Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Neurosci ; 42(47): 8897-8911, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36202617

RESUMEN

Metabotropic glutamate receptor Type 3 (mGlu3) controls the sleep/wake architecture, which plays a role in the glutamatergic pathophysiology of schizophrenia. Interestingly, mGlu3 receptor expression is decreased in the brain of schizophrenic patients. However, little is known about the molecular mechanisms regulating mGlu3 receptors at the cell membrane. Subcellular receptor localization is strongly dependent on protein-protein interactions. Here we show that mGlu3 interacts with PICK1 and that this scaffolding protein is important for mGlu3 surface expression and function in hippocampal primary cultures. Disruption of their interaction via an mGlu3 C-terminal mimicking peptide or an inhibitor of the PDZ domain of PICK1 altered the functional expression of mGlu3 receptors in neurons. We next investigated the impact of disrupting the mGlu3-PICK1 interaction on hippocampal theta oscillations in vitro and in vivo in WT male mice. We found a decreased frequency of theta oscillations in organotypic hippocampal slices, similar to what was previously observed in mGlu3 KO mice. In addition, hippocampal theta power was reduced during rapid eye movement sleep, non-rapid eye movement (NREM) sleep, and wake states after intraventricular administration of the mGlu3 C-terminal mimicking peptide. Targeting the mGlu3-PICK1 complex could thus be relevant to the pathophysiology of schizophrenia.SIGNIFICANCE STATEMENT Dysregulation of the glutamatergic system might play a role in the pathophysiology of schizophrenia. Metabotropic glutamate receptors Type 3 (mGlu3) have been proposed as potential targets for schizophrenia. Understanding the molecular mechanisms regulating mGlu3 receptor at the cell membrane is critical toward comprehending how their dysfunction contributes to the pathogenesis of schizophrenia. Here we describe that the binding of the signaling and scaffolding protein PICK1 to mGlu3 receptors is important for their localization and physiological functions. The identification of new proteins that associate specifically to mGlu3 receptors will advance our understanding of the regulatory mechanisms associated with their targeting and function and ultimately might provide new therapeutic strategies to counter these psychiatric conditions.


Asunto(s)
Proteínas Portadoras , Hipocampo , Receptores de Glutamato Metabotrópico , Animales , Masculino , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Hipocampo/metabolismo , Dominios PDZ , Receptores de Glutamato Metabotrópico/metabolismo
2.
Addict Biol ; 26(4): e12995, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33368923

RESUMEN

Prescription stimulants, such as d-amphetamine or methylphenidate are used to treat suffering from attention-deficit hyperactivity disorder (ADHD). They potently release dopamine (DA) and norepinephrine (NE) and cause phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 in the striatum. Whether other brain regions are also affected remains elusive. Here, we demonstrate that d-amphetamine and methylphenidate increase phosphorylation at Ser845 (pS845-GluA1) in the membrane fraction of mouse cerebellum homogenate. We identify Bergmann glial cells as the source of pS845-GluA1 and demonstrate a requirement for intact NE release. Consequently, d-amphetamine-induced pS845-GluA1 was prevented by ß1-adenoreceptor antagonist, whereas the blockade of DA D1 receptor had no effect. Together, these results indicate that NE regulates GluA1 phosphorylation in Bergmann glial cells in response to prescription stimulants.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Cerebelo/metabolismo , Dextroanfetamina/farmacología , Metilfenidato/farmacología , Fosfotransferasas , Animales , Masculino , Ratones , Norepinefrina/metabolismo , Fosforilación , Receptores de Dopamina D1/metabolismo
3.
J Neurochem ; 151(2): 204-226, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31245856

RESUMEN

The caudal part of the striatum, also named the tail of the striatum (TS), defines a fourth striatal domain. Determining whether rewarding, aversive and salient stimuli regulate the activity of striatal spiny projections neurons (SPNs) of the TS is therefore of paramount importance to understand its functions, which remain largely elusive. Taking advantage of genetically encoded biosensors (A-kinase activity reporter 3) to record protein kinase A signals and by analyzing the distribution of dopamine D1R- and D2R-SPNs in the TS, we characterized three subterritories: a D2R/A2aR-lacking, a D1R/D2R-intermingled and a D1R/D2R-SPNs-enriched area (corresponding to the amygdalostriatal transition). In addition, we provide evidence that the distribution of D1R- and D2R-SPNs in the TS is evolutionarily conserved (mouse, rat, gerbil). The in vivo analysis of extracellular signal-regulated kinase (ERK) phosphorylation in these TS subterritories in response to distinct appetitive, aversive and pharmacological stimuli revealed that SPNs of the TS are not recruited by stimuli triggering innate aversive responses, fasting, satiety, or palatable signals whereas a reduction in ERK phosphorylation occurred following learned avoidance. In contrast, D1R-SPNs of the intermingled and D2R/A2aR-lacking areas were strongly activated by both D1R agonists and psychostimulant drugs (d-amphetamine, cocaine, 3,4-methyl enedioxy methamphetamine, or methylphenidate), but not by hallucinogens. Finally, a similar pattern of ERK activation was observed by blocking selectively dopamine reuptake. Together, our results reveal that the caudal TS might participate in the processing of specific reward signals and discrete aversive stimuli. Cover Image for this issue: doi: 10.1111/jnc.14526. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Asunto(s)
Reacción de Prevención/fisiología , Cuerpo Estriado/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Recompensa , Estimulación Acústica/efectos adversos , Animales , Reacción de Prevención/efectos de los fármacos , Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Gerbillinae , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
4.
Neurobiol Dis ; 129: 13-28, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31051234

RESUMEN

Finding new targets to control or reduce seizure activity is essential to improve the management of epileptic patients. We hypothesized that activation of the pre-synaptic and inhibitory metabotropic glutamate receptor type 7 (mGlu7) reduces spontaneous seizures. We tested LSP2-9166, a recently developed mGlu7/4 agonist with unprecedented potency on mGlu7 receptors, in two paradigms of epileptogenesis. In a model of chemically induced epileptogenesis (pentylenetetrazole systemic injection), LSP2-9166 induces an anti-epileptogenic effect rarely observed in preclinical studies. In particular, we found a bidirectional modulation of seizure progression by mGlu4 and mGlu7 receptors, the latter preventing kindling. In the intra-hippocampal injection of kainic acid mouse model that mimics the human mesial temporal lobe epilepsy, we found that LSP2-9166 reduces seizure frequency and hippocampal sclerosis. LSP2-9166 also acts as an anti-seizure drug on established seizures in both models tested. Specific modulation of the mGlu7 receptor could represent a novel approach to reduce pathological network remodeling.


Asunto(s)
Aminobutiratos/farmacología , Anticonvulsivantes/farmacología , Hipocampo/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Convulsiones/metabolismo , Animales , Epilepsia/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/metabolismo , Excitación Neurológica/efectos de los fármacos , Ratones , Ratones Mutantes
5.
Stem Cells ; 36(2): 192-205, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29044892

RESUMEN

One strategy for stem cell-based therapy of the cerebral cortex involves the generation and transplantation of functional, histocompatible cortical-like neurons from embryonic stem cells (ESCs). Diploid parthenogenetic Pg-ESCs have recently emerged as a promising source of histocompatible ESC derivatives for organ regeneration but their utility for cerebral cortex therapy is unknown. A major concern with Pg-ESCs is genomic imprinting. In contrast with biparental Bp-ESCs derived from fertilized oocytes, Pg-ESCs harbor two maternal genomes but no sperm-derived genome. Pg-ESCs are therefore expected to have aberrant expression levels of maternally expressed (MEGs) and paternally expressed (PEGs) imprinted genes. Given the roles of imprinted genes in brain development, tissue homeostasis and cancer, their deregulation in Pg-ESCs might be incompatible with therapy. Here, we report that, unexpectedly, only one gene out of 7 MEGs and 12 PEGs was differentially expressed between Pg-ESCs and Bp-ESCs while 13 were differentially expressed between androgenetic Ag-ESCs and Bp-ESCs, indicating that Pg-ESCs but not Ag-ESCs, have a Bp-like imprinting compatible with therapy. In vitro, Pg-ESCs generated cortical-like progenitors and electrophysiologically active glutamatergic neurons that maintained the Bp-like expression levels for most imprinted genes. In vivo, Pg-ESCs participated to the cortical lineage in fetal chimeras. Finally, transplanted Pg-ESC derivatives integrated into the injured adult cortex and sent axonal projections in the host brain. In conclusion, mouse Pg-ESCs generate functional cortical-like neurons with Bp-like imprinting and their derivatives properly integrate into both the embryonic cortex and the injured adult cortex. Collectively, our data support the utility of Pg-ESCs for cortical therapy. Stem Cells 2018;36:192-205.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Animales , Metilación de ADN/genética , Metilación de ADN/fisiología , Electrofisiología , Impresión Genómica/genética , Impresión Genómica/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Partenogénesis/genética , Partenogénesis/fisiología
6.
Neurobiol Dis ; 113: 70-81, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432809

RESUMEN

BACKGROUND: Cerebrovascular dysfunction and inflammation occur in epilepsy. Here we asked whether pericytes, a pivotal cellular component of brain capillaries, undergo pathological modifications during experimental epileptogenesis and in human epilepsy. We evaluated whether pro-inflammatory cytokines, present in the brain during seizures, contribute to pericyte morphological modifications. METHODS: In vivo, unilateral intra-hippocampal kainic acid (KA) injections were performed in NG2DsRed/C57BL6 mice to induce status epilepticus (SE), epileptogenesis, and spontaneous recurrent seizures (SRS). NG2DsRed mice were used to visualize pericytes during seizure progression. The effect triggered by recombinant IL-1ß, TNFα, or IL-6 on pericytes was evaluated in NG2DsRed hippocampal slices and in human-derived cell culture. Human brain specimens obtained from temporal lobe epilepsy (TLE) with or without sclerosis (HS) and focal cortical dysplasia (FCD-IIb) were evaluated for pericyte-microglial cerebrovascular assembly. RESULTS: A disarray of NG2DsRed+ pericyte soma and ramifications was found 72 h post-SE and 1 week post-SE (epileptogenesis) in the hippocampus. Pericyte modifications topographically overlapped with IBA1+ microglia clustering around the capillaries with cases of pericytes lodged within the microglial cells. Microglial clustering around the NG2DsRed pericytes lingered at SRS. Pericyte proliferation (Ki67+) occurred 72 h post-SE and during epileptogenesis and returned towards control levels at SRS. Human epileptic brain tissues showed pericyte-microglia assemblies with IBA1/HLA microglial cells outlining the capillary wall in TLE-HS and FCD-IIb specimens. Inflammatory mediators contributed to pericyte modifications, in particular IL-1ß elicited pericyte morphological changes and pericyte-microglia clustering in NG2DsRed hippocampal slices. Modifications also occurred when pro-inflammatory cytokines were added to an in vitro culture of pericytes. CONCLUSIONS: These results indicate the occurrence of pericytosis during seizures and introduce a pericyte-microglial mediated mechanism of blood-brain barrier dysfunction in epilepsy.


Asunto(s)
Circulación Cerebrovascular/fisiología , Progresión de la Enfermedad , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Pericitos/metabolismo , Convulsiones/metabolismo , Adolescente , Adulto , Animales , Barrera Hematoencefálica/química , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Células Cultivadas , Niño , Preescolar , Femenino , Hipocampo/irrigación sanguínea , Hipocampo/química , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/química , Pericitos/química , Convulsiones/fisiopatología
7.
Epilepsia ; 59(1): 123-134, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29125184

RESUMEN

OBJECTIVE: Available evidence points to a role of cytochrome P450 (Cyp) drug biotransformation enzymes in central nervous system diseases, including epilepsy. Deviations in drug pharmacokinetic profiles may impact therapeutic outcomes. Here, we ask whether spontaneous recurrent seizure (SRS) activity is sufficient to modulate the expression of major Cyp enzymes in the liver and brain. METHODS: Unilateral intrahippocampal (IH) kainic acid (KA) injections were used to elicit nonconvulsive status epilepticus (SE), epileptogenesis, and SRS, as monitored by video-electroencephalography. Intraperitoneal (IP) KA injection was used to trigger generalized tonic-clonic SE. KA-injected mice and sham controls were sacrificed at 24-72 hours and 1 week post-SE (IH or IP KA), and during the chronic stage (SRS; 6 weeks post-IH KA). Liver and brain tissues were processed for histology, real-time quantitative polymerase chain reaction, Western blot, or microsomal enzymatic assay. Cyp2e1, Cyp3a13, glial fibrillary acidic protein (GFAP), IBA1, xenobiotic nuclear receptors nr1i2 (PXR), nr1i3 (CAR) and nr3c1 (glucocorticoid receptor [GR]) expression was examined. Serum samples were obtained to assay corticosterone levels, a GR activator. RESULTS: A significant increase of Cyp3a13 and Cyp2e1 transcript level and protein expression was found in the liver and hippocampi during SRS, as compared to control mice. In the ipsilateral hippocampus, Cyp2e1 and Cyp3a protein upregulation during SRS positively correlated to GFAP expression. GFAP+ , and not IBA1+ , cells colocalized with Cyp2e1 or Cyp3a expression. In the liver, a trend increase in Cyp3a microsomal activity was found during SRS as compared to control mice. The transcript levels of the Cyp upstream regulators GR, xenobiotic nr1i2, and nr1i3 receptors were unchanged at SRS. Corticosterone levels, a GR ligand, were increased in the blood post-SE. SIGNIFICANCE: SRS modifies Cyp expression in the liver and the hippocampus. Nuclear receptors or inflammatory pathways are candidate mechanisms of Cyp regulation during seizures.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Hipocampo/enzimología , Hígado/enzimología , Estado Epiléptico/enzimología , Estado Epiléptico/patología , Animales , Proteínas de Unión al Calcio/metabolismo , Receptor de Androstano Constitutivo , Corticosterona/sangre , Sistema Enzimático del Citocromo P-450/genética , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Agonistas de Aminoácidos Excitadores/toxicidad , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Recurrencia , Estadísticas no Paramétricas , Estado Epiléptico/sangre , Estado Epiléptico/inducido químicamente , Factores de Tiempo
8.
Neurobiol Dis ; 106: 110-123, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28688851

RESUMEN

Episodic Ataxia type 2 (EA2) is an autosomal dominant neuronal disorder linked to mutations in the Cav2.1 subunit of P/Q-type calcium channels. In vitro studies have established that EA2 mutations induce loss of channel activity and that EA2 mutants can exert a dominant negative effect, suppressing normal Cav2.1 activity through protein misfolding and trafficking defects. To date, the role of this mechanism in the disease pathogenesis is unknown because no animal model exists. To address this issue, we have generated a mouse bearing the R1497X nonsense mutation in Cav2.1 (Cav2.1R1497X). Phenotypic analysis of heterozygous Cav2.1R1497X mice revealed ataxia associated with muscle weakness and generalized absence epilepsy. Electrophysiological studies of the cerebellar circuits in heterozygous Cav2.1R1497X mice highlighted severe dysregulations in synaptic transmission of the two major excitatory inputs as well as alteration of the spontaneous activity of Purkinje cells. Moreover, these neuronal dysfunctions were associated with a strong suppression of Cav2.1 channel expression in the cerebellum of heterozygous Cav2.1R1497X mice. Finally, the presence of Cav2.1 in cerebellar lipid raft microdomains was strongly impaired in heterozygous Cav2.1R1497X mice. Altogether, these results reveal a pathogenic mechanism for EA2 based on a dominant negative activity of mutant channels.


Asunto(s)
Ataxia/genética , Ataxia/metabolismo , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Cerebelo/metabolismo , Neuronas/metabolismo , Nistagmo Patológico/genética , Nistagmo Patológico/metabolismo , Animales , Ataxia/patología , Cerebelo/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Genes Dominantes , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Debilidad Muscular/genética , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Neuronas/patología , Nistagmo Patológico/patología , Fenotipo , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Sinapsis/metabolismo , Técnicas de Cultivo de Tejidos
9.
J Neurosci ; 35(7): 3155-73, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25698751

RESUMEN

The charged multivesicular body proteins (Chmp1-7) are an evolutionarily conserved family of cytosolic proteins that transiently assembles into helical polymers that change the curvature of cellular membrane domains. Mutations in human CHMP2B cause frontotemporal dementia, suggesting that this protein may normally control some neuron-specific process. Here, we examined the function, localization, and interactions of neuronal Chmp2b. The protein was highly expressed in mouse brain and could be readily detected in neuronal dendrites and spines. Depletion of endogenous Chmp2b reduced dendritic branching of cultured hippocampal neurons, decreased excitatory synapse density in vitro and in vivo, and abolished activity-induced spine enlargement and synaptic potentiation. To understand the synaptic effects of Chmp2b, we determined its ultrastructural distribution by quantitative immuno-electron microscopy and its biochemical interactions by coimmunoprecipitation and mass spectrometry. In the hippocampus in situ, a subset of neuronal Chmp2b was shown to concentrate beneath the perisynaptic membrane of dendritic spines. In synaptoneurosome lysates, Chmp2b was stably bound to a large complex containing other members of the Chmp family, as well as postsynaptic scaffolds. The supramolecular Chmp assembly detected here corresponds to a stable form of the endosomal sorting complex required for transport-III (ESCRT-III), a ubiquitous cytoplasmic protein complex known to play a central role in remodeling of lipid membranes. We conclude that Chmp2b-containing ESCRT-III complexes are also present at dendritic spines, where they regulate synaptic plasticity. We propose that synaptic ESCRT-III filaments may function as a novel element of the submembrane cytoskeleton of spines.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Sinapsis/fisiología , Animales , Células Cultivadas , Simulación por Computador , Dendritas/metabolismo , Dendritas/ultraestructura , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Hipocampo/citología , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Mutación/genética , N-Metilaspartato/farmacología , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/ultraestructura , Densidad Postsináptica/metabolismo , Densidad Postsináptica/ultraestructura , Ratas , Ratas Sprague-Dawley , Sinapsis/ultraestructura , Proteína Fluorescente Roja
10.
J Biol Chem ; 289(5): 2600-9, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24352656

RESUMEN

Development of dendritic spines is important for synaptic function, and alteration in spine morphogenesis is often associated with mental disorders. Rich2 was an uncharacterized Rho-GAP protein. Here we searched for a role of this protein in spine morphogenesis. We found that it is enriched in dendritic spines of cultured hippocampal pyramidal neurons during early stages of development. Rich2 specifically stimulated the Rac1 GTPase in these neurons. Inhibition of Rac1 by EHT 1864 increased the size and decreased the density of dendritic spines. Similarly, Rich2 overexpression increased the size and decreased the density of dendritic spines, whereas knock-down of the protein by specific si-RNA decreased both size and density of spines. The morphological changes were reflected by the increased amplitude and decreased frequency of miniature EPSCs induced by Rich2 overexpression, while si-RNA treatment decreased both amplitude and frequency of these events. Finally, treatment of neurons with EHT 1864 rescued the phenotype induced by Rich2 knock-down. These results suggested that Rich2 controls dendritic spine morphogenesis and function via inhibition of Rac1.


Asunto(s)
Espinas Dendríticas/enzimología , Proteínas Activadoras de GTPasa/metabolismo , Neuronas/enzimología , Neuropéptidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Células COS , Chlorocebus aethiops , Potenciales Postsinápticos Excitadores/fisiología , Proteínas Activadoras de GTPasa/genética , Hipocampo/citología , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Ratones , Morfogénesis/fisiología , Neurogénesis/fisiología , Neuronas/ultraestructura , Neuropéptidos/genética , Técnicas de Placa-Clamp , Cultivo Primario de Células , Proteína de Unión al GTP rac1/genética
11.
Eur J Neurosci ; 41(2): 157-67, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25377770

RESUMEN

Type 1 metabotropic glutamate (mGlu1) receptors play a pivotal role in different forms of synaptic plasticity in the cerebellar cortex, e.g. long-term depression at glutamatergic synapses and rebound potentiation at GABAergic synapses. These various forms of plasticity might depend on the subsynaptic arrangement of the receptor in Purkinje cells that can be regulated by protein-protein interactions. This study investigated, by means of the freeze-fracture replica immunogold labelling method, the subcellular localization of mGlu1 receptors in the rodent cerebellum and whether Homer proteins regulate their subsynaptic distribution. We observed a widespread extrasynaptic localization of mGlu1 receptors and confirmed their peri-synaptic enrichment at glutamatergic synapses. Conversely, we detected mGlu1 receptors within the main body of GABAergic synapses onto Purkinje cell dendrites. Although Homer proteins are known to interact with the mGlu1 receptor C-terminus, we could not detect Homer3, the most abundant Homer protein in the cerebellar cortex, at GABAergic synapses by pre-embedding and post-embedding immunoelectron microscopy. We then hypothesized a critical role for Homer proteins in the peri-junctional localization of mGlu1 receptors at glutamatergic synapses. To disrupt Homer-associated protein complexes, mice were tail-vein injected with the membrane-permeable dominant-negative TAT-Homer1a. Freeze-fracture replica immunogold labelling analysis showed no significant alteration in the mGlu1 receptor distribution pattern at parallel fibre-Purkinje cell synapses, suggesting that other scaffolding proteins are involved in the peri-synaptic confinement. The identification of interactors that regulate the subsynaptic localization of the mGlu1 receptor at neurochemically distinct synapses may offer new insight into its trafficking and intracellular signalling.


Asunto(s)
Corteza Cerebelosa/metabolismo , Ácido Glutámico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Western Blotting , Proteínas Portadoras/metabolismo , Corteza Cerebelosa/ultraestructura , Proteínas de Andamiaje Homer , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Células de Purkinje/metabolismo , Células de Purkinje/ultraestructura , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Sinapsis/ultraestructura
12.
J Neurosci ; 33(23): 9699-715, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23739967

RESUMEN

Synaptic long-term potentiation (LTP) is a key mechanism involved in learning and memory, and its alteration is associated with mental disorders. Shank3 is a major postsynaptic scaffolding protein that orchestrates dendritic spine morphogenesis, and mutations of this protein lead to mental retardation and autism spectrum disorders. In the present study we investigated the role of a new Shank3-associated protein in LTP. We identified the Rho-GAP interacting CIP4 homolog 2 (Rich2) as a new Shank3 partner by proteomic screen. Using single-cell bioluminescence resonance energy transfer microscopy, we found that Rich2-Shank3 interaction is increased in dendritic spines of mouse cultured hippocampal neurons during LTP. We further characterized Rich2 as an endosomal recycling protein that controls AMPA receptor GluA1 subunit exocytosis and spine morphology. Knock-down of Rich2 with siRNA, or disruption of the Rich2-Shank3 complex using an interfering mimetic peptide, inhibited the dendritic spine enlargement and the increase in GluA1 subunit exocytosis typical of LTP. These results identify Rich2-Shank3 as a new postsynaptic protein complex involved in synaptic plasticity.


Asunto(s)
Exocitosis/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Potenciación a Largo Plazo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Espinas Dendríticas/metabolismo , Femenino , Proteínas Activadoras de GTPasa/genética , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Proteínas de Microfilamentos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Técnicas de Cultivo de Órganos , Unión Proteica/fisiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
13.
Neurobiol Dis ; 68: 47-56, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24768804

RESUMEN

Episodic ataxia type-2 (EA2) is a dominantly inherited human neurological disorder caused by loss of function mutations in the CACNA1A gene, which encodes the CaV2.1 subunit of P/Q-type voltage-gated calcium channels. It remains however unknown whether the deficit of cerebellar CaV2.1 in adult is in direct link with the disease. To address this issue, we have used lentiviral based-vector RNA interference (RNAi) to knock-down CaV2.1 expression in the cerebellum of adult mice. We show that suppression of the P/Q-type channels in Purkinje neurons induced motor abnormalities, such as imbalance and ataxic gait. Interestingly, moderate channel suppression caused no basal ataxia, while ß-adrenergic activation and exercise mimicked stress induced motor disorders. Moreover, stress-induced ataxia was stable, non-progressive and totally abolished by acetazolamide, a carbonic anhydrase inhibitor used to treat EA2. Altogether, these data reveal that P/Q-type channel suppression in adult mice supports the episodic status of EA2 disease.


Asunto(s)
Ataxia/etiología , Canales de Calcio Tipo N/metabolismo , Cerebelo/patología , Nistagmo Patológico/etiología , Células de Purkinje/metabolismo , Interferencia de ARN/fisiología , ARN Interferente Pequeño/fisiología , Animales , Ataxia/genética , Ataxia/patología , Ataxia/fisiopatología , Canales de Calcio Tipo N/genética , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Movimiento/fisiología , Nistagmo Patológico/genética , Nistagmo Patológico/patología , Nistagmo Patológico/fisiopatología , Equilibrio Postural/genética , ARN Interferente Pequeño/genética , Transducción Genética
14.
Neuroscience ; 510: 9-20, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36502959

RESUMEN

Psychostimulant drugs, such as cocaine, d-amphetamine and methylphenidate, alter a wide range of behaviors including locomotor activity and somatosensory perception. These altered behaviors are accompanied by the activation of specific neuronal populations within reward-, emotion- and locomotion-related circuits. However, whether such regulation occurs at the level of the spinal cord, a key node for neural circuits integrating and coordinating sensory and motor functions has never been addressed. By evaluating the temporal and spatial expression pattern of the phosphorylated form of the immediate early gene cFos at Ser32 (pS32-cFos), used as a proxy of neuronal activation, we demonstrate that, in adult male mice, d-amphetamine increases pS32-cFos expression in both inhibitory and excitatory neurons in dorsal and ventral horns at the lumbar spinal cord level. Interestingly, a fraction of neurons activated by a first exposure to d-amphetamine can be re-activated following d-amphetamine re-exposure. Similar expression patterns were observed in response to cocaine and methylphenidate, but not following morphine and dozilcipine administration. Finally, the blockade of dopamine reuptake was sufficient to recapitulate the increase in pS32-cFos expression induced by psychostimulant drugs. Our work provides evidence that cFos expression can be activated in lumbar spinal cord in response to acute psychostimulants administration.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Cocaína , Metilfenidato , Masculino , Ratones , Animales , Anfetamina/farmacología , Preparaciones Farmacéuticas , Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Metilfenidato/farmacología , Médula Espinal , Dextroanfetamina
15.
Front Neurosci ; 16: 1081010, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36570823

RESUMEN

Background: Shank3 is a scaffolding protein essential for the organization and function of the glutamatergic postsynapse. Monogenic mutations in SHANK3 gene are among the leading genetic causes of Autism Spectrum Disorders (ASD). The multiplicity of Shank3 isoforms seems to generate as much functional diversity and yet, there are no tools to study endogenous Shank3 proteins in an isoform-specific manner. Methods: In this study, we created a novel transgenic mouse line, the Shank3Venus/Venus knock in mouse, which allows to monitor the endogenous expression of the major Shank3 isoform in the brain, the full-length Shank3a isoform. Results: We show that the endogenous Venus-Shank3a protein is localized in spines and is mainly expressed in the striatum, hippocampus and cortex of the developing and adult brain. We show that Shank3Venus/+ and Shank3Venus/Venus mice have no behavioral deficiency. We further crossed Shank3Venus/Venus mice with Shank3ΔC/ΔC mice, a model of ASD, to track the Venus-tagged wild-type copy of Shank3a in physiological (Shank3Venus/+) and pathological (Shank3Venus/ΔC) conditions. We report a developmental delay in brain expression of the Venus-Shank3a isoform in Shank3Venus/ΔC mice, compared to Shank3Venus/+ control mice. Conclusion: Altogether, our results show that the Shank3Venus/Venus mouse line is a powerful tool to study endogenous Shank3a expression, in physiological conditions and in ASD.

16.
Nat Neurosci ; 25(7): 900-911, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35710984

RESUMEN

The cerebellum, a primary brain structure involved in the control of sensorimotor tasks, also contributes to higher cognitive functions including reward, emotion and social interaction. Although the regulation of these behaviors has been largely ascribed to the monoaminergic system in limbic regions, the contribution of cerebellar dopamine signaling in the modulation of these functions remains largely unknown. By combining cell-type-specific transcriptomics, histological analyses, three-dimensional imaging and patch-clamp recordings, we demonstrate that cerebellar dopamine D2 receptors (D2Rs) in mice are preferentially expressed in Purkinje cells (PCs) and regulate synaptic efficacy onto PCs. Moreover, we found that changes in D2R levels in PCs of male mice during adulthood alter sociability and preference for social novelty without affecting motor functions. Altogether, these findings demonstrate novel roles for D2R in PC function and causally link cerebellar D2R levels of expression to social behaviors.


Asunto(s)
Células de Purkinje , Receptores de Dopamina D2 , Animales , Cerebelo , Masculino , Ratones , Ratones Endogámicos C57BL , Células de Purkinje/fisiología , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Conducta Social
17.
Artículo en Inglés | MEDLINE | ID: mdl-32372941

RESUMEN

Epileptogenesis is the gradual process responsible for converting a healthy brain into an epileptic brain. This process can be triggered by a wide range of factors, including brain injury or tumors, infections, and status epilepticus. Epileptogenesis results in aberrant synaptic plasticity, neuroinflammation and seizure-induced cell death. As Matrix Metalloproteinases (MMPs) play a crucial role in cellular plasticity by remodeling the extracellular matrix (ECM), gelatinases (MMP-2 and MMP-9) were recently highlighted as key players in epileptogenesis. In this work, we engineered a biosensor to report in situ gelatinase activity in a model of epileptogenesis. This biosensor encompasses a gelatinase-sensitive activatable cell penetrating peptide (ACPP) coupled to a TAMRA fluorophore, allowing fluorescence uptake in cells displaying endogenous gelatinase activities. In a preclinical mouse model of temporal lobe epilepsy (TLE), the intrahippocampal kainate injection, ACPPs revealed a localized distribution of gelatinase activities, refining temporal cellular changes during epileptogenesis. The activity was found particularly but not only in the ipsilateral hippocampus, starting from the CA1 area and spreading to dentate gyrus from the early stages throughout chronic epilepsy, notably in neurons and microglial cells. Thus, our work shows that ACPPs are suitable molecular imaging probes for detecting the spatiotemporal pattern of gelatinase activity during epileptogenesis, suggesting their possible use as vectors to target cellular reactive changes with treatment for epileptogenesis.

18.
J Neurosci ; 28(34): 8604-14, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18716219

RESUMEN

The metabotropic glutamate receptor 7 (mGluR7) is widely expressed throughout the brain and primarily localized at presynaptic active zones, where it is thought to regulate neurotransmitter release. Protein interacting with C kinase 1 (PICK1), a postsynaptic density protein-95/disc-large tumor suppressor protein/zonula occludens-1 (PDZ)-domain protein, binds to the three C-terminal amino acids (-LVI) of the predominant mGluR7 splice variant, mGluR7a, and has been implicated in the synaptic clustering of this receptor. Here, we generated knock-in mice in which the C-terminal LVI coding sequence of exon 10 of the mGluR7 gene was replaced by three alanine codons (-AAA). Immunoprecipitation showed that the PICK1-mGluR7a interaction is disrupted in mGluR7a(AAA/AAA) mice. However, the synaptic localization of mGluR7a was not altered in cultured hippocampal neurons and brain sections prepared from the knock-in animals. In cerebellar granule cell cultures, the group III mGluR agonist l-AP-4 decreased the frequency of spontaneous excitatory currents in neurons derived from wild-type but not mGluR7a(AAA/AAA) mice, consistent with the interaction between mGluR7a and PICK1 being required for protein kinase C-mediated inhibition of glutamate release. At the behavioral level, the mGluR7a(AAA/AAA) mice showed no deficits in motor coordination, pain sensitivity, and anxiety but exhibited significant defects in hippocampus-dependent spatial working memory. In addition, they displayed a high susceptibility to the convulsant drug pentylenetetrazole. Together, these results indicate that PICK1 binding to the C-terminal region of mGluR7a is crucial for agonist-triggered presynaptic signaling in vivo.


Asunto(s)
Proteínas Portadoras/metabolismo , Convulsivantes , Antagonistas de Aminoácidos Excitadores , Trastornos de la Memoria/genética , Proteínas Nucleares/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Glutamato Metabotrópico/genética , Convulsiones/genética , Secuencias de Aminoácidos/genética , Animales , Conducta Animal , Encéfalo/patología , Proteínas de Ciclo Celular , Células Cultivadas , Cerebelo/patología , Cerebelo/fisiopatología , Predisposición Genética a la Enfermedad , Ácido Glutámico , Ligandos , Ratones , Ratones Transgénicos , Mutación , Inhibición Neural , Oocitos , Pentilenotetrazol , Terminales Presinápticos , Isoformas de Proteínas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Convulsiones/inducido químicamente , Transducción de Señal , Percepción Espacial , Sinapsis , Xenopus laevis
19.
J Physiol ; 587(1): 101-13, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19001039

RESUMEN

Extensive work has shown that activation of the cAMP-dependent protein kinase A (PKA) is crucial for long-term depression (LTD) of synaptic transmission in the hippocampus, a phenomenon that is thought to be involved in memory formation. Here we studied the role of an alternative target of cAMP, the exchange protein factor directly activated by cyclic AMP (Epac). We show that pharmacological activation of Epac by the selective agonist 8-(4-chlorophenylthio)-2'-O-methyl-cAMP (8-pCPT) induces LTD in the CA1 region. Paired-pulse facilitation of synaptic responses remained unchanged after induction of this LTD, suggesting that it depended on postsynaptic mechanisms. The 8-pCPT-induced LTD was blocked by the Epac signalling inhibitor brefeldin-A (BFA), Rap-1 antagonist geranylgeranyltransferase inhibitor (GGTI) and p38 mitogen activated protein kinase (P38-MAPK) inhibitor SB203580. This indicated a direct involvement of Epac in this form of LTD. As for other forms of LTD, a mimetic peptide of the PSD-95/Disc-large/ZO-1 homology (PDZ) ligand motif of the AMPA receptor subunit GluR2 blocked the Epac-LTD, suggesting involvement of PDZ protein interaction. The Epac-LTD also depended on mobilization of intracellular Ca(2+), proteasome activity and mRNA translation, but not transcription, as it was inhibited by thapsigargin, lactacystin and anisomycin, but not actinomycin-D, respectively. Finally, we found that the pituitary adenylate cyclase activating polypeptide (PACAP) can induce an LTD that was mutually occluded by the Epac-LTD and blocked by BFA or SB203580, suggesting that the Epac-LTD could be mobilized by stimulation of PACAP receptors. Altogether these results provided evidence for a new form of hippocampal LTD.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Hipocampo/fisiología , Depresión Sináptica a Largo Plazo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Animales , Brefeldino A/farmacología , Calcio/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Factores de Intercambio de Guanina Nucleótido/agonistas , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Receptores AMPA/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
20.
Cell Signal ; 19(9): 1996-2002, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17582742

RESUMEN

gamma-aminobutyric acid type B (GABA(B)) receptor is an allosteric complex made of two subunits, GABA(B1) and GABA(B2). GABA(B2) plays a major role in the coupling to G protein whereas GABA(B1) binds GABA. It has been shown that GABA(B) receptor activates ERK(1/2) in neurons of the central nervous system, but the molecular mechanisms underlying this event are poorly characterized. Here, we demonstrate that activation of GABA(B) receptor by either GABA or the selective agonist baclofen induces ERK(1/2) phosphorylation in cultured cerebellar granule neurons. We also show that CGP7930, a positive allosteric regulator specific of GABA(B2), alone can induce the phosphorylation of ERK(1/2). PTX, a G(i/o) inhibitor, abolishes both baclofen and CGP7930-mediated-ERK(1/2) phosphorylation. Moreover, both baclofen and CGP7930 induce ERK-dependent CREB phosphorylation. Furthermore, by using LY294002, a PI-3 kinase inhibitor, and a C-term of GRK-2 that has been reported to sequester Gbetagamma subunits, we demonstrate the role of Gbetagamma in GABA(B) receptor-mediated-ERK(1/2) phosphorylation. In conclusion, the activation of GABA(B) receptor leads to ERK(1/2) phosphorylation via the coupling of GABA(B2) to G(i/o) and by releasing Gbetagamma subunits which in turn induce the activation of CREB. These findings suggest a role of GABA(B) receptor in long-term change in the central nervous system.


Asunto(s)
Cerebelo/citología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Neuronas/enzimología , Receptores de GABA-B/metabolismo , Animales , Baclofeno/farmacología , Línea Celular , Cerebelo/efectos de los fármacos , Cerebelo/enzimología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Subunidades de Proteína/metabolismo , Ácido gamma-Aminobutírico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA