Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 23(13): 3827-3838, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949767

RESUMEN

Altered glutamatergic neurotransmission is thought to contribute to mental disorders and neurodegenerative diseases. Copy-number variation in genes associated with glutamatergic synapses represents a source of genetic variability, possibly underlying neurological and mental disease susceptibility. The GRIK4 gene encodes a high-affinity kainate receptor subunit of essentially unknown function, although de novo duplication of the 11q23.3-q24.1 locus to which it maps has been detected in autism and other disorders. To determine how changes in the dose of Grik4 affect synaptic activity, we studied mice overexpressing this gene in the forebrain. A mild gain in Grik4 enhances synaptic transmission, causing a persistent imbalance in inhibitory and excitatory activity and disturbing the circuits responsible for the main amygdala outputs. These changes in glutamatergic activity reverse when Grik4 levels are normalized; thus, they may account for the behavioral abnormalities in disorders like autism or schizophrenia.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Receptores de Ácido Kaínico/genética , Animales , Complejo Nuclear Basolateral/patología , Conducta Animal , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Dosificación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Glutamato/metabolismo , Receptores de Ácido Kaínico/metabolismo , Transmisión Sináptica/fisiología , Tetrodotoxina/farmacología
2.
Cereb Cortex ; 26(4): 1464-72, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25316333

RESUMEN

When native and recombinant kainate receptors (KARs) are compared, there is a mismatch in several of their functional properties. While both generate currents, synaptic responses mediated by KARs have rarely observed in cultured hippocampal neurons. The recent discovery of auxiliary proteins for KARs, such as Netos, offers an explanation for these discrepancies. We found that the GluK5 KAR subunit and the ancillary proteins, Neto1 and Neto2, are not expressed by hippocampal neurons in culture. Therefore, we used this model to directly test whether these proteins are required for the synaptic localization of KARs. Transfection of GluK4, GluK5, Neto1, or Neto2 into hippocampal neurons was associated with the appearance of synaptic KAR-mediated EPSCs. However, GluK4 or GluK5 alone produced synaptic activity in a significant proportion of cells and with reliable event frequency. While neurons expressing GluK4 or GluK5 subunits displayed synaptic responses with rapid kinetics, the expression of Neto proteins conferred these synaptic responses with their characteristic slow onset and decay rates. These data reveal some requirements for KAR targeting to the synapse, indicating a fundamental role of high affinity KAR subunits in this process.


Asunto(s)
Hipocampo/metabolismo , Lipoproteínas LDL/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Receptores de Ácido Kaínico/metabolismo , Sinapsis/metabolismo , Animales , Células Cultivadas , Potenciales Postsinápticos Excitadores , Células HEK293 , Hipocampo/fisiología , Humanos , Proteínas Relacionadas con Receptor de LDL , Lipoproteínas LDL/fisiología , Proteínas de la Membrana/fisiología , Ratones , Neuronas/fisiología , Subunidades de Proteína/metabolismo , Subunidades de Proteína/fisiología , Transporte de Proteínas , Receptores de Ácido Kaínico/fisiología , Receptores de N-Metil-D-Aspartato , Sinapsis/fisiología
3.
J Neurosci ; 35(40): 13619-28, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26446216

RESUMEN

The understanding of brain diseases requires the identification of the molecular, synaptic, and cellular disruptions underpinning the behavioral features that define the disease. The importance of genes related to synaptic function in brain disease has been implied in studies describing de novo germline mutations and copy number variants. Indeed, de novo copy number variations (deletion or duplication of a chromosomal region) of synaptic genes have been recently implicated as risk factors for mental retardation or autism. Among these genes is GRIK4, a gene coding for a glutamate receptor subunit of the kainate type. Here we show that mice overexpressing grik4 in the forebrain displayed social impairment, enhanced anxiety, and depressive states, accompanied by altered synaptic transmission, showing more efficient information transfer through the hippocampal trisynaptic circuit. Together, these data indicate that a single gene variation in the glutamatergic system results in behavioral symptomatology consistent with autism spectrum disorders as well as in alterations in synaptic function in regions involved in social activity. Autistic features of these mice represent powerful tools for improving diagnosis and testing of specific treatments targeting abnormalities in glutamatergic signaling related to autism spectrum disorders. SIGNIFICANCE STATEMENT: A genetic overlap exists between autism spectrum disorders (ASD), currently thought to represent a continuum of the same disorder with varying degrees of severity, and other neurodevelopmental and neuropsychiatric endophenotypes. We show that the duplication of a single gene coding for a high-affinity kainate receptor subunit (i.e., grik4) in a limited area of the brain recapitulates behavioral endophenotypes seen in humans diagnosed with autism (anhedonia, depression, anxiety, and altered social interaction), including some humans with GRIK4 duplications. Therefore, it should be possible to use mice overexpressing grik4 to directly address circuit dysfunctions associated with ASDs and test specific treatments of autism-related behaviors.


Asunto(s)
Trastorno del Espectro Autista/genética , Hipocampo/citología , Mutación/genética , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Transmisión Sináptica/genética , Animales , Animales Recién Nacidos , Trastorno del Espectro Autista/fisiopatología , Línea Celular Transformada , Adaptación a la Oscuridad/genética , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Conducta Exploratoria/fisiología , Preferencias Alimentarias , Guanilato-Quinasas/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Relaciones Interpersonales , Aprendizaje por Laberinto/fisiología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Sacarosa/administración & dosificación , Natación/fisiología
4.
J Neurosci ; 35(13): 5171-9, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834043

RESUMEN

Kainate receptors (KARs) are found ubiquitously in the CNS and are present presynaptically and postsynaptically regulating synaptic transmission and excitability. Functional studies have proven that KARs act as ion channels as well as potentially activating G-proteins, thus indicating the existance of a dual signaling system for KARs. Nevertheless, it is not clear how these ion channels activate G-proteins and which of the KAR subunits is involved. Here we performed a proteomic analysis to define proteins that interact with the C-terminal domain of GluK1 and we identified a variety of proteins with many different functions, including a Go α subunit. These interactions were verified through distinct in vitro and in vivo assays, and the activation of the Go protein by GluK1 was validated in bioluminescence resonance energy transfer experiments, while the specificity of this association was confirmed in GluK1-deficient mice. These data reveal components of the KAR interactome, and they show that GluK1 and Go proteins are natural partners, accounting for the metabotropic effects of KARs.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteómica , Receptores de Ácido Kaínico/química , Receptores de Ácido Kaínico/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Células HEK293 , Humanos , Ácido Kaínico/farmacología , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína , Receptores de Ácido Kaínico/genética
5.
Neuron ; 63(3): 357-71, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19679075

RESUMEN

Regulation of surface insertion and internalization of AMPA and NMDA receptors has emerged as a key mechanism for the control of synaptic strength. Regulatory elements for synaptic kainate receptors (KARs) are, however, largely undetermined. We have found that SNAP25 is critical for the synaptic removal of KARs, acting via GluK5 (i.e., KA2) subunits. SNAP25 coimmunoprecipitates with protein complexes containing PICK1, GRIP1, and GluK5 and colocalizes with GluK5 in both hippocampal neurons and transfected HEK293 cells. In hippocampal slices, purified SNAP25 antibodies and blocking peptides caused a GluK5-dependent run-up of KARs-mediated EPSC (EPSC(KAR)) recorded from CA3 pyramidal neurons when included in the patch pipette and prevented activity-dependent long-term depression of EPSC(KAR). As EPSC(KAR) LTD, SNAP25/PICK1/GluK5 interactions are dynamically regulated by PKC.


Asunto(s)
Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Receptores de Ácido Kaínico/metabolismo , Sinapsis/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Línea Celular Transformada , Línea Celular Tumoral , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Hipocampo/citología , Hipocampo/ultraestructura , Humanos , Inmunoprecipitación/métodos , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naftalenos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/fisiología , Neuroblastoma , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neurotoxinas/farmacología , Proteínas Nucleares/metabolismo , Técnicas de Placa-Clamp/métodos , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Piridinas/farmacología , Ratas , Receptores de Ácido Kaínico/deficiencia , Proteína 25 Asociada a Sinaptosomas/genética , Transfección , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
6.
J Neurosci ; 28(35): 8844-50, 2008 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-18753386

RESUMEN

Acid-sensitive K+ channels of the tandem P-domain K+-channel family (TASK-1 and TASK-3) have been implicated in peripheral and central respiratory chemosensitivity; however, because of the lack of decisive pharmacological agents, the final proof of the role of the TASK channel in the chemosensory control of breathing has been missing. In the mouse, TASK-1 and TASK-3 channels are dispensable for central respiratory chemosensitivity (Mulkey et al., 2007). Here, we have used knock-out animals to determine whether TASK-1 and TASK-3 channels play a role in the carotid body function and chemosensory control of breathing exerted by the carotid body chemoreceptors. Ventilatory responses to hypoxia (10% O2 in inspired air) and moderate normoxic hypercapnia (3-6% CO2 in inspired air) were significantly reduced in TASK-1 knock-out mice. In contrast, TASK-3-deficient mice showed responses to both stimuli that were similar to those developed by their wild-type counterparts. TASK-1 channel deficiency resulted in a marked reduction of the hypoxia (by 49%)- and CO2 (by 68%)-evoked increases in the carotid sinus nerve chemoafferent discharge recorded in the in vitro superfused carotid body/carotid sinus nerve preparations. Deficiency in both TASK-1 and TASK-3 channels increased baseline chemoafferent activity but did not cause a further reduction of the carotid body chemosensory responses. These observations provide direct evidence that TASK-1 channels contribute significantly to the increases in the carotid body chemoafferent discharge in response to a decrease in arterial P(O2) or an increase in P(CO2)/[H+]. TASK-1 channels therefore play a key role in the control of ventilation by peripheral chemoreceptors.


Asunto(s)
Cuerpo Carotídeo/fisiología , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Ventilación Pulmonar/genética , Respiración/genética , Análisis de Varianza , Animales , Tronco Encefálico , Dióxido de Carbono/farmacología , Hipercapnia/genética , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Pletismografía Total/métodos , Canales de Potasio/deficiencia , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Ventilación Pulmonar/efectos de los fármacos , Médula Espinal , Volumen de Ventilación Pulmonar/fisiología , Vigilia
7.
J Pharmacol Exp Ther ; 327(1): 277-86, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18660435

RESUMEN

TASK two-pore-domain leak K(+) channels occur throughout the brain. However, TASK-1 and TASK-3 knockout (KO) mice have few neurological impairments and only mildly reduced sensitivities to inhalational anesthetics, contrasting with the anticipated functions and importance of these channels. TASK-1/-3 channel expression can compensate for the absence of GABA(A) receptors in GABA(A) alpha6 KO mice. To investigate the converse, we analyzed the behavior of TASK-1 and -3 KO mice after administering drugs with preferential efficacies at GABA(A) receptor subtypes: benzodiazepines (diazepam and flurazepam, active at alpha1betagamma2, alpha2betagamma2, alpha3betagamma2, and alpha5betagamma2 subtypes), zolpidem (alpha1betagamma2 subtype), propofol (beta2-3-containing receptors), gaboxadol (alpha4betadelta and alpha6betadelta subtypes), pregnanolone, and pentobarbital (many subtypes). TASK-1 KO mice showed increased motor impairment in rotarod and beam-walking tests after diazepam and flurazepam administration but not after zolpidem. They also showed prolonged loss of righting reflex induced by propofol and pentobarbital. Autoradiography indicated no change in GABA(A) receptor ligand binding levels. These altered behavioral responses to GABAergic drugs suggest functional up-regulation of alpha2beta2/3gamma2 and alpha3beta2/3gamma2 receptor subtypes in TASK-1 KO mice. In addition, female, but not male, TASK-1 KO mice were more sensitive to gaboxadol, suggesting an increased influence of alpha4betadelta or alpha6betadelta subtypes. The benzodiazepine sensitivity of TASK-3 KO mice was marginally increased. Our results underline that TASK-1 channels perform such key functions in the brain that compensation is needed for their absence. Furthermore, because inhalation anesthetics act partially through GABA(A) receptors, the up-regulation of GABA(A) receptor function in TASK-1 KO mice might mask TASK-1 channel's significance as a target for inhalation anesthetics.


Asunto(s)
Ataxia/inducido químicamente , Hipnóticos y Sedantes/farmacología , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Receptores de GABA-A/fisiología , Animales , Ansiolíticos/farmacología , Benzodiazepinas/farmacología , Encéfalo/metabolismo , Diazepam/farmacología , Femenino , Flurazepam/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Pentobarbital/farmacología , Propofol/farmacología , Proteína Quinasa C/fisiología , Receptores de GABA-A/efectos de los fármacos , Factores Sexuales , Médula Espinal/metabolismo
8.
EMBO J ; 27(1): 179-87, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18034154

RESUMEN

TASK1 (KCNK3) and TASK3 (KCNK9) are two-pore domain potassium channels highly expressed in adrenal glands. TASK1/TASK3 heterodimers are believed to contribute to the background conductance whose inhibition by angiotensin II stimulates aldosterone secretion. We used task1-/- mice to analyze the role of this channel in adrenal gland function. Task1-/- exhibited severe hyperaldosteronism independent of salt intake, hypokalemia, and arterial 'low-renin' hypertension. The hyperaldosteronism was fully remediable by glucocorticoids. The aldosterone phenotype was caused by an adrenocortical zonation defect. Aldosterone synthase was absent in the outer cortex normally corresponding to the zona glomerulosa, but abundant in the reticulo-fasciculata zone. The impaired mineralocorticoid homeostasis and zonation were independent of the sex in young mice, but were restricted to females in adults. Patch-clamp experiments on adrenal cells suggest that task3 and other K+ channels compensate for the task1 absence. Adrenal zonation appears as a dynamic process that even can take place in adulthood. The striking changes in the adrenocortical architecture in task1-/- mice are the first demonstration of the causative role of a potassium channel in development/differentiation.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Homeostasis/genética , Mineralocorticoides/antagonistas & inhibidores , Mineralocorticoides/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Glándulas Suprarrenales/patología , Aldosterona/sangre , Aldosterona/metabolismo , Animales , Femenino , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Potasio/sangre , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Renina/sangre
9.
J Pharmacol Exp Ther ; 323(3): 924-34, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17875609

RESUMEN

The TASK-3 channel is an acid-sensitive two-pore-domain K+ channel, widely expressed in the brain and probably involved in regulating numerous neuronal populations. Here, we characterized the behavioral and pharmacological phenotypes of TASK-3 knockout (KO) mice. Circadian locomotor activity measurements revealed that the nocturnal activity of the TASK-3 KO mice was increased by 38% (P < 0.01) compared with wild-type littermate controls, light phase activity being similar. Although TASK-3 channels are abundant in cerebellar granule cells, the KO mice performed as well as the wild-type mice in walking on a rotating rod or along a 1.2-cm-diameter beam. However, they fell more frequently from a narrower 0.8-cm beam. The KO mice showed impaired working memory in the spontaneous alternation task, with the alternation percentage being 62 +/- 3% for the wild-type mice and 48 +/- 4% (P < 0.05) for the KO mice. Likewise, during training for the Morris water-maze spatial memory task, the KO mice were slower to find the hidden platform, and in the probe trial, the female KO mice visited fewer times the platform quadrant than the male KO and wild-type mice. In pharmacological tests, the TASK-3 KO mice showed reduced sensitivity to the inhalation anesthetic halothane and the cannabinoid receptor agonist WIN55212-2 mesylate [(R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate] but unaltered responses to the alpha2 adrenoceptor agonist dexmedetomidine, the i.v. anesthetic propofol, the opioid receptor agonist morphine, and the local anesthetic lidocaine. Overall, our results suggest important contributions of TASK-3 channels in the neuronal circuits regulating circadian rhythms, cognitive functions, and mediating specific pharmacological effects.


Asunto(s)
Anestésicos por Inhalación , Conducta Animal/fisiología , Ritmo Circadiano/fisiología , Aprendizaje por Laberinto/fisiología , Actividad Motora/fisiología , Canales de Potasio/fisiología , Anestésicos por Inhalación/farmacología , Anestésicos por Inhalación/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dolor/tratamiento farmacológico , Dimensión del Dolor , Umbral del Dolor/fisiología , Canales de Potasio/genética
10.
J Neurosci ; 27(35): 9329-40, 2007 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-17728447

RESUMEN

The ability of neurons, such as cerebellar granule neurons (CGNs), to fire action potentials (APs) at high frequencies during sustained depolarization is usually explained in relation to the functional properties of voltage-gated ion channels. Two-pore domain potassium (K(2P)) channels are considered to simply hyperpolarize the resting membrane potential (RMP) by increasing the potassium permeability of the membrane. However, we find that CGNs lacking the TASK-3 type K(2P) channel exhibit marked accommodation of action potential firing. The accommodation phenotype was not associated with any change in the functional properties of the underlying voltage-gated sodium channels, nor could it be explained by the more depolarized RMP that resulted from TASK-3 channel deletion. A functional rescue, involving the introduction of a nonlinear leak conductance with a dynamic current clamp, was able to restore wild-type firing properties to adult TASK-3 knock-out CGNs. Thus, in addition to the accepted role of TASK-3 channels in limiting neuronal excitability, by increasing the resting potassium conductance TASK-3 channels also increase excitability by supporting high-frequency firing once AP threshold is reached.


Asunto(s)
Potenciales de Acción/fisiología , Cerebelo/citología , Neuronas/fisiología , Canales de Potasio/fisiología , Potenciales de Acción/genética , Anestésicos Locales/farmacología , Animales , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Hibridación in Situ/métodos , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/efectos de la radiación , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/deficiencia , Estructura Terciaria de Proteína/fisiología , Tetraetilamonio/farmacología , Tetrodotoxina/farmacología
11.
Alcohol ; 41(3): 163-76, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17591542

RESUMEN

In rodent models, gamma-aminobutyric acid A (GABAA) receptors with the alpha6 and delta subunits, expressed in the cerebellar and cochlear nucleus granule cells, have been linked to ethanol sensitivity and voluntary ethanol drinking. Here, we review the findings. When considering both in vivo contributions and data on cloned receptors, the evidence for direct participation of the alpha6-containing receptors to increased ethanol sensitivity is poor. The alpha6 subunit-knockout mouse lines do not have any changed sensitivity to ethanol, although these mice do display increased benzodiazepine sensitivity. However, in general the compensations occurring in knockout mice (regardless of which particular gene is knocked out) tend to fog interpretations of drug actions at the systems level. For example, the alpha6 knockout mice have increased TASK-1 channel expression in their cerebellar granule cells, which could influence sensitivity to ethanol in the opposite direction to that obtained with the alpha6 knockouts. Indeed, TASK-1 knockout mice are more impaired than wild types in motor skills when given ethanol; this might explain why GABAA receptor alpha6 knockout mice have unchanged ethanol sensitivities. As an alternative to studying knockout mice, we examined the claimed delta subunit-dependent/gamma2 subunit-independent ethanol/[3H]Ro 15-4513 binding sites on GABAA receptors. We looked at [3H]Ro 15-4513 binding in HEK 293 cell membrane homogenates containing rat recombinant alpha6/4beta3delta receptors and in mouse brain sections. Specific high-affinity [3H]Ro 15-4513 binding could not be detected under any conditions to the recombinant receptors or to the cerebellar sections of gamma2(F77I) knockin mice, nor was this binding to brain sections of wild-type C57BL/6 inhibited by 1-100 mM ethanol. Since ethanol may act on many receptor and channel protein targets in neuronal membranes, we consider the alpha6 (and alpha4) subunit-containing GABAA receptors unlikely to be directly responsible for any major part of ethanol's actions. Therefore, we finish the review by discussing more generally alcohol and GABAA receptors and by suggesting potential future directions for this research.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Receptores de GABA-A/efectos de los fármacos , Animales , Azidas/metabolismo , Azidas/farmacología , Benzodiazepinas/metabolismo , Benzodiazepinas/farmacología , Unión Competitiva/efectos de los fármacos , Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Tolerancia a Medicamentos , Etanol/antagonistas & inhibidores , Humanos , Ratones , Ratas , Receptores de GABA-A/genética
12.
PLoS One ; 2(6): e533, 2007 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-17579707

RESUMEN

To identify the underlying reason for the controversial performance of tetracycline (Tet)-controlled regulated gene expression in mammalian neurons, we investigated each of the three components that comprise the Tet inducible systems, namely tetracyclines as inducers, tetracycline-transactivator (tTA) and reverse tTA (rtTA), and tTA-responsive promoters (P(tets)). We have discovered that stably integrated P(tet) becomes functionally silenced in the majority of neurons when it is inactive during development. P(tet) silencing can be avoided when it is either not integrated in the genome or stably-integrated with basal activity. Moreover, long-term, high transactivator levels in neurons can often overcome integration-induced P(tet) gene silencing, possibly by inducing promoter accessibility.


Asunto(s)
Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Regulación de la Expresión Génica , Silenciador del Gen/efectos de los fármacos , Tetraciclina/farmacología , Transactivadores/genética , Animales , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación in Situ , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Transactivadores/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
13.
J Neurosci ; 27(10): 2560-9, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344393

RESUMEN

Tonic inhibition has emerged as a key regulator of neuronal excitability in the CNS. Thalamic relay neurons of the dorsal lateral geniculate nucleus (dLGN) exhibit a tonic GABA(A) receptor (GABA(A)R)-mediated conductance that is correlated with delta-subunit expression. Indeed, consistent with the absence of delta-subunit expression, no tonic conductance is found in the adjacent ventral LGN. We show that, in contrast to the situation in cerebellar granule cells, thalamic delta-subunit-containing GABA(A)Rs (delta-GABA(A)Rs) do not contribute to a spillover component of IPSCs in dLGN. However, tonic activation of thalamic delta-GABA(A)Rs is sensitive to the global level of inhibition, showing an absolute requirement on the synaptic release of GABA. Thus, the tonic conductance is abolished when transmitter release probability is reduced or action potential-evoked release is blocked. We further show that continuous activation of delta-GABA(A)Rs introduces variability into the timing of low-threshold rebound bursts. Hence, activation of delta-GABA(A)Rs could act to destabilize thalamocortical oscillations and therefore have an important impact on behavioral state.


Asunto(s)
Neuronas/fisiología , Receptores de GABA-A/fisiología , Sinapsis/fisiología , Tálamo/fisiología , Animales , Cloruros/metabolismo , Umbral Diferencial , Conductividad Eléctrica , Cuerpos Geniculados/citología , Cuerpos Geniculados/fisiología , Homeostasis/fisiología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibición Neural/fisiología , Isoformas de Proteínas/fisiología , Tiempo de Reacción , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
14.
J Pharmacol Exp Ther ; 317(2): 615-26, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16397088

RESUMEN

Inhalation anesthetics activate and cannabinoid agonists inhibit TWIK-related acid-sensitive K(+) channels (TASK)-1 two-pore domain leak K(+) channels in vitro. Many neuromodulators, such as noradrenaline, might also manifest some of their actions by modifying TASK channel activity. Here, we have characterized the basal behavioral phenotype of TASK-1 knockout mice and tested their sensitivity to the inhalation anesthetics halothane and isoflurane, the alpha(2) adrenoreceptor agonist dexmedetomidine, and the cannabinoid agonist WIN55212-2 mesylate [R-(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrrolo[1,2,3,-de]-1,4-benzoxazinyl]-(1-naphtalenyl)methanone mesylate)]. TASK-1 knockout mice had a largely normal behavioral phenotype. Male, but not female, knockout mice displayed an enhanced acoustic startle response. The knockout mice showed increased sensitivity to thermal nociception in a hot-plate test but not in a tail-flick test. The analgesic, sedative, and hypothermic effects of WIN55212-2 (2-6 mg/kg s.c.) were reduced in TASK-1 knockout mice. These results implicate TASK-1-containing channels in supraspinal pain pathways, in particular those modulated by endogenous cannabinoids. TASK-1 knockout mice were less sensitive to the anesthetic effects of halothane and isoflurane than wild-type littermates, requiring higher anesthetic concentrations to induce immobility as reflected by loss of the tail-withdrawal reflex. Our results support the idea that the activation of multiple background K(+) channels is crucial for the high potency of inhalation anesthetics. Furthermore, TASK-1 knockout mice were less sensitive to the sedative effects of dexmedetomidine (0.03 mg/kg s.c.), suggesting a role for the TASK-1 channels in the modulation of function of the adrenergic locus coeruleus nuclei and/or other neuronal systems.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2 , Anestésicos por Inhalación/farmacología , Conducta Animal/efectos de los fármacos , Agonistas de Receptores de Cannabinoides , Dexmedetomidina/farmacología , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Animales , Benzoxazinas , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Morfolinas/farmacología , Naftalenos/farmacología , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/genética
15.
Mol Pharmacol ; 69(4): 1468-76, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16424077

RESUMEN

A genetic knockout was used to determine the specific contribution of TWIK-related acid-sensitive K+ (TASK)-1 channels to the function of dorsal lateral geniculate nucleus (DLG) thalamocortical relay (TC) neurons. Disruption of TASK-1 function produced an approximately 19% decrease in amplitude of the standing outward current (ISO) and a 3 +/- 1-mV depolarizing shift in resting membrane potential (Vrest) of DLG neurons. We estimated that current through TASK-1 homodimers or TASK-1/TASK-3 heterodimers contribute(s) approximately one third of the current sensitive to TASK channel modulators in DLG TC neurons. The effects of the TASK channel blocker bupivacaine (20 microM), of muscarine (50 microM), and of H+ on ISO were reduced to approximately 60%, 59%, and shifted to more acidic pH values, respectively. The blocking effect of anandamide on ISO [30 microM; 23 +/- 3% current decrease in wild type (WT)] was absent in TASK-1 knockout (TASK-1-/-) mice (9 +/- 6% current increase). Comparable results were obtained with the more stable anand-amide derivative methanandamide (20 microM; 20 +/- 2% decrease in WT; 4 +/- 6% increase in TASK-1-/-). Current-clamp recordings revealed a muscarine-induced shift in TC neuron activity from burst to tonic firing in both mouse genotypes. Electrocorticograms and sleep/wake times were unchanged in TASK-1-/- mice. In conclusion, our findings demonstrate a significant contribution of TASK-1 channels to ISO in DLG TC neurons, although the genetic knockout of TASK-1 did not produce severe deficits in the thalamocortical system.


Asunto(s)
Cuerpos Geniculados/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Anestésicos Locales/farmacología , Animales , Ácidos Araquidónicos/farmacología , Conducta Animal , Benzoxazinas , Bupivacaína/farmacología , Electroencefalografía , Endocannabinoides , Cuerpos Geniculados/citología , Cuerpos Geniculados/efectos de los fármacos , Hibridación in Situ , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Morfolinas/farmacología , Muscarina/farmacología , Naftalenos/farmacología , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Alcamidas Poliinsaturadas , Canales de Potasio de Dominio Poro en Tándem/genética
16.
J Neurosci ; 25(49): 11455-67, 2005 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-16339039

RESUMEN

Two-pore domain potassium (K2P) channel expression is believed to underlie the developmental emergence of a potassium leak conductance [IK(SO)] in cerebellar granule neurons (CGNs), suggesting that K2P function is an important determinant of the input conductance and resting membrane potential. To investigate the role that different K2P channels may play in the regulation of CGN excitability, we generated a mouse lacking TASK-1, a K2P channel known to have high expression levels in CGNs. In situ hybridization and real-time PCR studies in wild-type and TASK-1 knock-outs (KOs) demonstrated that the expression of other K2P channels was unaltered in CGNs. TASK-1 knock-out mice were healthy and bred normally but exhibited compromised motor performance consistent with altered cerebellar function. Whole-cell recordings from adult cerebellar slice preparations revealed that the resting excitability of mature CGNs was no different in TASK-1 KO and littermate controls. However, the modulation of IK(SO) by extracellular Zn2+, ruthenium red, and H+ was altered. The IK(SO) recorded from TASK-1 knock-out CGNs was no longer sensitive to alkalization and was blocked by Zn2+ and ruthenium red. These results suggest that a TASK-1-containing channel population has been replaced by a homodimeric TASK-3 population in the TASK-1 knock-out. These data directly demonstrate that TASK-1 channels contribute to the properties of IK(SO) in adult CGNs. However, TASK channel subunit composition does not alter the resting excitability of CGNs but does influence sensitivity to endogenous modulators such as Zn2+ and H+.


Asunto(s)
Cerebelo/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Subunidades de Proteína/fisiología , Animales , Células Cultivadas , Cerebelo/citología , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Trastornos de la Destreza Motora/genética , Trastornos de la Destreza Motora/fisiopatología , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Canales de Potasio de Dominio Poro en Tándem/genética , Subunidades de Proteína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...