Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Nature ; 464(7293): 1376-80, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20393464

RESUMEN

Schizophrenia is a complex disorder that interferes with the function of several brain systems required for cognition and normal social behaviour. Although the most notable clinical aspects of the disease only become apparent during late adolescence or early adulthood, many lines of evidence suggest that schizophrenia is a neurodevelopmental disorder with a strong genetic component. Several independent studies have identified neuregulin 1 (NRG1) and its receptor ERBB4 as important risk genes for schizophrenia, although their precise role in the disease process remains unknown. Here we show that Nrg1 and ErbB4 signalling controls the development of inhibitory circuitries in the mammalian cerebral cortex by cell-autonomously regulating the connectivity of specific GABA (gamma-aminobutyric acid)-containing interneurons. In contrast to the prevalent view, which supports a role for these genes in the formation and function of excitatory synapses between pyramidal cells, we found that ErbB4 expression in the mouse neocortex and hippocampus is largely confined to certain classes of interneurons. In particular, ErbB4 is expressed by many parvalbumin-expressing chandelier and basket cells, where it localizes to axon terminals and postsynaptic densities receiving glutamatergic input. Gain- and loss-of-function experiments, both in vitro and in vivo, demonstrate that ErbB4 cell-autonomously promotes the formation of axo-axonic inhibitory synapses over pyramidal cells, and that this function is probably mediated by Nrg1. In addition, ErbB4 expression in GABA-containing interneurons regulates the formation of excitatory synapses onto the dendrites of these cells. By contrast, ErbB4 is dispensable for excitatory transmission between pyramidal neurons. Altogether, our results indicate that Nrg1 and ErbB4 signalling is required for the wiring of GABA-mediated circuits in the postnatal cortex, providing a new perspective to the involvement of these genes in the aetiology of schizophrenia.


Asunto(s)
Corteza Cerebral/metabolismo , Receptores ErbB/metabolismo , Interneuronas/metabolismo , Vías Nerviosas/fisiología , Neurregulina-1/metabolismo , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo , Animales , Diferenciación Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Dendritas/metabolismo , Embrión de Mamíferos , Receptores ErbB/deficiencia , Receptores ErbB/genética , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Técnicas In Vitro , Ratones , Inhibición Neural/genética , Inhibición Neural/fisiología , Células Piramidales/metabolismo , Receptor ErbB-4 , Esquizofrenia/genética , Esquizofrenia/metabolismo , Sinapsis/metabolismo
5.
J Neurosci ; 33(46): 18298-310, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227739

RESUMEN

The CRMP2 and CRMP4 proteins are strongly expressed in the developing nervous system, mediating neurite outgrowth, neuronal polarity, and axon guidance. In the present study, we demonstrate the interaction of the CRMP2 and CRMP4 proteins with the GluK5 subunit of the kainate (KA) receptor (KAR) and investigated the role of KARs in modulating the development of cultured mouse DRG neurons. We found that KARs modulate neuronal maturation and neurite outgrowth in a bidirectional manner. Accordingly, low concentrations of KA delayed maturation and enhanced neurite outgrowth, whereas maturation was promoted by higher concentrations of KA that attenuated neuritic elongation. The effects of weak KAR activation were prevented by blocking their noncanonical signaling and involved a differential regulation of CRMP2. Whereas the delay in maturation involves PKC-mediated phosphorylation of CRMP2 at T555 leading to a downregulation of membrane Cav2.2, the promotion of neurite outgrowth is achieved by dephosphorylation at T514 at the growth cones, the latter reflecting PKC-driven enhancement of GSK3ß phosphorylation at S9. Together, these findings indicate that noncanonical KAR signaling influences neuronal development by modulating CRMP2 activity.


Asunto(s)
Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Animales Recién Nacidos , Células COS , Chlorocebus aethiops , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica/fisiología
6.
Cell Rep ; : 114427, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38986610

RESUMEN

Kainate (KA)-type glutamate receptors (KARs) are implicated in various neuropsychiatric and neurological disorders through their ionotropic and metabotropic actions. However, compared to AMPA- and NMDA-type receptor functions, many aspects of KAR biology remain incompletely understood. Our study demonstrates an important role of KARs in organizing climbing fiber (CF)-Purkinje cell (PC) synapses and synaptic plasticity in the cerebellum, independently of their ion channel or metabotropic functions. The amino-terminal domain (ATD) of the GluK4 KAR subunit binds to C1ql1, provided by CFs, and associates with Bai3, an adhesion-type G protein-coupled receptor expressed in PC dendrites. Mice lacking GluK4 exhibit no KAR-mediated responses, reduced C1ql1 and Bai3 levels, and fewer CF-PC synapses, along with impaired long-term depression and oculomotor learning. Remarkably, introduction of the ATD of GluK4 significantly improves all these phenotypes. These findings demonstrate that KARs act as synaptic scaffolds, orchestrating synapses by forming a KAR-C1ql1-Bai3 complex in the cerebellum.

7.
Crit Care Nurse ; 43(4): 30-38, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37524366

RESUMEN

BACKGROUND: The risk of medication errors in intensive care units is high, primarily in the drug administration phase. LOCAL PROBLEM: Management of high-alert medications within intensive care units in the study institution varied widely. The aim of this quality improvement project was to protocolize and centralize the management of high-alert medications in acute care settings and to implement smart intravenous infusion pump technology in intensive care units. METHODS: The project was conducted in 4 phases: (1) protocolization and standardization of intravenous mixtures, (2) centralization of intravenous mixture preparation in the Pharmacy Department, (3) programming of the smart pumps, and (4) dissemination and staged implementation of intravenous mixture protocols. Smart pumps (Alaris, CareFusion) were used to deliver the medicines, and the manufacturer's software (Alaris Guardrails, CareFusion) was used to analyze data regarding adherence to the drug library and the number of programming errors detected. RESULTS: Morphine, remifentanil, fentanyl, midazolam, dexmedetomidine, and propofol were included. After implementation of the smart pumps, 3283 infusions were started; of these, 2198 were programmed through the drug library, indicating 67% compliance with the safety software. The pumps intercepted 398 infusion-related programming errors that led to cancellation or reprogramming of drug infusions. CONCLUSIONS: Protocolization and centralization of the preparation of high-alert sedative and analgesic medications for critically ill patients and the administration of these drugs using smart pump technology decrease variability of clinical practice and intercept potentially serious medication errors.


Asunto(s)
Analgesia , Seguridad del Paciente , Humanos , Errores de Medicación/prevención & control , Unidades de Cuidados Intensivos , Cuidados Críticos , Bombas de Infusión , Infusiones Intravenosas
8.
Sci Rep ; 12(1): 14690, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-36038626

RESUMEN

The molecular mechanisms underlying circuit re-wiring in the mature brain remains ill-defined. An eloquent example of adult circuit remodelling is the hippocampal mossy fiber (MF) sprouting found in diseases such as temporal lobe epilepsy. The molecular determinants underlying this retrograde re-wiring remain unclear. This may involve signaling system(s) controlling axon specification/growth during neurodevelopment reactivated during epileptogenesis. Since adenosine A2A receptors (A2AR) control axon formation/outgrowth and synapse stabilization during development, we now examined the contribution of A2AR to MF sprouting. A2AR blockade significantly attenuated status epilepticus(SE)-induced MF sprouting in a rat pilocarpine model. This involves A2AR located in dentate granule cells since their knockdown selectively in dentate granule cells reduced MF sprouting, most likely through the ability of A2AR to induce the formation/outgrowth of abnormal secondary axons found in rat hippocampal neurons. These A2AR should be activated by extracellular ATP-derived adenosine since a similar prevention/attenuation of SE-induced hippocampal MF sprouting was observed in CD73 knockout mice. These findings demonstrate that A2AR contribute to epilepsy-related MF sprouting, most likely through the reactivation of the ability of A2AR to control axon formation/outgrowth observed during neurodevelopment. These results frame the CD73-A2AR axis as a regulator of circuit remodeling in the mature brain.


Asunto(s)
Adenosina , Epilepsia del Lóbulo Temporal , Receptor de Adenosina A2A/metabolismo , Animales , Epilepsia del Lóbulo Temporal/inducido químicamente , Ratones , Fibras Musgosas del Hipocampo , Pilocarpina/farmacología , Ratas , Sinapsis/fisiología
9.
J Neurosci ; 30(1): 242-54, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20053906

RESUMEN

Protein kinase C (PKC) enhances NMDA receptor (NMDAR)-mediated currents and promotes NMDAR delivery to the cell surface via SNARE-dependent exocytosis. Although the mechanisms of PKC potentiation are established, the molecular target of PKC is unclear. Here we show that synaptosomal-associated protein of 25 kDa (SNAP-25), a SNARE protein, is functionally relevant to PKC-dependent NMDAR insertion, and identify serine residue-187 as the molecular target of PKC phosphorylation. Constitutively active PKC delivered via the patch pipette potentiated NMDA (but not AMPA) whole-cell currents in hippocampal neurons. Expression of RNAi targeting SNAP-25 or mutant SNAP-25(S187A) and/or acute disruption of the SNARE complex by treatment with BoNT A, BoNT B or SNAP-25 C-terminal blocking peptide abolished NMDAR potentiation. A SNAP-25 peptide and function-blocking antibody suppressed PKC potentiation of NMDA EPSCs at mossy fiber-CA3 synapses. These findings identify SNAP-25 as the target of PKC phosphorylation critical to PKC-dependent incorporation of synaptic NMDARs and document a postsynaptic action of this major SNARE protein relevant to synaptic plasticity.


Asunto(s)
Marcación de Gen , Proteína Quinasa C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Fosforilación , Unión Proteica/fisiología , Proteína Quinasa C/genética , Transporte de Proteínas/fisiología , Ratas , Receptores de N-Metil-D-Aspartato/genética , Proteína 25 Asociada a Sinaptosomas/genética , Xenopus laevis
10.
EMBO J ; 26(20): 4359-67, 2007 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17898803

RESUMEN

Agonists of kainate receptors (KARs) cause both the opening of the associated ion channels and the activation of signalling pathways driven by G-proteins and PKC. Here we report the existence of an unknown mechanism of KAR autoregulation, involving the interplay of this two signalling mechanisms. Repetitive activation of native KARs evoked the rundown of the ionotropic responses in a manner that was dependent on the activation of PKC. Experiments on recombinant GluR5 expressed in neuroblastoma cells indicated that KARs trigger the activation of PKC and induce the internalization of membrane receptors. This phenomenon depends on the PKC-mediated phosphorylation of serines 879 and 885 of the GluR5-2b subunits, since mutation of these two residues abolished internalization. These results reveal that the non-canonical signalling of KARs is associated with a sensitive mechanism that detects afferent activity. Such a mechanism represents an active way to limit overactivation of the KAR system, by regulating the number of KARs in the cell membrane.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteína Quinasa C/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Calcio/metabolismo , Línea Celular Tumoral , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Proteínas de Unión al GTP/metabolismo , Humanos , Ratones , Modelos Biológicos , Neuronas/metabolismo , Ratas , Transducción de Señal
11.
Neuropharmacology ; 191: 108558, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33862031

RESUMEN

Cognition and behavior are tightly linked to synaptic function. A growing body of evidence suggests that aberrant neurotransmission, caused by changes in synaptic protein expression levels, may be a major cause underlying different brain disorders. These changes in expression result in abnormal synaptic organization or function, leading to impaired neurotransmission and unbalanced circuit operations. Here, we review the data supporting the involvement of mutations in genes coding for kainate receptor (KAR) subunits in the pathogenesis of psychiatric disorders and Down syndrome (DS). We show that most of these mutations do not affect the biophysical properties or the receptors, but rather alter subunit expression levels. On the basis of reports studying KAR genes mutations in mouse models of autism spectrum disorders and DS, we illustrate how deviations from the physiological regulatory role that these receptors play in neurotransmitter release and plasticity give rise to synaptic alterations that lead to behavioral and cognitive deficits underlying these disorders.


Asunto(s)
Síndrome de Down/fisiopatología , Trastornos Mentales/fisiopatología , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/fisiología , Animales , Síndrome de Down/genética , Síndrome de Down/metabolismo , Humanos , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Ratones , Plasticidad Neuronal , Receptores de Ácido Kaínico/metabolismo , Transmisión Sináptica
12.
Neuroscience ; 456: 17-26, 2021 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31866560

RESUMEN

Extensive research over the past decades has characterized multiple forms of synaptic plasticity, identifying them as key processes that allow the brain to operate in a dynamic manner. Within the wide variety of synaptic plasticity modulators, kainate receptors are receiving increasing attention, given their diversity of signaling mechanisms and cellular expression profile. Here, we summarize the experimental evidence about the involvement of kainate receptor signaling in the regulation of short- and long-term plasticity, from the perspective of the regulation of neurotransmitter release. In light of this evidence, we propose that kainate receptors may be considered homeostatic modulators of neurotransmitter release, able to bidirectionally regulate plasticity depending on the functional history of the synapse.


Asunto(s)
Plasticidad Neuronal , Receptores de Ácido Kaínico , Homeostasis , Potenciación a Largo Plazo , Receptores de Ácido Kaínico/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
13.
Neuron ; 52(6): 1037-46, 2006 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17178406

RESUMEN

A prominent feature of ionotropic glutamate receptors from the AMPA and kainate subtypes is their profound desensitization in response to glutamate-a process thought to protect the neuron from overexcitation. In AMPA receptors, it is well established that desensitization results from rearrangements of the interface formed between agonist-binding domains of adjacent subunits; however, it is unclear how this mechanism applies to kainate receptors. Here we show that stabilization of the binding domain dimer by the generation of intermolecular disulfide bonds apparently blocked desensitization of the kainate receptor GluR6. This result establishes a common desensitization mechanism in both AMPA and kainate receptors. Surprisingly, however, surface expression of these nondesensitizing mutants was drastically reduced and did not depend on channel activity. Therefore, in addition to its role at the synapse, we now propose an intracellular role for desensitization in controlling maturation and trafficking of glutamate receptors.


Asunto(s)
Receptores de Ácido Kaínico/fisiología , Animales , Células Cultivadas , Cisteína/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Ácido Kaínico/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Modelos Biológicos , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oocitos , Técnicas de Placa-Clamp/métodos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Quinoxalinas/farmacología , Receptores AMPA/fisiología , Receptores de Ácido Kaínico/química , Relación Estructura-Actividad , Transfección/métodos , Xenopus , Receptor de Ácido Kaínico GluK2
14.
J Neurosci Methods ; 343: 108814, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32569785

RESUMEN

Preclinical MRI approaches constitute a key tool to study a wide variety of neurological and psychiatric illnesses, allowing a more direct investigation of the disorder substrate and, at the same time, the possibility of back-translating such findings to human subjects. However, the lack of consensus on the optimal experimental scheme used to acquire the data has led to relatively high heterogeneity in the choice of protocols, which can potentially impact the comparison between results obtained by different groups, even using the same animal model. This is especially true for diffusion-weighted MRI data, where certain experimental choices can impact not only on the accuracy and precision of the extracted biomarkers, but also on their biological meaning. With this in mind, we extensively examined preclinical imaging studies that used diffusion-weighted MRI to investigate neurodegenerative, neurodevelopmental and psychiatric disorders in rodent models. In this review, we discuss the main findings for each preclinical model, with a special focus on the analysis and comparison of the different acquisition strategies used across studies and their impact on the heterogeneity of the findings.


Asunto(s)
Imagen de Difusión por Resonancia Magnética , Imagen por Resonancia Magnética , Animales , Biomarcadores , Modelos Animales de Enfermedad , Roedores
15.
Neuron ; 48(2): 279-88, 2005 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16242408

RESUMEN

AMPA-type glutamate receptors are specifically inhibited by the noncompetitive antagonists GYKI-53655 and CP-465,022, which act through sites and mechanisms that are not understood. Using receptor mutagenesis, we found that these antagonists bind at the interface between the S1 and S2 glutamate binding core and channel transmembrane domains, specifically interacting with S1-M1 and S2-M4 linkers, thereby disrupting the transduction of agonist binding into channel opening. We also found that the antagonists' affinity is higher for agonist-unbound receptors than for activated nondesensitized receptors, further depending on the level of S1 and S2 domain closure. These results provide evidence for substantial conformational changes in the S1-M1 and S2-M4 linkers following agonist binding and channel opening, offering a conceptual frame to account for noncompetitive antagonism of AMPA receptors.


Asunto(s)
Benzodiazepinas/farmacología , Potenciales de la Membrana/fisiología , Quinazolinas/farmacología , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/fisiología , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Western Blotting/métodos , Encéfalo/efectos de los fármacos , Línea Celular , Clonación Molecular/métodos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Modelos Neurológicos , Mutagénesis/fisiología , Técnicas de Placa-Clamp/métodos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Conformación Proteica , Quinazolinas/farmacocinética , Ratas , Receptores AMPA/química , Transfección/métodos , Tritio/farmacocinética , Xenopus laevis
16.
Adicciones ; 21(3): 195-202, 2009.
Artículo en Español | MEDLINE | ID: mdl-19718490

RESUMEN

OBJECTIVE: To assess the metric properties of the abridged, 10-item version of the Cocaine Craving Questionnaire-Now (CCQ-N-10) in its Spanish adaptation. METHOD: The sample consists of 63 people beginning treatment for cocaine-abuse (n=23) or dependence (n=40) disorders according to DSM-IV criteria. Participants completed an assessment battery that included the extended, 45-item version of the Cocaine Craving Questionnaire (CCQ-N-45) - in which the CCQ-N-10 is embedded -, the Cocaine Craving Scale (CCS), a Visual Analog Craving Scale (VAS), the Severity of Dependence Scale (SDS), and the Clinical Psychiatric Impression (CPI). RESULTS: Cronbach's values for the CCQ-N-10, CCQ-N-45, SDS and CCS scales were 0.95, 0.88, 0.79 and 0.93, respectively, indicating that all instruments show high internal consistency. Test-retest reliability for the CCQ-N-10 was fair (Intraclass Correlation Coefficient=0.59; p<0.001). A factor analysis of the CCQ-N-10 (KMO=0.85) shows a one-factor structure explaining 68.6% of the variance, with factor loadings ranging from 0.64 to 0.92. Confirmatory factor analysis supports the unidimensionality of the scale (CF =0.91; SRMR=0.06). The CCQ-N-10 explains 86.6% of the variance accounted for by the CCQ-N extended version (CCQ-N-45) and shows convergent validity with related measures such as the CCS (r=0.64, p<0.001), VAS (r=0.65, p<0.001), SDS (r=0.53, p<0.001) and CPI (r=0.50, p<0.001). Persons fulfilling criteria for cocaine-dependence disorder, compared to those who present cocaine abuse, show higher CCQ-N-10 scores (F(1;60.6)=7.16, p=0.010). CONCLUSIONS: The Spanish adaptation of the CCQ-N-10 shows metric adequacy, and can be considered as a suitable assessment instrument in relation to cocaineuse disorders.


Asunto(s)
Trastornos Relacionados con Cocaína/diagnóstico , Trastornos Relacionados con Cocaína/psicología , Encuestas y Cuestionarios , Adulto , Femenino , Humanos , Lenguaje , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Adulto Joven
17.
Nat Commun ; 10(1): 4991, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676751

RESUMEN

Overinhibition is assumed one of the main causes of cognitive deficits (e.g. memory impairment) in mouse models of Down syndrome (DS). Yet the mechanisms that drive such exaggerated synaptic inhibition and their behavioral effects remain unclear. Here we report the existence of bidirectional alterations to the synaptic inhibition on CA1 pyramidal cells in the Ts2Cje mouse model of DS which are associated to impaired spatial memory. Furthermore, we identify triplication of the kainate receptor (KAR) encoding gene Grik1 as the cause of these phenotypes. Normalization of Grik1 dosage in Ts2Cje mice specifically restored spatial memory and reversed the bidirectional alterations to CA1 inhibition, but not the changes in synaptic plasticity or the other behavioral modifications observed. We propose that modified information gating caused by disturbed inhibitory tone rather than generalized overinhibition underlies some of the characteristic cognitive deficits in DS.


Asunto(s)
Región CA1 Hipocampal/fisiología , Dendritas/fisiología , Síndrome de Down/fisiopatología , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Memoria Espacial/fisiología , Animales , Región CA1 Hipocampal/citología , Modelos Animales de Enfermedad , Síndrome de Down/patología , Femenino , Humanos , Masculino , Ratones
18.
ACS Chem Neurosci ; 10(11): 4685-4695, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31622082

RESUMEN

Selective pharmacological tool compounds are invaluable for understanding the functions of the various ionotropic glutamate receptor subtypes. For the kainate receptors, these compounds are few. Here we have synthesized nine novel quinoxaline-2,3-diones with substitutions in the 7-position to investigate the structure-activity relationship at kainate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Compound 11 exhibited the highest binding affinity across GluK1-3 while having selectivity toward kainate vs AMPA receptors. Compound 11 potently inhibited glutamate evoked currents at homomeric GluK1 and GluK3 receptors in HEK293 cells with Kb values of 65 and 39 nM, respectively. The binding mode of 11 in the ligand binding domain of GluK1 was investigated by X-ray crystallography, revealing that 11 stabilizes the receptor in an open conformation, consistent with its demonstrated antagonism. Furthermore, 11 was tested for analgesic effects in the mouse tail flick test where it significantly increased tail flick latency at doses where 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]-quinoxaline-7-sulfonamide (NBQX) was ineffective.


Asunto(s)
Analgésicos/metabolismo , Cristalografía por Rayos X/métodos , Antagonistas de Aminoácidos Excitadores/metabolismo , Quinoxalinas/metabolismo , Receptores de Ácido Kaínico/antagonistas & inhibidores , Receptores de Ácido Kaínico/metabolismo , Analgésicos/química , Analgésicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/química , Antagonistas de Aminoácidos Excitadores/farmacología , Células HEK293 , Humanos , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Quinoxalinas/química , Quinoxalinas/farmacología , Ratas , Receptores de Ácido Kaínico/química , Relación Estructura-Actividad
20.
Neuron ; 39(3): 543-53, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12895426

RESUMEN

The potent neurotoxin kainate activates ion channel-forming receptors. However, it can also activate a G protein-coupled signaling pathway to inhibit transmitter release in central neurons. It remains unclear whether the same receptor complex is involved in both signaling activities. Here we show that in a population of dorsal root ganglion cells, exposure to kainate elicits a G protein-dependent increase in intracellular Ca2+. Furthermore, in these cells a brief exposure to kainate inhibited the K+-induced Ca2+ increase, a process that was sensitive to the G protein inhibitor Pertussis toxin and inhibitors of protein kinase C. This metabotropic action did not require ion channel activity and was not observed in neurons prepared from mice deficient for the ion channel-forming subunit GluR5. These results indicate that GluR5, an ion channel-forming subunit, signals through a second messenger cascade, inhibiting voltage-dependent Ca2+ channels. Thus, such a system represents a noncanonical signaling route of ion channel-forming receptors.


Asunto(s)
Receptores de Ácido Kaínico/fisiología , Transducción de Señal/fisiología , Animales , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA