONCLUSION: In the production of lentivirus expressing K(+) channels, titers are increased by blocking K(+) currents in the virus-producing cells. This identifies a crucial issue in the production of viruses expressing membrane channels, and should facilitate basic and gene therapeutic research on channelopathies.


Asunto(s)
Vectores Genéticos , Lentivirus/genética , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Bario/farmacología , Cationes Bivalentes/farmacología , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Vectores Genéticos/metabolismo , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Ratas Sprague-Dawley , Sinapsinas/genética , Sinapsinas/metabolismo , Transfección , Carga Viral
16.
Am J Physiol Regul Integr Comp Physiol ; 308(1): R18-27, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25411361

RESUMEN

Psychological disorders are prevalent in patients with inflammatory bowel disease; the underlying mechanisms remain unknown. We tested the hypothesis that ulcerative colitis-like inflammation induced by dextran sodium sulfate (DSS) exacerbates the ongoing spontaneous activity in colon-projecting afferent neurons that induces abdominal discomfort and anxiety, and depressive-like behaviors in rats. In this study, we used the conditioned place preference and standard tests for anxiety- and depression-like behaviors. DSS rats developed anxiety- and depression-like behaviors 10 to 20 days after the start of inflammation. Single-fiber recordings showed an increase in the frequency of spontaneous activity in L6-S1 dorsal root ganglion (DRG) roots. Prolonged desensitization of transient receptor potential vanilloid 1 (TRPV1)-expressing colonic afferents by resiniferatoxin (RTX) suppressed the spontaneous activity, as well as the anxiety- and depressive-like behaviors. Reduction in spontaneous activity in colon afferents by intracolonic administration of lidocaine produced robust conditioned place preference (CPP) in DSS rats, but not in control rats. Patch-clamp studies demonstrated a significant decrease in the resting membrane potential, lower rheobase, and sensitization of colon-projecting L6-S1 DRG neurons to generate trains of action potentials in response to current injection in DSS rats. DSS inflammation upregulated the mRNA levels of transient receptor potential ankyrin 1 and TRPV1 channels and downregulated that of Kv1.1 and Kv1.4 channels. Ulcerative colitis-like inflammation in rats induces anxiety- and depression-like behaviors, as well as ongoing abdominal discomfort by exacerbating the spontaneous activity in the colon-projecting afferent neurons. Alterations in the expression of voltage- and ligand-gated channels are associated with the induction of mood disorders following colon inflammation.


Asunto(s)
Dolor Abdominal/etiología , Ansiedad/etiología , Conducta Animal , Colitis Ulcerosa/complicaciones , Colon/inervación , Depresión/etiología , Dolor Abdominal/tratamiento farmacológico , Dolor Abdominal/metabolismo , Dolor Abdominal/fisiopatología , Dolor Abdominal/psicología , Potenciales de Acción , Anestésicos Locales/farmacología , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/prevención & control , Ansiedad/psicología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/fisiopatología , Colitis Ulcerosa/psicología , Condicionamiento Psicológico , Depresión/metabolismo , Depresión/fisiopatología , Depresión/prevención & control , Depresión/psicología , Sulfato de Dextran , Modelos Animales de Enfermedad , Diterpenos/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Lidocaína/farmacología , ARN Mensajero/metabolismo , Ratas , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Factores de Tiempo
17.
Biochim Biophys Acta ; 1828(3): 990-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23196347

RESUMEN

Rapid inactivation of voltage-gated potassium channel plays an important role in shaping the electrical signaling in neurons and other excitable cells. N-type ("ball and chain") inactivation, as the most extensively studied inactivation model, is assumed to be the inactivation mechanism of Kv1.4 channel. The inactivation ball inactivates the channel by interacting with the hydrophobic wall of inner pore and occluding it. Recently, we have proved that the electrostatic interaction between two charged segments in the NH(2)-termainal plays an important role through promoting the inactivation process of the Kv1.4 channel. This study investigates the effect of inserting negatively or positively charged short peptides at NH(2)-terminal on the inactivation of Kv1.4 channel. The results that inserting negatively-charged peptide (either myc or D-peptide) at different sites of NH(2)-terminal, deceleraes inactivation process of Kv1.4 channel to a different extent with inserting site changing and that the mutant Kv1.4-D50 exhibits a more slower inactivation rate than Kv1.4-K50 further identified the role of electrostatic interactions in the "ball and chain" inactivation mechanism.


Asunto(s)
Canal de Potasio Kv1.4/química , Péptidos/química , Secuencia de Aminoácidos , Animales , Biofisica/métodos , Células CHO , Cricetinae , Electrofisiología/métodos , Proteínas Fluorescentes Verdes/metabolismo , Iones , Canal de Potasio Kv1.4/metabolismo , Potenciales de la Membrana , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/química , Homología de Secuencia de Aminoácido , Electricidad Estática
18.
Pflugers Arch ; 466(11): 2153-65, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24463703

RESUMEN

Neuropathic pain and pain arising from local inflammation are characterized by increased release of inflammatory mediators like interleukin-6 (IL-6) by immune cells. The levels of IL-6 is increased in various painfull conditions and correlates with the severity of thermal and mechanical hypersensitivity. Deletion of the IL-6 signal transducer glycoprotein 130 (gp130) reduces inflammation associated with hypersensitivity to thermal and mechanical stimuli. In this study, we show that nociceptor-specific deletion of gp130 alters excitability parameters that are linked to changes in the potassium conductance. In SNS-gp130(-/-) sensory neurons, the resting membrane potential was reduced. Moreover the repolarization speed of the action potential and afterhypolarization was augmented, however, voltage-gated Na(+) and Ca(2+) current were not obviously altered. The main difference between gp130-deficient and control neurons was a significant increase in the conductance of both delayed rectifier as well as A-type potassium currents. Taqman RT-PCR analysis revealed significantly higher levels of Kcna4 mRNA, encoding A-type Kv1.4 potassium channel, in neuron cultures from SNS-gp130(-/-) versus control mice, which may account for the electrophysiological data. No difference in other voltage-gated ion channel mRNAs was observed. The present data show for the first time increased A-type K(+) currents and expression of voltage-gated potassium channel Kcna4 (Kv1.4) in SNS-gp130(-/-) nociceptors. This suggests that gp130 acts as a break for the expression of potassium channels and important regulator hub for nociceptor excitability.


Asunto(s)
Glicoproteínas/deficiencia , Canal de Potasio Kv1.4/metabolismo , Nociceptores/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Calcio/metabolismo , Glicoproteínas/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Canal de Potasio Kv1.4/genética , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , ARN Mensajero/genética , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Sodio/metabolismo , Regulación hacia Arriba
19.
Am J Physiol Heart Circ Physiol ; 306(7): H1054-65, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24486512

RESUMEN

In the present study, we examined if and how cardiac ion channels are modified by type 2 diabetes mellitus (T2DM). Subendocardial (Endo) myocytes and subepicardial (Epi) myocytes were isolated from left ventricles of Otsuka-Long-Evans-Tokushima Fatty rats (OLETF) rats, a rat model of T2DM, and Otsuka-Long-Evans-Tokushima (LETO) rats (nondiabetic control rats). Endo and Epi myocytes were used for whole cell patch-clamp recordings and for protein and mRNA analyses. Action potential durations in Endo and Epi myocytes were longer in OLETF rats than in LETO rats, and the difference was larger in Endo myocytes. Steady-state transient outward K+ current (Ito) density was reduced in Endo but not Epi myocytes of OLETF rats compared with LETO rats, although the contribution of the fast component of Ito recovery from inactivation was smaller in both Endo and Epi myocytes of OLETF rats than in LETO rats. Kv4.2 protein was reduced only in Endo myocytes in OLETF rats, although voltage-gated K+ channel-interacting protein 2 (KChIP2) protein levels in both Endo and Epi myocytes were lower in OLETF rats than in LETO rats. Corresponding regional differences in mRNA levels of KChIP2 and Kv4.2 were observed between OLETF and LETO rats. mRNA levels of Iroquois homeobox 5 in Endo myocytes were 53% higher in OLETF rats than in LETO rats. Densities of inward rectifier K+ current and L-type Ca2+ current and mRNA levels of Kv4.3 and Kv1.4 were similar in OLETF and LETO rats. In conclusion, T2DM induces Endo-predominant prolongation of the action potential duration via a reduction of the fast component of Ito recovery from inactivation and reduced steady-state Ito, in which downregulation of Kv4.2 and KChIP2 may be involved. Increased Iroquois homeobox 5 expression may underlie Kv4.2 downregulation in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/etiología , Proteínas de Interacción con los Canales Kv/metabolismo , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Canales de Potasio Shal/metabolismo , Potenciales de Acción , Animales , Glucemia/metabolismo , Canales de Calcio Tipo L/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/sangre , Cardiomiopatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Electrocardiografía , Proteínas de Homeodominio/metabolismo , Cinética , Proteínas de Interacción con los Canales Kv/genética , Canal de Potasio Kv1.4/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas OLETF , Canales de Potasio Shal/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA