Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Oncol Rep ; 45(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34105734

RESUMEN

The aim of the present study was to explore and verify the potential mechanism of seminoma progression. Data on 132 RNA­seq and 156 methylation sites from stage II/III and I seminoma specimens were downloaded from The Cancer Genome Atlas database. An initial filter of |fold­change| >2 and false discovery rate <0.05 were used to identify differentially expressed genes (DEGs) which were associated with differential methylation site genes; these genes were considered potential candidates for further investigation by survival analysis. Potassium voltage­gated channel subfamily C member 1 (KCNC1) expression was verified in seminoma human tissues and three seminoma cell lines. The invasive, proliferative and apoptotic abilities of the human testicular tumor Ntera­2 and normal human testis Hs1.Tes cell lines were assessed following aberrant KCNC1 expression. KCNC1 was identified as a DEG, in which hypermethylation inhibited its expression and it was associated with poor overall survival in patients with seminoma. The present results demonstrated that KCNC1 is negatively correlated with methylation. Due to the abnormal expression of KCNC1 in seminoma cells, it was suggested that KCNC1 could be used as a diagnostic indicator and therapeutic target for the progression of seminoma.


Asunto(s)
Metilación de ADN , Seminoma/genética , Canales de Potasio Shaw/genética , Neoplasias Testiculares/genética , Adulto , Apoptosis/genética , Proliferación Celular/genética , Técnicas de Inactivación de Genes , Humanos , Inmunohistoquímica , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Seminoma/metabolismo , Seminoma/mortalidad , Seminoma/patología , Canales de Potasio Shaw/biosíntesis , Tasa de Supervivencia , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/mortalidad , Neoplasias Testiculares/patología , Transfección
2.
Int J Mol Sci ; 21(19)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003279

RESUMEN

Kv3.1 channel is abundantly expressed in neurons and its dysfunction causes sleep loss, neurodegenerative diseases and depression. Fluoxetine, a serotonin selective reuptake inhibitor commonly used to treat depression, acts also on Kv3.1. To define the relationship between Kv3.1 and serotonin receptors (SR) pharmacological modulation, we showed that 1C11, a serotonergic cell line, expresses different voltage gated potassium (VGK) channels subtypes in the presence (differentiated cells (1C11D)) or absence (not differentiated cells (1C11ND)) of induction. Only Kv1.2 and Kv3.1 transcripts increase even if the level of Kv3.1b transcripts is highest in 1C11D and, after fluoxetine, in 1C11ND but decreases in 1C11D. The Kv3.1 channel protein is expressed in 1C11ND and 1C11D but is enhanced by fluoxetine only in 1C11D. Whole cell measurements confirm that 1C11 cells express (VGK) currents, increasing sequentially as a function of cell development. Moreover, SR 5HT1b is highly expressed in 1C11D but fluoxetine increases the level of transcript in 1C11ND and significantly decreases it in 1C11D. Serotonin dosage shows that fluoxetine at 10 nM blocks serotonin reuptake in 1C11ND but slows down its release when cells are differentiated through a decrease of 5HT1b receptors density. We provide the first experimental evidence that 1C11 expresses Kv3.1b, which confirms its major role during differentiation. Cells respond to the fluoxetine effect by upregulating Kv3.1b expression. On the other hand, the possible relationship between the fluoxetine effect on the kinetics of 5HT1b differentiation and Kv3.1bexpression, would suggest the Kv3.1b channel as a target of an antidepressant drug as well as it was suggested for 5HT1b.


Asunto(s)
Fluoxetina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Canales de Potasio Shaw/genética , Animales , Células CHO , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cricetulus , Depresión/tratamiento farmacológico , Depresión/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Canal de Potasio Kv.1.2/genética , Neuronas Serotoninérgicas/metabolismo , Serotonina/genética , Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
3.
Neuropediatrics ; 51(5): 368-372, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32392612

RESUMEN

Patients with neurofibromatosis type 1 (NF1) have an increased risk for West syndrome (WS), but the underlying mechanisms linking NF1 and WS are unknown. In contrast to other neurocutaneous syndromes, intracerebral abnormalities explaining the course of infantile spasms (IS) are often absent and the seizure outcome is usually favorable. Several studies have investigated a potential genotype-phenotype correlation between NF1 and seizure susceptibility, but an association was not identified. Therefore, we identified three patients with NF1-related WS (NF1-WS) in a cohort of 51 NF1 patients and performed whole-exome sequencing (WES) to identify genetic modifiers. In two NF1 patients with WS and good seizure outcome, we did not identify variants in epilepsy-related genes. However, in a single patient with NF1-WS and transition to drug-resistant epilepsy, we identified a de novo variant in KCNC2 (c.G499T, p.D167Y) coding for Kv3.2 as a previously undescribed potassium channel to be correlated to epilepsy. Electrophysiological studies of the identified KCNC2 variant demonstrated both a strong loss-of-function effect for the current amplitude and a gain-of-function effect for the channel activation recommending a complex network effect. These results suggest that systematic genetic analysis for potentially secondary genetic etiologies in NF1 patients and severe epilepsy presentations should be done.


Asunto(s)
Neurofibromatosis 1/genética , Canales de Potasio Shaw/genética , Espasmos Infantiles/genética , Comorbilidad , Humanos , Lactante , Secuenciación del Exoma
4.
Ann Clin Lab Sci ; 49(4): 457-467, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31471334

RESUMEN

This study aims to explore the regulatory mechanism of hypoxia-inducible factor HIF-1α on Kv3.4. Oral squamous cell carcinoma (OSCC) cell lines SCC3 and CAL27 were used in this study. Western blotting and qRT-PCR methods were used to detect Kv3.4 expression levels in OSCC and their adjacent tissues. The expression changes of Kv3.4 and HIF-1α in a hypoxic environment were detected in cell lines. The stable OSCC cell lines with knockouts of HIF-1α and Kv3.4 were constructed. Transwell and CCK-8 assays were used to detect changes in the invasion, migration and proliferation ability after transfection. Chromatin immunoprecipitation and luciferase reporter gene assays were used to determine the regulatory and binding sites of HIF-1α on Kv3.4. The expression level of Kv3.4 in oral cancer tissue was higher than normal oral epithelium's regular value. The expression level of HIF-1α and Kv3.4 increased under hypoxia. Knocking out HIF-1α and Kv3.4 could reduce the invasion, migration and proliferation of cells. A down regulation of HIF-1α will reduce the Kv3.4 expression level. Overexpressing Kv3.4 after knocking down HIF-1α partially restored the proliferation and invasion of cell lines. Therefore, HIF-1α regulates the invasion, migration and proliferation of oral cancer cells by regulating Kv3.4 expression.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Boca/patología , Canales de Potasio Shaw/metabolismo , Secuencia de Bases , Hipoxia de la Célula/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Neoplasias de la Boca/genética , Invasividad Neoplásica , Regiones Promotoras Genéticas/genética , Unión Proteica , Canales de Potasio Shaw/genética
5.
Am J Physiol Heart Circ Physiol ; 317(3): H496-H504, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31274353

RESUMEN

Accumulating evidence supports that the brain renin-angiotensin system (RAS), including prorenin (PR) and its receptor (PRR), two newly discovered RAS players, contribute to sympathoexcitation in salt-sensitive hypertension. Still, whether PR also contributed to elevated circulating levels of neurohormones such as vasopressin (VP) during salt-sensitive hypertension, and if so, what are the precise underlying mechanisms, remains to be determined. To address these questions, we obtained patch-clamp recordings from hypothalamic magnocellular neurosecretory neurons (MNNs) that synthesize the neurohormones oxytocin and VP in acute hypothalamic slices obtained from sham and deoxycorticosterone acetate (DOCA)-salt-treated hypertensive rats. We found that focal application of PR markedly increased membrane excitability and firing responses in MNNs of DOCA-salt, compared with sham rats. This effect included a shorter latency to spike initiation and increased numbers of spikes in response to depolarizing stimuli and was mediated by a more robust inhibition of A-type K+ channels in DOCA-salt compared with sham rats. On the other hand, the afterhyperpolarizing potential mediated by the activation of Ca2+-dependent K+ channel was not affected by PR. mRNA expression of PRR, VP, and the Kv4.3 K+ channel subunit in the supraoptic nucleus of DOCA-salt hypertensive rats was increased compared with sham rats. Finally, we report a significant decrease of plasma VP levels in neuron-selective PRR knockdown mice treated with DOCA-salt, compared with wild-type DOCA-salt-treated mice. Together, these results support that activation of PRR contributes to increased excitability and firing discharge of MNNs and increased plasma levels of VP in DOCA-salt hypertension.NEW & NOTEWORTHY Our studies support that prorenin (PR) and its receptor (PRR) within the hypothalamus contribute to elevated plasma vasopressin levels in deoxycorticosterone acetate-salt hypertension, in part because of an exacerbated effect of PR on magnocellular neurosecretory neuron excitability; Moreover, our study implicates A-type K+ channels as key underlying molecular targets mediating these effects. Thus, PR/PRR stands as a novel therapeutic target for the treatment of neurohumoral activation in salt-sensitive hypertension.


Asunto(s)
Presión Sanguínea , Hipertensión/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Sistema Renina-Angiotensina , Renina/metabolismo , Vasopresinas/sangre , Animales , Acetato de Desoxicorticosterona , Modelos Animales de Enfermedad , Hipertensión/sangre , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Hipotálamo/fisiopatología , Masculino , Potenciales de la Membrana , Ratones Noqueados , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/metabolismo , Ratas Wistar , Tiempo de Reacción , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Cloruro de Sodio Dietético , Factores de Tiempo , Regulación hacia Arriba
6.
Hear Res ; 365: 77-89, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29773471

RESUMEN

Noise exposure has been shown to produce long-lasting increases in spontaneous activity in central auditory structures in animal models, and similar pathologies are thought to contribute to clinical phenomena such as hyperacusis or tinnitus in humans. Here we demonstrate that multi-unit spontaneous neuronal activity in the inferior colliculus (IC) of mice is significantly elevated four weeks following noise exposure at recording sites with frequency tuning within or near the noise exposure band, and this selective central auditory pathology can be normalised through administration of a novel compound that modulates activity of Kv3 voltage-gated ion channels. The compound had no statistically significant effect on IC spontaneous activity without noise exposure, nor on thresholds or frequency tuning of tone-evoked responses either with or without noise exposure. Administration of the compound produced some reduction in the magnitude of evoked responses to a broadband noise, but unlike effects on spontaneous rates, these effects on evoked responses were not specific to recording sites with frequency tuning within the noise exposure band. Thus, the results suggest that modulators of Kv3 channels can selectively counteract increases in spontaneous activity in the auditory midbrain associated with noise exposure.


Asunto(s)
Estimulación Acústica/métodos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Imidazoles/farmacología , Colículos Inferiores/efectos de los fármacos , Pirimidinas/farmacología , Canales de Potasio Shaw/efectos de los fármacos , Animales , Vías Auditivas/efectos de los fármacos , Vías Auditivas/metabolismo , Umbral Auditivo/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/farmacocinética , Colículos Inferiores/metabolismo , Masculino , Ratones Endogámicos CBA , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Pirimidinas/farmacocinética , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Transducción de Señal/efectos de los fármacos
8.
PLoS One ; 12(5): e0173565, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28467418

RESUMEN

The autosomal dominant spinocerebellar ataxias (SCAs) are a diverse group of neurological disorders anchored by the phenotypes of motor incoordination and cerebellar atrophy. Disease heterogeneity is appreciated through varying comorbidities: dysarthria, dysphagia, oculomotor and/or retinal abnormalities, motor neuron pathology, epilepsy, cognitive impairment, autonomic dysfunction, and psychiatric manifestations. Our study focuses on SCA13, which is caused by several allelic variants in the voltage-gated potassium channel KCNC3 (Kv3.3). We detail the clinical phenotype of four SCA13 kindreds that confirm causation of the KCNC3R423H allele. The heralding features demonstrate congenital onset with non-progressive, neurodevelopmental cerebellar hypoplasia and lifetime improvement in motor and cognitive function that implicate compensatory neural mechanisms. Targeted expression of human KCNC3R423H in Drosophila triggers aberrant wing veins, maldeveloped eyes, and fused ommatidia consistent with the neurodevelopmental presentation of patients. Furthermore, human KCNC3R423H expression in mammalian cells results in altered glycosylation and aberrant retention of the channel in anterograde and/or endosomal vesicles. Confirmation of the absence of plasma membrane targeting was based on the loss of current conductance in cells expressing the mutant channel. Mechanistically, genetic studies in Drosophila, along with cellular and biophysical studies in mammalian systems, demonstrate the dominant negative effect exerted by the mutant on the wild-type (WT) protein, which explains dominant inheritance. We demonstrate that ocular co-expression of KCNC3R423H with Drosophila epidermal growth factor receptor (dEgfr) results in striking rescue of the eye phenotype, whereas KCNC3R423H expression in mammalian cells results in aberrant intracellular retention of human epidermal growth factor receptor (EGFR). Together, these results indicate that the neurodevelopmental consequences of KCNC3R423H may be mediated through indirect effects on EGFR signaling in the developing cerebellum. Our results therefore confirm the KCNC3R423H allele as causative for SCA13, through a dominant negative effect on KCNC3WT and links with EGFR that account for dominant inheritance, congenital onset, and disease pathology.


Asunto(s)
Receptores ErbB/metabolismo , Canales de Potasio Shaw/genética , Degeneraciones Espinocerebelosas/genética , Animales , Células CHO , Cricetinae , Cricetulus , Drosophila melanogaster , Femenino , Humanos , Masculino , Linaje , Transporte de Proteínas
9.
Epilepsia ; 58(4): 543-547, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28166365

RESUMEN

OBJECTIVE: Perampanel (PER) was used in 12 patients with Unverricht-Lundborg disease (ULD) to evaluate its efficacy against myoclonus and seizures. METHODS: We treated 11 patients with EPM1 mutations (6 F, 5 M, aged 13-62 years) and a 43-year-old man with de novo KCNC1 mutation. PER was introduced by 2 mg steps at 2-4 week intervals until 6 mg/day, with a possible dose reduction or dose increase. RESULTS: Ten patients had a clear clinical response of myoclonus, and five were able to reduce concomitant therapy. Improvement was noted sometimes as soon as with 2 mg/day. Epileptic seizures stopped on PER in the six patients who still had experienced generalized tonic-clonic or myoclonic seizures (100%). Some abatement of efficacy on myoclonus was seen in two patients who still retained some benefit. Weight gain was reported in six patients (50%). Psychological and behavioral side-effects were observed in six patients (50%) and led to withdrawal of PER in three cases and dose reduction in three, with abatement of the problems. SIGNIFICANCE: This study provides evidence that for ULD patients, PER may show marked efficacy even in severe cases, particularly against myoclonus, but also against seizures. PER should thus be tried in ULD patients whose seizures are not satisfactorily controlled. Its use is limited because of psychological and behavioral side effects, with higher doses of approximately 6 mg/day or greater likely risk factors.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Piridonas/uso terapéutico , Síndrome de Unverricht-Lundborg/tratamiento farmacológico , Adolescente , Adulto , Electroencefalografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Proteínas de Neoplasias/genética , Nitrilos , Receptores de Superficie Celular/genética , Canales de Potasio Shaw/genética , Resultado del Tratamiento , Síndrome de Unverricht-Lundborg/genética , Adulto Joven
10.
Histopathology ; 69(1): 91-8, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26648458

RESUMEN

AIMS: The expression of the voltage-gated potassium channel Kv3.4 was investigated in both oral squamous cell carcinomas (OSCC) and oral leucoplakias to establish its clinical significance during the development and progression of OSCC. MATERIALS AND METHODS: Tissue specimens from 62 patients with oral leucoplakia were collected prospectively and 100 patients with OSCC who underwent surgical treatment were collected retrospectively, and Kv3.4 expression was analysed by immunohistochemistry. RESULTS: Thirty-nine of 100 tumours exhibited Kv3.4-positive expression, and staining was associated with the degree of differentiation (P = 0.05) but showed no impact on patient prognosis. Abnormal Kv3.4 expression was detected in 16% (7 of 43) hyperplastic lesions and at a significantly higher proportion in oral dysplasias (50%, 8 of 16 cases; P = 0.008), whereas expression was negligible in normal adjacent epithelia. Furthermore, patients carrying Kv3.4-positive lesions exhibited a higher progression risk than those with Kv3.4-negative lesions; however, histology but not Kv3.4 expression predicted oral cancer development significantly in this prospective cohort. CONCLUSION: This study provides original evidence to demonstrate the early occurrence and high prevalence of abnormal Kv3.4 expression in oral leucoplakias. Our results support a role for Kv3.4 potassium channel in OSCC tumorigenesis rather than tumour progression and disease outcome.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Leucoplasia Bucal/metabolismo , Neoplasias de la Boca/metabolismo , Canales de Potasio Shaw/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/diagnóstico , Transformación Celular Neoplásica , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Leucoplasia Bucal/diagnóstico , Masculino , Persona de Mediana Edad , Boca/metabolismo , Boca/patología , Neoplasias de la Boca/diagnóstico , Pronóstico , Estudios Prospectivos , Canales de Potasio Shaw/genética
11.
Nat Commun ; 6: 10173, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26673941

RESUMEN

High-frequency action potential (AP) transmission is essential for rapid information processing in the central nervous system. Voltage-dependent Kv3 channels play an important role in this process thanks to their high activation threshold and fast closure kinetics, which reduce the neuron's refractory period. However, premature Kv3 channel closure leads to incomplete membrane repolarization, preventing sustainable AP propagation. Here, we demonstrate that Kv3.1b channels solve this problem by producing resurgent K(+) currents during repolarization, thus ensuring enough repolarizing power to terminate each AP. Unlike previously described resurgent Na(+) and K(+) currents, Kv3.1b's resurgent current does not originate from recovery of channel block or inactivation but results from a unique combination of steep voltage-dependent gating kinetics and ultra-fast voltage-sensor relaxation. These distinct properties are readily transferrable onto an orthologue Kv channel by transplanting the voltage-sensor's S3-S4 loop, providing molecular insights into the mechanism by which Kv3 channels contribute to high-frequency AP transmission.


Asunto(s)
Potenciales de Acción/genética , Oocitos/metabolismo , Potasio/metabolismo , Canales de Potasio Shaw/genética , Animales , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Cadenas de Markov , Modelos Moleculares , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas , Técnicas de Placa-Clamp , Canales de Potasio Shaw/metabolismo , Xenopus
12.
PLoS One ; 10(9): e0137138, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26348848

RESUMEN

The intrinsic electrical properties of a neuron depend on expression of voltage gated potassium (Kv) channel isoforms, as well as their distribution and density in the plasma membrane. Recently, we showed that N-glycosylation site occupancy of Kv3.1b modulated its placement in the cell body and neurites of a neuronal-derived cell line, B35 neuroblastoma cells. To extrapolate this mechanism to other N-glycosylated Kv channels, we evaluated the impact of N-glycosylation occupancy of Kv3.1a and Kv1.1 channels. Western blots revealed that wild type Kv3.1a and Kv1.1 α-subunits had complex and oligomannose N-glycans, respectively, and that abolishment of the N-glycosylation site(s) generated Kv proteins without N-glycans. Total internal reflection fluorescence microscopy images revealed that N-glycans of Kv3.1a contributed to its placement in the cell membrane while N-glycans had no effect on the distribution of Kv1.1. Based on particle analysis of EGFP-Kv proteins in the adhered membrane, glycosylated forms of Kv3.1a, Kv1.1, and Kv3.1b had differences in the number, size or density of Kv protein clusters in the cell membrane of neurites and cell body of B35 cells. Differences were also observed between the unglycosylated forms of the Kv proteins. Cell dissociation assays revealed that cell-cell adhesion was increased by the presence of complex N-glycans of Kv3.1a, like Kv3.1b, whereas cell adhesion was similar in the oligomannose and unglycosylated Kv1.1 subunit containing B35 cells. Our findings provide direct evidence that N-glycans of Kv3.1 splice variants contribute to the placement of these glycoproteins in the plasma membrane of neuronal-derived cells while those of Kv1.1 were absent. Further when the cell membrane distribution of the Kv channel was modified by N-glycans then the cell-cell adhesion properties were altered. Our study demonstrates that N-glycosylation of Kv3.1a, like Kv3.1b, provides a mechanism for the distribution of these proteins to the cell body and outgrowths and thereby can generate different voltage-dependent conductances in these membranes.


Asunto(s)
Canal de Potasio Kv.1.1/genética , Proteínas de la Membrana/metabolismo , Polisacáridos/metabolismo , Canales de Potasio Shaw/metabolismo , Empalme Alternativo/genética , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Glicosilación , Canal de Potasio Kv.1.1/metabolismo , Proteínas de la Membrana/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Neuritas/metabolismo , Neuronas/metabolismo , Polisacáridos/química , Ratas , Canales de Potasio Shaw/genética
13.
FEBS Lett ; 588(1): 86-91, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24291260

RESUMEN

The Kv3.1 channel plays a crucial role in regulating the high-frequency firing properties of neurons. Here, we determined whether Src regulates the subcellular distributions of the Kv3.1b channel. Co-expression of active Src induced a dramatic redistribution of Kv3.1b to the endoplasmic reticulum. Furthermore, co-expression of the Kv3.1b channel with active Src induced a remarkable decrease in the pool of Kv3.1b at the cell surface. Moreover, the co-expression of active Src results in a significant decrease in the peak current densities of the Kv3.1b channel, and a substantial alteration in the voltage dependence of its steady-state inactivation. Taken together, these results indicate that Src kinase may play an important role in regulating membrane trafficking of Kv3.1b channels.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Canales de Potasio Shaw/metabolismo , Animales , Western Blotting , Células COS , Membrana Celular/fisiología , Chlorocebus aethiops , Células HEK293 , Humanos , Potenciales de la Membrana/fisiología , Ratones , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Técnicas de Placa-Clamp , Transporte de Proteínas , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Ratas , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/fisiología
14.
Biochim Biophys Acta ; 1840(1): 595-604, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24161696

RESUMEN

BACKGROUND: Vacancy of occupied N-glycosylation sites of glycoproteins is quite disruptive to a multicellular organism, as underlined by congenital disorders of glycosylation. Since a neuronal component is typically associated with this disease, we evaluated the impact of N-glycosylation processing of a neuronal voltage gated potassium channel, Kv3.1b, expressed in a neuronal-derived cell line, B35 neuroblastoma cells. METHODS: Total internal reflection fluorescence and differential interference contrast microscopy measurements of live B35 cells expressing wild type and glycosylation mutant Kv3.1b proteins were used to evaluate the distribution of the various forms of the Kv3.1b protein in the cell body and outgrowths. Cell adhesion assays were also employed. RESULTS: Microscopy images revealed that occupancy of both N-glycosylation sites of Kv3.1b had relatively similar amounts of Kv3.1b in the outgrowth and cell body while vacancy of one or both sites led to increased accumulation of Kv3.1b in the cell body. Further both the fully glycosylated and partially glycosylated N229Q Kv3.1b proteins formed higher density particles in outgrowths compared to cell body. Cellular assays demonstrated that the distinct spatial arrangements altered cell adhesion properties. CONCLUSIONS: Our findings provide direct evidence that occupancy of the N-glycosylation sites of Kv3.1b contributes significantly to its lateral heterogeneity in membranes of neuronal-derived cells, and in turn alters cellular properties. GENERAL SIGNIFICANCE: Our study demonstrates that N-glycans of Kv3.1b contain information regarding the association, clustering, and distribution of Kv3.1b in the cell membrane, and furthermore that decreased occupancy caused by congenital disorders of glycosylation may alter the biological activity of Kv3.1b.


Asunto(s)
Membrana Celular/metabolismo , Neuroblastoma/patología , Neuronas/metabolismo , Polisacáridos/metabolismo , Canales de Potasio Shaw/metabolismo , Western Blotting , Adhesión Celular , Proliferación Celular , Glicósido Hidrolasas/metabolismo , Glicosilación , Humanos , Neuroblastoma/metabolismo , Neuronas/citología , Canales de Potasio Shaw/genética , Fracciones Subcelulares , Células Tumorales Cultivadas
15.
J Neurophysiol ; 110(5): 1097-106, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23741043

RESUMEN

The neuropeptide vasoactive intestinal peptide (VIP) is expressed at high levels in the neurons of the suprachiasmatic nucleus (SCN). While VIP is known to be important to the input and output pathways from the SCN, the physiological effects of VIP on electrical activity of SCN neurons are not well known. Here the impact of VIP on firing rate of SCN neurons was investigated in mouse slice cultures recorded during the night. The application of VIP produced an increase in electrical activity in SCN slices that lasted several hours after treatment. This is a novel mechanism by which this peptide can produce long-term changes in central nervous system physiology. The increase in action potential frequency was blocked by a VIP receptor antagonist and lost in a VIP receptor knockout mouse. In addition, inhibitors of both the Epac family of cAMP binding proteins and cAMP-dependent protein kinase (PKA) blocked the induction by VIP. The persistent increase in spike rate following VIP application was not seen in SCN neurons from mice deficient in Kv3 channel proteins and was dependent on the clock protein PER1. These findings suggest that VIP regulates the long-term firing rate of SCN neurons through a VIPR2-mediated increase in the cAMP pathway and implicate the fast delayed rectifier (FDR) potassium currents as one of the targets of this regulation.


Asunto(s)
Neuronas/fisiología , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/farmacología , Animales , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Proteínas Circadianas Period/metabolismo , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/fisiología , Transducción de Señal , Núcleo Supraquiasmático/efectos de los fármacos
16.
Pflugers Arch ; 465(8): 1209-21, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23443853

RESUMEN

Aberrant ion channel expression in the plasma membrane is characteristic for many tumor entities and has been attributed to neoplastic transformation, tumor progression, metastasis, and therapy resistance. The present study aimed to define the function of these "oncogenic" channels for radioresistance of leukemia cells. Chronic myeloid leukemia cells were irradiated (0-6 Gy X ray), ion channel expression and activity, Ca(2+)- and protein signaling, cell cycle progression, and cell survival were assessed by quantitative reverse transcriptase-polymerase chain reaction, patch-clamp recording, fura-2 Ca(2+)-imaging, immunoblotting, flow cytometry, and clonogenic survival assays, respectively. Ionizing radiation-induced G2/M arrest was preceded by activation of Kv3.4-like voltage-gated potassium channels. Channel activation in turn resulted in enhanced Ca(2+) entry and subsequent activation of Ca(2+)/calmodulin-dependent kinase-II, and inactivation of the phosphatase cdc25B and the cyclin-dependent kinase cdc2. Accordingly, channel inhibition by tetraethylammonium and blood-depressing substance-1 and substance-2 or downregulation by RNA interference led to release from radiation-induced G2/M arrest, increased apoptosis, and decreased clonogenic survival. Together, these findings indicate the functional significance of voltage-gated K(+) channels for the radioresistance of myeloid leukemia cells.


Asunto(s)
Ciclo Celular/genética , Supervivencia Celular/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Apoptosis/genética , Proteína Quinasa CDC2 , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , División Celular/genética , Línea Celular Tumoral , Células Cultivadas , Ciclina B/genética , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Fase G2/genética , Humanos , Células K562 , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Tolerancia a Radiación/genética , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
17.
Biophys J ; 103(4): 669-76, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22947928

RESUMEN

Membrane proteins that respond to changes in transmembrane voltage are critical in regulating the function of living cells. The voltage-sensing domains (VSDs) of voltage-gated ion channels are extensively studied to elucidate voltage-sensing mechanisms, and yet many aspects of their structure-function relationship remain elusive. Here, we transplanted homologous amino acid motifs from the tetrameric voltage-activated potassium channel Kv3.1 to the monomeric VSD of Ciona intestinalis voltage-sensitive phosphatase (Ci-VSP) to explore which portions of Kv3.1 subunits depend on the tetrameric structure of Kv channels and which properties of Kv3.1 can be transferred to the monomeric Ci-VSP scaffold. By attaching fluorescent proteins to these chimeric VSDs, we obtained an optical readout to establish membrane trafficking and kinetics of voltage-dependent structural rearrangements. We found that motifs extending from 10 to roughly 100 amino acids can be readily transplanted from Kv3.1 into Ci-VSP to form engineered VSDs that efficiently incorporate into the plasma membrane and sense voltage. Some of the functional features of these engineered VSDs are reminiscent of Kv3.1 channels, indicating that these properties do not require interactions between Kv subunits or between the voltage sensing and the pore domains of Kv channels.


Asunto(s)
Ciona intestinalis/enzimología , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Ingeniería de Proteínas/métodos , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Datos de Secuencia Molecular , Células PC12 , Monoéster Fosfórico Hidrolasas/química , Porosidad , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales de Potasio Shaw/química
18.
J Neurophysiol ; 106(6): 3230-44, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21957229

RESUMEN

Fast onset and high-level neurospecific transgene expression in vivo is of importance for many areas in neuroscience, from basic to translational, and can significantly reduce the amount of vector load required to maintain transgene expression in vivo. In this study, we tested various cis elements to optimize transgene expression at transcriptional, posttranscriptional, and posttranslational levels and combined them together to create the high-level neuronal transgene expression cassette pUNISHER. Using a second-generation adenoviral vector system in combination with the pUNISHER cassette, we characterized its rate of onset of detectable expression and levels of expression compared with a neurospecific expression cassette driven by the 470-bp human synapsin promoter in vitro and in vivo. Our results demonstrate in primary neurons that the pUNISHER cassette, in a recombinant adenovirus type 5 background, led to a faster rate of onset of detectable transgene expression and higher level of transgene expression. More importantly, this cassette led to highly correlated neuronal expression in vivo and to stable transgene expression up to 30 days in the auditory brain stem with no toxicity on the characteristics of synaptic transmission and plasticity at the calyx of Held synapse. Thus the pUNISHER cassette is an ideal high-level neuronal expression cassette for use in vivo for neuroscience applications.


Asunto(s)
Sistema Nervioso Central/citología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Neuronas/metabolismo , Adenoviridae/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Línea Celular Transformada , Citomegalovirus/genética , Potenciales Postsinápticos Excitadores/genética , Regulación de la Expresión Génica/genética , Técnicas de Transferencia de Gen/instrumentación , Proteínas Fluorescentes Verdes/genética , Humanos , Técnicas In Vitro , Potenciales de la Membrana/genética , Ratones , Técnicas de Placa-Clamp , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Regiones Promotoras Genéticas/genética , Procesamiento Postranscripcional del ARN , Ratas , Ratas Wistar , Secuencias Reguladoras de Ácidos Nucleicos , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Transmisión Sináptica/genética , Factores de Tiempo , Transfección
19.
FEBS Lett ; 585(20): 3322-7, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-21945320

RESUMEN

The sialic acid of complex N-glycans can be biochemically engineered by substituting the physiological precursor N-acetylmannosamine with non-natural N-acylmannosamines. The Kv3.1 glycoprotein, a neuronal voltage-gated potassium channel, contains sialic acid. Western blots of the Kv3.1 glycoprotein isolated from transfected B35 neuroblastoma cells incubated with N-acylmannosamines verified sialylated N-glycans attached to the Kv3.1 glycoprotein. Outward ionic currents of Kv3.1 transfected B35 cells treated with N-pentanoylmannosamine or N-propanoylmannosamine had slower activation and inactivation rates than those of untreated cells. Therefore, the N-acyl side chain of sialic acid is intimately connected with the activation and inactivation rates of this glycosylated potassium channel.


Asunto(s)
Hexosaminas/farmacología , Ingeniería Metabólica , Ácido N-Acetilneuramínico/metabolismo , Canales de Potasio Shaw/metabolismo , Línea Celular Tumoral , Glicosilación , Hexosaminas/metabolismo , Humanos , Transporte Iónico/genética , Ácido N-Acetilneuramínico/genética , Canales de Potasio Shaw/genética
20.
PLoS One ; 5(11): e13843, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21079792

RESUMEN

As a first step towards discovery of genes expressed from only one allele in the CNS, we used a tiling array assay for DNA sequences that are both methylated and unmethylated (the MAUD assay). We analyzed regulatory regions of the entire mouse brain transcriptome, and found that approximately 10% of the genes assayed showed dual DNA methylation patterns. They include a large subset of genes that display marks of both active and silent, i.e., poised, chromatin during development, consistent with a link between differential DNA methylation and lineage-specific differentiation within the CNS. Sixty-five of the MAUD hits and 57 other genes whose function is of relevance to CNS development and/or disorders were tested for allele-specific expression in F(1) hybrid clonal neural stem cell (NSC) lines. Eight MAUD hits and one additional gene showed such expression. They include Lgi1, which causes a subtype of inherited epilepsy that displays autosomal dominance with incomplete penetrance; Gfra2, a receptor for glial cell line-derived neurotrophic factor GDNF that has been linked to kindling epilepsy; Unc5a, a netrin-1 receptor important in neurodevelopment; and Cspg4, a membrane chondroitin sulfate proteoglycan associated with malignant melanoma and astrocytoma in human. Three of the genes, Camk2a, Kcnc4, and Unc5a, show preferential expression of the same allele in all clonal NSC lines tested. The other six genes show a stochastic pattern of monoallelic expression in some NSC lines and bi-allelic expression in others. These results support the estimate that 1-2% of genes expressed in the CNS may be subject to allelic exclusion, and demonstrate that the group includes genes implicated in major disorders of the CNS as well as neurodevelopment.


Asunto(s)
Sistema Nervioso Central/metabolismo , Cromatina/genética , Metilación de ADN , Perfilación de la Expresión Génica , Alelos , Animales , Antígenos/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Diferenciación Celular/genética , Línea Celular , Sistema Nervioso Central/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Netrina , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Proteínas/genética , Proteoglicanos/genética , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio Shaw/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA