Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Alzheimers Dis ; 92(4): 1241-1256, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36872774

RESUMEN

BACKGROUND: Amyloid-ß protein precursor (AßPP) is enriched in neurons. However, the mechanism underlying AßPP regulation of neuronal activity is poorly understood. Potassium channels are critically involved in neuronal excitability. In hippocampus, A-type potassium channels are highly expressed and involved in determining neuronal spiking. OBJECTIVE: We explored hippocampal local field potential (LFP) and spiking in the presence and absence of AßPP, and the potential involvement of an A-type potassium channel. METHODS: We used in vivo extracellular recording and whole-cell patch-clamp recording to determine neuronal activity, current density of A-type potassium currents, and western blot to detect changes in related protein levels. RESULTS: Abnormal LFP was observed in AßPP-/- mice, including reduced beta and gamma power, and increased epsilon and ripple power. The firing rate of glutamatergic neurons reduced significantly, in line with an increased action potential rheobase. Given that A-type potassium channels regulate neuronal firing, we measured the protein levels and function of two major A-type potassium channels and found that the post-transcriptional level of Kv1.4, but not Kv4.2, was significantly increased in the AßPP-/- mice. This resulted in a marked increase in the peak time of A-type transient outward potassium currents in both glutamatergic and gamma-aminobutyric acid-ergic (GABAergic) neurons. Furthermore, a mechanistic experiment using human embryonic kidney 293 (HEK293) cells revealed that the AßPP deficiency-induced increase in Kv1.4 may not involve protein-protein interaction between AßPP and Kv1.4. CONCLUSION: This study suggests that AßPP modulates neuronal firing and oscillatory activity in the hippocampus, and Kv1.4 may be involved in mediating the modulation.


Asunto(s)
Precursor de Proteína beta-Amiloide , Canal de Potasio Kv1.4 , Canales de Potasio , Animales , Humanos , Ratones , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Células HEK293 , Hipocampo/metabolismo , Potasio , Canales de Potasio/metabolismo , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo
2.
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
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.
Pain ; 160(8): 1876-1882, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31335655

RESUMEN

Chronic muscle pain is a prominent symptom of the hand-arm vibration syndrome (HAVS), an occupational disease induced by exposure to vibrating power tools, but the underlying mechanism remains unknown. We evaluated the hypothesis that vibration induces an interleukin 6 (IL-6)-mediated downregulation of the potassium voltage-gated channel subfamily A member 4 (KV1.4) in nociceptors leading to muscle pain. Adult male rats were submitted to a protocol of mechanical vibration of the right hind limb. Twenty-four hours after vibration, muscle hyperalgesia was observed, concomitant to increased levels of IL-6 in the gastrocnemius muscle and decreased expression of KV1.4 in the dorsal root ganglia. Local injection of neutralizing antibodies against IL-6 attenuated the muscle hyperalgesia induced by vibration, whereas antisense knockdown of this channel in the dorsal root ganglia mimicked the muscle hyperalgesia observed in the model of HAVS. Finally, knockdown of the IL-6 receptor signaling subunit glycoprotein 130 (gp130) attenuated both vibration-induced muscle hyperalgesia and downregulation of KV1.4. These results support the hypothesis that IL-6 plays a central role in the induction of muscle pain in HAVS. This likely occurs through intracellular signaling downstream to the IL-6 receptor subunit gp130, which decreases the expression of KV1.4 in nociceptors.


Asunto(s)
Ganglios Espinales/metabolismo , Síndrome por Vibración de la Mano y el Brazo/metabolismo , Hiperalgesia/metabolismo , Interleucina-6/metabolismo , Canal de Potasio Kv1.4/metabolismo , Músculo Esquelético/metabolismo , Dolor Musculoesquelético/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Síndrome por Vibración de la Mano y el Brazo/genética , Interleucina-6/genética , Canal de Potasio Kv1.4/genética , Masculino , Dolor Musculoesquelético/genética , Ratas , Ratas Sprague-Dawley , Vibración
6.
Nat Rev Dis Primers ; 5(1): 30, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31048702

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease caused by antibodies against the acetylcholine receptor (AChR), muscle-specific kinase (MuSK) or other AChR-related proteins in the postsynaptic muscle membrane. Localized or general muscle weakness is the predominant symptom and is induced by the antibodies. Patients are grouped according to the presence of antibodies, symptoms, age at onset and thymus pathology. Diagnosis is straightforward in most patients with typical symptoms and a positive antibody test, although a detailed clinical and neurophysiological examination is important in antibody-negative patients. MG therapy should be ambitious and aim for clinical remission or only mild symptoms with near-normal function and quality of life. Treatment should be based on MG subgroup and includes symptomatic treatment using acetylcholinesterase inhibitors, thymectomy and immunotherapy. Intravenous immunoglobulin and plasma exchange are fast-acting treatments used for disease exacerbations, and intensive care is necessary during exacerbations with respiratory failure. Comorbidity is frequent, particularly in elderly patients. Active physical training should be encouraged.


Asunto(s)
Miastenia Gravis/diagnóstico , Miastenia Gravis/terapia , Acetilcolinesterasa/genética , Acetilcolinesterasa/fisiología , Corticoesteroides/uso terapéutico , Agrina/genética , Agrina/fisiología , Antiinflamatorios no Esteroideos/uso terapéutico , Autoanticuerpos/análisis , Autoanticuerpos/sangre , Biomarcadores/análisis , Biomarcadores/sangre , Blefaroptosis/etiología , Colágeno/genética , Colágeno/fisiología , Cortactina/genética , Cortactina/fisiología , Electromiografía/métodos , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Miastenia Gravis/fisiopatología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/genética , Receptores Colinérgicos/fisiología , Receptores Nicotínicos/genética , Factores de Riesgo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología
7.
Neurosci Lett ; 704: 159-163, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-30965109

RESUMEN

Background ADAM22 and ADAM23 are transmembrane proteins that bind the secreted synaptic protein LGI1 and associate with Kv1.1/Kv1.4 potassium channels. However, the roles of these proteins in regulated voltage-gated potassium currents are poorly understood. Methods Cultured cells were transfected to express ADAM22, ADAM23, and Kv1.1/Kv1.4. Voltage-gated potassium currents were measured by whole-cell patch-clamp. Immunostaining Kv1.1 with fluorescent antibodies and fluorescently tagged Kv1.1 subunits was used to measure the effects of ADAM proteins on cell-surface and total expression of Kv1.1 channels. LGI1-conditioned media was added to assess the effect on LGI1 on Kv1.1 currents. Results Cells transfected with Kv1.1/Kv1.4 showed voltage-gated potassium currents (Kv1.1 currents). ADAM23 was a powerful negative regulator of Kv1.1 currents and caused decreased surface expression of Kv1.1 subunits. This decrease in current was not mediated by clathrin-dependent endocytosis. LGI1-conditioned media did not affect the negative regulation of Kv1.1 currents by ADAM23. ADAM22 had no significant effect on Kv1.1 currents by itself, but in the presence of LGI1-conditioned media markedly potentiated Kv1.1 currents without changing channel activation kinetics. Conclusions ADAM22 and ADAM23 have opposite effects on Kv1.1 currents. The relative expression of these proteins, and the availability of LGI1 may shape the expression of Kv1.1 currents in different neuronal membrane domains.


Asunto(s)
Proteínas ADAM/metabolismo , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv1.4/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas ADAM/genética , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv1.4/genética , Ratones , Proteínas del Tejido Nervioso/genética
8.
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
9.
Neuropharmacology ; 146: 65-73, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30465811

RESUMEN

Voltage-gated potassium channels (VGKCs) are transmembrane ion channels specific for potassium. Currently there are nine kinds of VGKCs. Kv1.4 is one of shaker-related potassium channels. It is a representative alpha subunit of potassium channels that can inactivate A type-currents, leading to N pattern inactivation. Inactivation of Kv channels plays an important role in shaping electrical signaling properties of neuronal and muscular cells. The shape of N pattern inactivation can be modified by removing the N-terminal (NT) domain which results in non-inactivated currents and C pattern inactivation. In a previous work, we have reported the regulatory effect of metergoline on Kv1.4 and Nav1.2 channel activity. In the present study, we constructed a mutant of deleted 61 residues from NT of Kv1.4 channels (Kv1.4 Δ2-61) and found that it induced an outward peak and steady-state currents We also studied the modulation effect of metergoline on the activity of this Kv1.4 Δ2-61 mutant channel without having the N-terminal quick inactivation domain. Our results revealed that treatment with metergoline inhibited NT deleted Kv1.4 mutant channel activity in a concentration-dependent manner which was reversible. Interestingly, metergoline treatment induced little effects on the outward peak current in the deleted Kv1.4 mutant channel. However, metergoline treatment conspicuously inhibited steady state currents of Kv1.4 Δ2-61 channels with acceleration current mode. The acceleration of steady-state current of deleted Kv1.4 mutant channel occurred in a concentration-dependent manner. This means that metergoline can accelerate C pattern inactivation of Kv1.4 Δ2-61 channel by acting as an open state dependent channel blocker. We also performed site-directed mutations in V561A and K532Y, also known as C-type inactivation sites. V561A, K532Y, and V561A + K532Y substitution mutants significantly attenuated the acceleration effect of metergoline on C pattern inactivation of hKv1.4 channel currents. In docking modeling study, predicted binding residues for metergoline were analyzed for six amino acids. Among them, the K532 residue known as the C-type inactivation site was analyzed to be a major site of action. Then various mutants were constructed. K532 substitution mutant significantly abolished the effect of metergoline on Kv1.4 currents among various mutants whereas other changes had slight inhibitory effects. Furthermore, we found that metergoline had specificity for Kv1.4, but not for Kv1.5 currents. In addition, the A type current in rat neuronal cell was inhibited and accelerated of inactivation. This result further shows that metergoline might interact with Lys532 residue and then accelerate C pattern inactivation of Kv1.4 channels with channel type specificity. Taken together, these results demonstrate the molecular basis involved in the effect of metergoline, an ergot alkaloid, on human Kv1.4 channel, providing a novel interaction ligand.


Asunto(s)
Antidepresivos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Metergolina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Sitios de Unión , Cinética , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Lectinas Tipo C , Modelos Moleculares , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Neuronas/fisiología , Oocitos , Canales de Potasio con Entrada de Voltaje , Ratas , Relación Estructura-Actividad , Xenopus laevis
10.
Sci Rep ; 8(1): 3568, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29476105

RESUMEN

Cortical dysplasia (CD) is a common cause for intractable epilepsy. Hyperactivation of the mechanistic target of rapamycin (mTOR) pathway has been implicated in CD; however, the mechanisms by which mTOR hyperactivation contribute to the epilepsy phenotype remain elusive. Here, we investigated whether constitutive mTOR hyperactivation in the hippocampus is associated with altered voltage-gated ion channel expression in the neuronal subset-specific Pten knockout (NS-Pten KO) mouse model of CD with epilepsy. We found that the protein levels of Kv1.1, but not Kv1.2, Kv1.4, or Kvß2, potassium channel subunits were increased, along with altered Kv1.1 distribution, within the hippocampus of NS-Pten KO mice. The aberrant Kv1.1 protein levels were present in young adult (≥postnatal week 6) but not juvenile (≤postnatal week 4) NS-Pten KO mice. No changes in hippocampal Kv1.1 mRNA levels were found between NS-Pten KO and WT mice. Interestingly, mTOR inhibition with rapamycin treatment at early and late stages of the pathology normalized Kv1.1 protein levels in NS-Pten KO mice to WT levels. Together, these studies demonstrate altered Kv1.1 protein expression in association with mTOR hyperactivation in NS-Pten KO mice and suggest a role for mTOR signaling in the modulation of voltage-gated ion channel expression in this model.


Asunto(s)
Epilepsia/genética , Canal de Potasio Kv.1.1/genética , Malformaciones del Desarrollo Cortical/genética , Fosfohidrolasa PTEN/genética , Serina-Treonina Quinasas TOR/genética , Animales , Modelos Animales de Enfermedad , Epilepsia/complicaciones , Epilepsia/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv1.4/genética , Malformaciones del Desarrollo Cortical/complicaciones , Malformaciones del Desarrollo Cortical/patología , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
11.
Cell Biochem Biophys ; 75(1): 25-33, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28054303

RESUMEN

Different classes of Kv1 potassium channels have different trafficking patterns despite having very similar amino acid sequences. Two amino acids responsible for these differences have been identified in the outer pore turret region of Kv1.1 and Kv1.4. Here we tested a series of substitutions at these two determinants on Kv1.4. All P506 substitutions tested resulted in a significant decrease in surface protein, total protein, and protein half-life, indicating that proline is required at 506 to stabilize protein conformation and increase trafficking to the cell surface. All K533 substitutions tested had no effect on total protein, suggesting that the lysine at 533 is not important for maintaining Kv1.4 protein conformation. However, a basic or long polar amino acid, such as K, R, or Q, at this position favored high surface protein and efficient trafficking of Kv1.4, whereas an acidic or short amino acid, such as D, E, S, L, N, or H, at this position induced partial high endoplasmic reticulum-retention. This intracellular retention was not due to protein misfolding. We propose that these four prolines and four lysines in a Kv1.4 homotetramer might provide a binding site for a putative endoplasmic reticulum-export molecule to ensure high cell surface protein expression of the channel.


Asunto(s)
Canal de Potasio Kv1.4/metabolismo , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Células CHO , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Cricetulus , Semivida , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/genética , Dominios Proteicos , Transporte de Proteínas , Ratas
12.
Phys Rev E ; 96(6-1): 062404, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29347346

RESUMEN

Protein and lipid nanodomains are prevalent on the surface of mammalian cells. In particular, it has been recently recognized that ion channels assemble into surface nanoclusters in the soma of cultured neurons. However, the interactions of these molecules with surface nanodomains display a considerable degree of heterogeneity. Here, we investigate this heterogeneity and develop statistical tools based on the recurrence of individual trajectories to identify subpopulations within ion channels in the neuronal surface. We specifically study the dynamics of the K^{+} channel Kv1.4 and the Na^{+} channel Nav1.6 on the surface of cultured hippocampal neurons at the single-molecule level. We find that both these molecules are expressed in two different forms with distinct kinetics with regards to surface interactions, emphasizing the complex proteomic landscape of the neuronal surface. Further, the tools presented in this work provide new methods for the analysis of membrane nanodomains, transient confinement, and identification of populations within single-particle trajectories.


Asunto(s)
Membrana Celular/metabolismo , Hipocampo/metabolismo , Canal de Potasio Kv1.4/metabolismo , Microscopía Fluorescente/métodos , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuronas/metabolismo , Animales , Células Cultivadas , Hipocampo/citología , Procesamiento de Imagen Asistido por Computador , Canal de Potasio Kv1.4/genética , Microdominios de Membrana/metabolismo , Movimiento (Física) , Canal de Sodio Activado por Voltaje NAV1.6/genética , Neuronas/citología , Proteoma , Ratas , Transfección
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.
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
15.
J Biol Rhythms ; 30(5): 396-407, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26152125

RESUMEN

Neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, display daily rhythms in electrical activity with more depolarized resting potentials and higher firing rates during the day than at night. Although these daily variations in the electrical properties of SCN neurons are required for circadian rhythms in physiology and behavior, the mechanisms linking changes in neuronal excitability to the molecular clock are not known. Recently, we reported that mice deficient for either Kcna4 (Kv1.4(-/-)) or Kcnd2 (Kv4.2(-/-); but not Kcnd3, Kv4.3(-/-)), voltage-gated K(+) (Kv) channel pore-forming subunits that encode subthreshold, rapidly activating, and inactivating K(+) currents (IA), have shortened (0.5 h) circadian periods in SCN firing and in locomotor activity compared with wild-type (WT) mice. In the experiments here, we used a mouse (Per2(Luc)) line engineered with a bioluminescent reporter construct, PERIOD2::LUCIFERASE (PER2::LUC), replacing the endogenous Per2 locus, to test the hypothesis that the loss of Kv1.4- or Kv4.2-encoded IA channels also modifies circadian rhythms in the expression of the clock protein PERIOD2 (PER2). We found that SCN explants from Kv1.4(-/-)Per2(Luc) and Kv4.2(-/-) Per2(Luc), but not Kv4.3(-/-)Per2(Luc), mice have significantly shorter (by approximately 0.5 h) circadian periods in PER2 rhythms, compared with explants from Per2(Luc) mice, revealing that the membrane properties of SCN neurons feedback to regulate clock (PER2) expression. The combined loss of both Kv1.4- and Kv4.2-encoded IA channels in Kv1.4(-/-)/Kv4.2(-/-)Per2(Luc) SCN explants did not result in any further alterations in PER2 rhythms. Interestingly, however, mice lacking both Kv1.4 and Kv4.2 show a striking (approximately 1.8 h) advance in their daily activity onset in a light cycle compared with WT mice, suggesting additional roles for Kv1.4- and Kv4.2-encoded IA channels in controlling the light-dependent responses of neurons within and/or outside of the SCN to regulate circadian phase of daily activity.


Asunto(s)
Ritmo Circadiano/fisiología , Canal de Potasio Kv1.4/fisiología , Proteínas Circadianas Period/metabolismo , Canales de Potasio Shal/fisiología , Núcleo Supraquiasmático/fisiología , Animales , Ritmo Circadiano/genética , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Canal de Potasio Kv1.4/genética , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/genética , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp , Proteínas Circadianas Period/genética , Canales de Potasio Shal/genética , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo , Técnicas de Cultivo de Tejidos
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...