Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Cell Biol ; 220(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33683284

RESUMEN

Mutations in the human ALS2 gene cause recessive juvenile-onset amyotrophic lateral sclerosis and related motor neuron diseases. Although the ALS2 protein has been identified as a guanine-nucleotide exchange factor for the small GTPase Rab5, its physiological roles remain largely unknown. Here, we demonstrate that the Drosophila homologue of ALS2 (dALS2) promotes postsynaptic development by activating the Frizzled nuclear import (FNI) pathway. dALS2 loss causes structural defects in the postsynaptic subsynaptic reticulum (SSR), recapitulating the phenotypes observed in FNI pathway mutants. Consistently, these developmental phenotypes are rescued by postsynaptic expression of the signaling-competent C-terminal fragment of Drosophila Frizzled-2 (dFz2). We further demonstrate that dALS2 directs early to late endosome trafficking and that the dFz2 C terminus is cleaved in late endosomes. Finally, dALS2 loss causes age-dependent progressive defects resembling ALS, including locomotor impairment and brain neurodegeneration, independently of the FNI pathway. These findings establish novel regulatory roles for dALS2 in endosomal trafficking, synaptic development, and neuronal survival.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Endosomas/metabolismo , Endosomas/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Densidad Postsináptica/metabolismo , Densidad Postsináptica/fisiología , Esclerosis Amiotrófica Lateral/genética , Animales , Transporte Biológico/fisiología , Muerte Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiología , Endosomas/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Mutación/genética , Fenotipo , Densidad Postsináptica/genética , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
2.
Int J Mol Sci ; 21(15)2020 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756473

RESUMEN

BACKGROUND: Antipsychotic agents modulate key molecules of the postsynaptic density (PSD), including the Homer1a gene, implicated in dendritic spine architecture. How the antipsychotic receptor profile, dose, and duration of administration may influence synaptic plasticity and the Homer1a pattern of expression is yet to be determined. METHODS: In situ hybridization for Homer1a was performed on rat tissue sections from cortical and striatal regions of interest (ROI) after acute or chronic administration of three antipsychotics with divergent receptor profile: Haloperidol, asenapine, and olanzapine. Univariate and multivariate analyses of the effects of topography, treatment, dose, and duration of antipsychotic administration were performed. RESULTS: All acute treatment regimens were found to induce a consistently higher expression of Homer1a compared to chronic ones. Haloperidol increased Homer1a expression compared to olanzapine in striatum at the acute time-point. A dose effect was also observed for acute administration of haloperidol. CONCLUSIONS: Biological effects of antipsychotics on Homer1a varied strongly depending on the combination of their receptor profile, dose, duration of administration, and throughout the different brain regions. These molecular data may have translational valence and may reflect behavioral sensitization/tolerance phenomena observed with prolonged antipsychotics.


Asunto(s)
Antipsicóticos/farmacología , Encéfalo/efectos de los fármacos , Proteínas de Andamiaje Homer/genética , Plasticidad Neuronal/efectos de los fármacos , Animales , Antipsicóticos/efectos adversos , Encéfalo/metabolismo , Mapeo Encefálico , Dibenzocicloheptenos , Relación Dosis-Respuesta a Droga , Duración de la Terapia , Haloperidol/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Hibridación in Situ , Modelos Animales , Plasticidad Neuronal/genética , Olanzapina/farmacología , Densidad Postsináptica/efectos de los fármacos , Densidad Postsináptica/genética , Ratas , Distribución Tisular/efectos de los fármacos
3.
Aging (Albany NY) ; 12(3): 2169-2225, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32012119

RESUMEN

The prevalence of smoking is significantly higher in persons with schizophrenia (SCZ) than in the general population. However, the biological mechanisms of the comorbidity of smoking and SCZ are largely unknown. This study aimed to reveal shared biological pathways for the two diseases by analyzing data from two genome-wide association studies with a total sample size of 153,898. With pathway-based analysis, we first discovered 18 significantly enriched pathways shared by SCZ and smoking, which were classified into five groups: postsynaptic density, cadherin binding, dendritic spine, long-term depression, and axon guidance. Then, by using an integrative analysis of genetic, epigenetic, and expression data, we found not only 34 critical genes (e.g., PRKCZ, ARHGEF3, and CDKN1A) but also various risk-associated SNPs in these genes, which convey susceptibility to the comorbidity of the two disorders. Finally, using both in vivo and in vitro data, we demonstrated that the expression profiles of the 34 genes were significantly altered by multiple psychotropic drugs. Together, this multi-omics study not only reveals target genes for new drugs to treat SCZ but also reveals new insights into the shared genetic vulnerabilities of SCZ and smoking behaviors.


Asunto(s)
Encéfalo/metabolismo , Fumar Cigarrillos/genética , Esquizofrenia/genética , Orientación del Axón/genética , Cadherinas/genética , Cadherinas/metabolismo , Fumar Cigarrillos/epidemiología , Comorbilidad , Metilación de ADN , Bases de Datos Factuales , Bases de Datos Genéticas , Espinas Dendríticas/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Depresión Sináptica a Largo Plazo/genética , Farmacogenética , Densidad Postsináptica/genética , Esquizofrenia/epidemiología
4.
Cereb Cortex ; 30(4): 2573-2585, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-31800021

RESUMEN

It is generally accepted that formation and storage of memory relies on alterations of the structure and function of brain circuits. However, the structural data, which show learning-induced and long-lasting remodeling of synapses, are still very sparse. Here, we reconstruct 1927 dendritic spines and their postsynaptic densities (PSDs), representing a postsynaptic part of the glutamatergic synapse, in the hippocampal area CA1 of the mice that underwent spatial training. We observe that in young adult (5 months), mice volume of PSDs, but not the volume of the spines, is increased 26 h after the training. The training-induced growth of PSDs is specific for the dendritic spines that lack smooth endoplasmic reticulum and spine apparatuses, and requires autophosphorylation of αCaMKII. Interestingly, aging alters training-induced ultrastructural remodeling of dendritic spines. In old mice, both the median volumes of dendritic spines and PSDs shift after training toward bigger values. Overall, our data support the hypothesis that formation of memory leaves long-lasting footprint on the ultrastructure of brain circuits; however, the form of circuit remodeling changes with age.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Espinas Dendríticas/enzimología , Memoria a Largo Plazo/fisiología , Densidad Postsináptica/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Espinas Dendríticas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/fisiología , Densidad Postsináptica/genética , Densidad Postsináptica/ultraestructura
5.
Artículo en Inglés | MEDLINE | ID: mdl-31404590

RESUMEN

Type-5 metabotropic glutamate receptors (mGlu5) have been implicated in the mechanism of resilience to stress. They form part of the postsynaptic density (PSD), a thickening of the glutamatergic synapse that acts as a multimodal hub for multiple cellular signaling. Perinatal stress in rats triggers alterations that make adult offspring less resilient to stress. In the present study, we examined the expression of gene encoding the mGlu5 (Grm5), as well as those encoding the short and long isoforms of Homer proteins in different brain regions of the offspring of dams exposed to repeated episodes of restraint stress during pregnancy ("perinatally stressed" or PRS offspring). To this end, we investigated unconditioned behavioral response using the light/dark box test, as well as the expression of PSD genes (Homer1a, Homer1b, and Grm5), in the medial prefrontal cortex, cortex, caudate-putamen, amygdala, and dorsal hippocampus. PRS rats spent significantly less time in the light area than the control group. In the amygdala, Homer1a mRNA levels were significantly increased in PRS rats, whereas Homer1b and Grm5 mRNA levels were reduced. In contrast, the transcript encoding for Homer1a was significantly reduced in the medial prefrontal cortex, caudate-putamen, and dorsal hippocampus of PRS rats. We also evaluated the relative ratio between Homer1a and Homer1b/Grm5 expression, finding a significant shift toward the expression of Homer1a in the amygdala and toward Homer1b/Grm5 in the other brain regions. These topographic patterns of Homer1a, Homer1b, and mGlu5 gene expression were significantly correlated with risk-taking behavior measured in the light/dark box test. Remarkably, in the amygdala and in other brain regions, Homer1b and Grm5 expression showed positive correlation with time spent in the light box, whereas Homer1a in the amygdala showed a negative correlation with risk-taking behavior, in contrast with all other brain regions analyzed, wherein these correlations were positive. These results suggest that perinatal stress programs the developmental expression of PSD molecules involved in mGlu5 signaling in discrete brain regions, with a predominant role for the amygdala.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Andamiaje Homer/biosíntesis , Densidad Postsináptica/metabolismo , Receptor del Glutamato Metabotropico 5/biosíntesis , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Animales , Femenino , Expresión Génica , Proteínas de Andamiaje Homer/genética , Masculino , Densidad Postsináptica/genética , Embarazo , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/genética , Restricción Física
6.
J Clin Invest ; 129(6): 2390-2403, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31063986

RESUMEN

A disintegrine and metalloproteinase 10 (ADAM10) is implicated in synaptic function through its interaction with postsynaptic receptors and adhesion molecules. Here, we report that levels of active ADAM10 are increased in Huntington's disease (HD) mouse cortices and striata and in human postmortem caudate. We show that, in the presence of polyglutamine-expanded (polyQ-expanded) huntingtin (HTT), ADAM10 accumulates at the postsynaptic densities (PSDs) and causes excessive cleavage of the synaptic protein N-cadherin (N-CAD). This aberrant phenotype is also detected in neurons from HD patients where it can be reverted by selective silencing of mutant HTT. Consistently, ex vivo delivery of an ADAM10 synthetic inhibitor reduces N-CAD proteolysis and corrects electrophysiological alterations in striatal medium-sized spiny neurons (MSNs) of 2 HD mouse models. Moreover, we show that heterozygous conditional deletion of ADAM10 or delivery of a competitive TAT-Pro-ADAM10709-729 peptide in R6/2 mice prevents N-CAD proteolysis and ameliorates cognitive deficits in the mice. Reduction in synapse loss was also found in R6/2 mice conditionally deleted for ADAM10. Taken together, these results point to a detrimental role of hyperactive ADAM10 at the HD synapse and provide preclinical evidence of the therapeutic potential of ADAM10 inhibition in HD.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Disfunción Cognitiva/enzimología , Enfermedad de Huntington/enzimología , Proteínas de la Membrana/metabolismo , Densidad Postsináptica/enzimología , Proteína ADAM10/genética , Adulto , Anciano , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Proteínas de la Membrana/genética , Ratones Transgénicos , Persona de Mediana Edad , Densidad Postsináptica/genética , Densidad Postsináptica/patología
7.
J Clin Invest ; 129(2): 820-833, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30667373

RESUMEN

Molecular signaling mechanisms underlying Alzheimer's disease (AD) remain unclear. Maintenance of memory and synaptic plasticity depend on de novo protein synthesis, dysregulation of which is implicated in AD. Recent studies showed AD-associated hyperphosphorylation of mRNA translation factor eukaryotic elongation factor 2 (eEF2), which results in inhibition of protein synthesis. We tested to determine whether suppression of eEF2 phosphorylation could improve protein synthesis capacity and AD-associated cognitive and synaptic impairments. Genetic reduction of the eEF2 kinase (eEF2K) in 2 AD mouse models suppressed AD-associated eEF2 hyperphosphorylation and improved memory deficits and hippocampal long-term potentiation (LTP) impairments without altering brain amyloid ß (Aß) pathology. Furthermore, eEF2K reduction alleviated AD-associated defects in dendritic spine morphology, postsynaptic density formation, de novo protein synthesis, and dendritic polyribosome assembly. Our results link eEF2K/eEF2 signaling dysregulation to AD pathophysiology and therefore offer a feasible therapeutic target.


Asunto(s)
Enfermedad de Alzheimer , Espinas Dendríticas , Quinasa del Factor 2 de Elongación , Potenciación a Largo Plazo , Densidad Postsináptica , Transducción de Señal/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Quinasa del Factor 2 de Elongación/genética , Quinasa del Factor 2 de Elongación/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Factor 2 de Elongación Peptídica/genética , Factor 2 de Elongación Peptídica/metabolismo , Fosforilación/genética , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Densidad Postsináptica/patología
8.
Biochemistry ; 57(27): 4005-4009, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29913061

RESUMEN

The strength of each excitatory synapse in the central nervous system is regulated by its prior activity in a process called synaptic plasticity. The initiation of synaptic plasticity occurs when calcium ions enter the postsynaptic compartment and encounter a subcellular structure called the postsynaptic density (PSD). The PSD is attached to the postsynaptic membrane just underneath the concentrated plaque of neurotransmitter receptors. It is comprised of a core set of 30-60 proteins, approximately 20 of which are scaffold proteins. The rest include protein kinases and phosphatases, some of which respond to calcium ion; small GTPases and their regulators; chaperones; ubiquitins; and proteases. The assembly of the PSD involves competitive binding among a variety of specific protein binding sites to form a dynamic network. A biochemical challenge for the future is to understand how the dynamic regulation of the structure, composition, and activity of the PSD mediates synaptic plasticity and how mutations in PSD proteins lead to mental and neurodegenerative diseases.


Asunto(s)
Ácido Glutámico/metabolismo , Red Nerviosa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal , Densidad Postsináptica/metabolismo , Animales , Humanos , Potenciación a Largo Plazo , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Mutación , Red Nerviosa/citología , Red Nerviosa/patología , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Densidad Postsináptica/genética , Densidad Postsináptica/patología
9.
J Neurochem ; 145(6): 449-463, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29473168

RESUMEN

The Shank proteins are crucial scaffolding elements of the post-synaptic density (PSD). One of the best-characterized domains in Shank is the PDZ domain, which binds to C-terminal segments of several other PSD proteins. We carried out a detailed structural analysis of Shank3 PDZ domain-peptide complexes, to understand determinants of binding affinity towards different ligand proteins. Ligand peptides from four different proteins were cocrystallized with the Shank3 PDZ domain, and binding affinities were determined calorimetrically. In addition to conserved class I interactions between the first and third C-terminal peptide residue and Shank3, side chain interactions of other residues in the peptide with the PDZ domain are important factors in defining affinity. Structural conservation suggests that the binding specificities of the PDZ domains from different Shanks are similar. Two conserved buried water molecules in PDZ domains may affect correct local folding of ligand recognition determinants. The solution structure of a tandem Shank3 construct containing the SH3 and PDZ domains showed that the two domains are close to each other, which could be of relevance, when recognizing and binding full target proteins. The SH3 domain did not affect the affinity of the PDZ domain towards short target peptides, and the schizophrenia-linked Shank3 mutation R536W in the linker between the domains had no effect on the structure or peptide interactions of the Shank3 SH3-PDZ unit. Our data show the spatial arrangement of two adjacent Shank domains and pinpoint affinity determinants for short PDZ domain ligands with limited sequence homology.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Dominios PDZ/fisiología , Densidad Postsináptica/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Dicroismo Circular , Cristalización , Simulación de Dinámica Molecular , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Densidad Postsináptica/química , Densidad Postsináptica/fisiología , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Dispersión de Radiación , Esquizofrenia/genética , Agua/metabolismo , Rayos X
10.
J Biol Chem ; 293(7): 2232-2246, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29269412

RESUMEN

Altering the expression of Tomosyn-1 (Tomo-1), a soluble, R-SNARE domain-containing protein, significantly affects behavior in mice, Drosophila, and Caenorhabditis elegans Yet, the mechanisms that modulate Tomo-1 expression and its regulatory activity remain poorly defined. Here, we found that Tomo-1 expression levels influence postsynaptic spine density. Tomo-1 overexpression increased dendritic spine density, whereas Tomo-1 knockdown (KD) decreased spine density. These findings identified a novel action of Tomo-1 on dendritic spines, which is unique because it occurs independently of Tomo-1's C-terminal R-SNARE domain. We also demonstrated that the ubiquitin-proteasome system (UPS), which is known to influence synaptic strength, dynamically regulates Tomo-1 protein levels. Immunoprecipitated and affinity-purified Tomo-1 from cultured rat hippocampal neurons was ubiquitinated, and the levels of ubiquitinated Tomo-1 dramatically increased upon pharmacological proteasome blockade. Moreover, Tomo-1 ubiquitination appeared to be mediated through an interaction with the E3 ubiquitin ligase HRD1, as immunoprecipitation of Tomo-1 from neurons co-precipitated HRD1, and this interaction increases upon proteasome inhibition. Further, in vitro reactions indicated direct, HRD1 concentration-dependent Tomo-1 ubiquitination. We also noted that the UPS regulates both Tomo-1 expression and functional output, as HRD1 KD in hippocampal neurons increased Tomo-1 protein level and dendritic spine density. Notably, the effect of HRD1 KD on spine density was mitigated by additional KD of Tomo-1, indicating a direct HRD1/Tomo-1 effector relationship. In summary, our results indicate that the UPS is likely to participate in tuning synaptic efficacy and spine dynamics by precise regulation of neuronal Tomo-1 levels.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas R-SNARE/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Espinas Dendríticas/enzimología , Espinas Dendríticas/genética , Femenino , Hipocampo/citología , Hipocampo/enzimología , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas/enzimología , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Unión Proteica , Proteínas R-SNARE/genética , Ratas , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
11.
J Autism Dev Disord ; 47(11): 3600-3607, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28856484

RESUMEN

Approximately one-third of children with autism spectrum disorder (ASD) reportedly lose skills within the first 3 years, yet a causal mechanism remains elusive. Considering evidence of strong genetic effects for ASD and findings that distinct phenotypes in ASD associate with specific genetic events, we examined rates of parent-reported regression in the Simons Simplex Collection with likely gene disrupting mutations from five distinct classes: FMRP target genes, genes encoding chromatin modifiers, genes expressed preferentially in embryos, genes encoding postsynaptic density proteins, and essential genes. Children with ASD and mutations in postsynaptic density genes were more likely to experience regression, while a trend suggested that children with ASD and mutations in embryonic genes were less likely to have skill losses.


Asunto(s)
Trastorno del Espectro Autista/genética , Mutación , Adolescente , Trastorno del Espectro Autista/diagnóstico , Niño , Preescolar , Cromatina/genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Genes Esenciales , Humanos , Fenotipo , Densidad Postsináptica/genética
12.
Nat Neurosci ; 20(8): 1150-1161, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28671696

RESUMEN

The postsynaptic density (PSD) contains a collection of scaffold proteins used for assembling synaptic signaling complexes. However, it is not known how the core-scaffold machinery associates in protein-interaction networks or how proteins encoded by genes involved in complex brain disorders are distributed through spatiotemporal protein complexes. Here using immunopurification, proteomics and bioinformatics, we isolated 2,876 proteins across 41 in vivo interactomes and determined their protein domain composition, correlation to gene expression levels and developmental integration to the PSD. We defined clusters for enrichment of schizophrenia, autism spectrum disorders, developmental delay and intellectual disability risk factors at embryonic day 14 and adult PSD in mice. Mutations in highly connected nodes alter protein-protein interactions modulating macromolecular complexes enriched in disease risk candidates. These results were integrated into a software platform, Synaptic Protein/Pathways Resource (SyPPRes), enabling the prioritization of disease risk factors and their placement within synaptic protein interaction networks.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Densidad Postsináptica/genética , Sinapsis/metabolismo , Animales , Encefalopatías/genética , Encefalopatías/metabolismo , Modelos Animales de Enfermedad , Guanilato-Quinasas/genética , Proteínas de la Membrana/genética , Ratones Transgénicos , Esquizofrenia/genética , Esquizofrenia/metabolismo , Transducción de Señal/genética , Sinapsis/genética
13.
Mol Neurobiol ; 54(3): 1759-1776, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26884267

RESUMEN

The postsynaptic density (PSD) consists of a lattice-like array of interacting proteins that organizes and stabilizes synaptic receptors, ion channels, structural proteins, and signaling molecules required for normal synaptic transmission and synaptic function. The scaffolding and hub protein postsynaptic density protein-95 (PSD-95) is a major element of central chemical synapses and interacts with glutamate receptors, cell adhesion molecules, and cytoskeletal elements. In fact, PSD-95 can regulate basal synaptic stability as well as the activity-dependent structural plasticity of the PSD and, therefore, of the excitatory chemical synapse. Several studies have shown that PSD-95 is highly enriched at excitatory synapses and have identified multiple protein structural domains and protein-protein interactions that mediate PSD-95 function and trafficking to the postsynaptic region. PSD-95 is also a target of several signaling pathways that induce posttranslational modifications, including palmitoylation, phosphorylation, ubiquitination, nitrosylation, and neddylation; these modifications determine the synaptic stability and function of PSD-95 and thus regulate the fates of individual dendritic spines in the nervous system. In the present work, we review the posttranslational modifications that regulate the synaptic localization of PSD-95 and describe their functional consequences. We also explore the signaling pathways that induce such changes.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/análisis , Homólogo 4 de la Proteína Discs Large/metabolismo , Densidad Postsináptica/química , Densidad Postsináptica/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Homólogo 4 de la Proteína Discs Large/genética , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Densidad Postsináptica/genética , Sinapsis/química , Sinapsis/genética , Sinapsis/metabolismo
14.
Biochim Biophys Acta ; 1862(10): 1918-25, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27460706

RESUMEN

Autism is a severe neurodevelopmental disorder with a large population prevalence, characterized by abnormal reciprocal social interactions, communication deficits, and repetitive behaviors with restricted interests. The BTBR T(+)Itpr3(tf) (BTBR) mice have emerged as strong candidates to serve as models of a range of autism-relevant behaviors. Increasing evidences suggest that interleukin (IL)-6, one of the most important neuroimmune factors, was involved in the pathophysiology of autism. It is of great importance to further investigate whether therapeutic interventions in autism can be achieved through the manipulation of IL-6. Our previous studies showed that IL-6 elevation in the brain could mediate autistic-like behaviors, possibly through the imbalances of neural circuitry and impairments of synaptic plasticity. In this study, we evaluate whether inhibiting IL-6 signaling in the brain is sufficient to modulate the autism-like behaviors on the BTBR mice. The results showed that chronic infusion of an analog of the endogenous IL-6 trans-signaling blocker sgp130Fc protein increased the sociability in BTBR mice. Furthermore, no change was observed in the number of excitatory synapse, level of synaptic proteins, density of dentitic spine and postsynaptic density in BTBR cortices after inhibiting IL-6 trans-signaling. However, inhibition of IL-6 trans-signaling increased the evoked glutamate release in synaptoneurosomes from the cerebral cortex of BTBR mice. Our findings suggest that inhibition of excessive production of IL-6 may have selective therapeutic efficacy in treating abnormal social behaviors in autism.


Asunto(s)
Trastorno Autístico/metabolismo , Conducta Animal , Corteza Cerebral/metabolismo , Interleucina-6/metabolismo , Plasticidad Neuronal , Animales , Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/genética , Trastorno Autístico/patología , Corteza Cerebral/patología , Receptor gp130 de Citocinas/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Ratones , Ratones Transgénicos , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Densidad Postsináptica/patología , Proteínas Recombinantes/farmacología , Transducción de Señal
15.
Front Neural Circuits ; 10: 31, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27199672

RESUMEN

Mutation of the metabotropic glutamate receptor type 7 (mGlu7) induces absence-like epileptic seizures, but its precise role in the somatosensory thalamocortical network remains unknown. By combining electrophysiological recordings, optogenetics, and pharmacology, we dissected the contribution of the mGlu7 receptor at mouse thalamic synapses. We found that mGlu7 is functionally expressed at both glutamatergic and GABAergic synapses, where it can inhibit neurotransmission and regulate short-term plasticity. These effects depend on the PDZ-ligand of the receptor, as they are lost in mutant mice. Interestingly, the very low affinity of mGlu7 receptors for glutamate raises the question of how it can be activated, namely at GABAergic synapses and in basal conditions. Inactivation of the receptor activity with the mGlu7 negative allosteric modulator (NAM), ADX71743, enhances thalamic synaptic transmission. In vivo administration of the NAM induces a lethargic state with spindle and/or spike-and-wave discharges accompanied by a behavioral arrest typical of absence epileptic seizures. This provides evidence for mGlu7 receptor-mediated tonic modulation of a physiological function in vivo preventing synchronous and potentially pathological oscillations.


Asunto(s)
Corteza Cerebral/citología , Vías Nerviosas/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Tálamo/fisiología , Animales , Benzoxazoles/química , Benzoxazoles/farmacología , Corteza Cerebral/fisiología , Channelrhodopsins , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , GABAérgicos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Densidad Postsináptica/efectos de los fármacos , Densidad Postsináptica/genética , Receptores de GABA-A/fisiología , Receptores de Glutamato Metabotrópico/genética , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética
16.
Neuropsychopharmacology ; 41(3): 886-95, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26211730

RESUMEN

Genome-wide association studies (GWAS) have identified several common variants associated with bipolar disorder (BD), but the biological meaning of these findings remains unclear. Integrative genomics-the integration of GWAS signals with gene expression data-may illuminate genes and gene networks that have key roles in the pathogenesis of BD. We applied weighted gene co-expression network analysis (WGCNA), which exploits patterns of co-expression among genes, to brain transcriptome data obtained by sequencing of poly-A RNA derived from postmortem dorsolateral prefrontal cortex from people with BD, along with age- and sex-matched controls. WGCNA identified 33 gene modules. Many of the modules corresponded closely to those previously reported in human cortex. Three modules were associated with BD, enriched for genes differentially expressed in BD, and also enriched for signals in prior GWAS of BD. Functional analysis of genes within these modules revealed significant enrichment of several functionally related sets of genes, especially those involved in the postsynaptic density (PSD). These results provide convergent support for the hypothesis that dysregulation of genes involved in the PSD is a key factor in the pathogenesis of BD. If replicated in larger samples, these findings could point toward new therapeutic targets for BD.


Asunto(s)
Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Corteza Prefrontal/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Análisis por Micromatrices
17.
J Biol Chem ; 290(8): 5105-5116, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25555912

RESUMEN

Synapse-associated protein 102 (SAP102) is a scaffolding protein abundantly expressed early in development that mediates glutamate receptor trafficking during synaptogenesis. Mutations in human SAP102 have been reported to cause intellectual disability, which is consistent with its important role during early postnatal development. SAP102 contains PDZ, SH3, and guanylate kinase (GK)-like domains, which mediate specific protein-protein interactions. SAP102 binds directly to N-methyl-D-aspartate receptors (NMDARs), anchors receptors at synapses, and facilitates transduction of NMDAR signals. Proper localization of SAP102 at the postsynaptic density is essential to these functions. However, how SAP102 is targeted to synapses is unclear. In the current study we find that synaptic localization of SAP102 is regulated by alternative splicing. The SAP102 splice variant that possesses a C-terminal insert (I2) between the SH3 and GK domains is highly enriched at dendritic spines. We also show that there is an intramolecular interaction between the SH3 and GK domains in SAP102 but that the I2 splicing does not influence SH3-GK interaction. Previously, we have shown that SAP102 expression promotes spine lengthening. We now find that the spine lengthening effect is independent of the C-terminal alternative splicing of SAP102. In addition, expression of I2-containing SAP102 isoforms is regulated developmentally. Knockdown of endogenous I2-containing SAP102 isoforms differentially affect NMDAR surface expression in a subunit-specific manner. These data shed new light on the role of SAP102 in the regulation of NMDAR trafficking.


Asunto(s)
Empalme Alternativo/fisiología , Neuropéptidos/metabolismo , Proteínas Nucleares/metabolismo , Densidad Postsináptica/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Factores de Transcripción/metabolismo , Animales , Células HEK293 , Humanos , Neuropéptidos/genética , Proteínas Nucleares/genética , Densidad Postsináptica/genética , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Factores de Transcripción/genética , Dominios Homologos src
18.
Mol Psychiatry ; 20(9): 1091-100, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25330739

RESUMEN

Numerous investigations support decreased glutamatergic signaling as a pathogenic mechanism of schizophrenia, yet the molecular underpinnings for such dysregulation are largely unknown. In the post-mortem dorsolateral prefrontal cortex (DLPFC), we found striking decreases in tyrosine phosphorylation of N-methyl-D aspartate (NMDA) receptor subunit 2 (GluN2) that is critical for neuroplasticity. The decreased GluN2 activity in schizophrenia may not be because of downregulation of NMDA receptors as MK-801 binding and NMDA receptor complexes in postsynaptic density (PSD) were in fact increased in schizophrenia cases. At the postreceptor level, however, we found striking reductions in the protein kinase C, Pyk 2 and Src kinase activity that in tandem can decrease GluN2 activation. Given that Src serves as a hub of various signaling mechanisms affecting GluN2 phosphorylation, we postulated that Src hypoactivity may result from convergent alterations of various schizophrenia susceptibility pathways and thus mediate their effects on NMDA receptor signaling. Indeed, the DLPFC of schizophrenia cases exhibit increased PSD-95 and erbB4 and decreased receptor-type tyrosine-protein phosphatase-α (RPTPα) and dysbindin-1, each of which reduces Src activity via protein interaction with Src. To test genomic underpinnings for Src hypoactivity, we examined genome-wide association study results, incorporating 13 394 cases and 34 676 controls. We found no significant association of individual variants of Src and its direct regulators with schizophrenia. However, a protein-protein interaction-based network centered on Src showed significant enrichment of gene-level associations with schizophrenia compared with other psychiatric illnesses. Our results together demonstrate striking decreases in NMDA receptor signaling at the postreceptor level and propose Src as a nodal point of convergent dysregulations affecting NMDA receptor pathway via protein-protein associations.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Familia-src Quinasas/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratones Noqueados , Plasticidad Neuronal , Fosforilación , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Corteza Prefrontal/metabolismo , Mapas de Interacción de Proteínas , Esquizofrenia/enzimología , Esquizofrenia/patología , Transducción de Señal , Familia-src Quinasas/genética
19.
Mol Psychiatry ; 20(4): 424-32, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25048004

RESUMEN

The postsynaptic density (PSD) contains a complex set of proteins of known relevance to neuropsychiatric disorders, and schizophrenia specifically. We enriched for this anatomical structure, in the anterior cingulate cortex, of 20 schizophrenia samples and 20 controls from the Stanley Medical Research Institute, and used unbiased shotgun proteomics incorporating label-free quantitation to identify differentially expressed proteins. Quantitative investigation of the PSD revealed more than 700 protein identifications and 143 differentially expressed proteins. Prominent among these were altered expression of proteins involved in clathrin-mediated endocytosis (CME) (Dynamin-1, adaptor protein 2) and N-methyl-D-aspartate (NMDA)-interacting proteins such as CYFIP2, SYNPO, SHANK3, ESYT and MAPK3 (all P<0.0015). Pathway analysis of the differentially expressed proteins implicated the cellular processes of endocytosis, long-term potentiation and calcium signaling. Both single-gene and gene-set enrichment analyses in genome-wide association data from the largest schizophrenia sample to date of 13,689 cases and 18,226 controls show significant association of HIST1H1E and MAPK3, and enrichment of our PSD proteome. Taken together, our data provide robust evidence implicating PSD-associated proteins and genes in schizophrenia, and suggest that within the PSD, NMDA-interacting and endocytosis-related proteins contribute to disease pathophysiology.


Asunto(s)
Regulación de la Expresión Génica/genética , Genómica , Giro del Cíngulo/patología , Densidad Postsináptica , Proteómica , Esquizofrenia , Animales , Antipsicóticos/farmacología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Femenino , Estudios de Asociación Genética , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/metabolismo , Haloperidol/farmacología , Humanos , Masculino , N-Metilaspartato/genética , N-Metilaspartato/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Densidad Postsináptica/patología , Ratas , Reproducibilidad de los Resultados , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sinaptotagminas/metabolismo , Espectrometría de Masas en Tándem
20.
J Neurosci ; 34(49): 16482-95, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471585

RESUMEN

Frontotemporal dementia (FTD) is a neurodegenerative behavioral disorder that selectively affects the salience network, including the ventral striatum and insula. Tau mutations cause FTD, but how mutant tau impairs the salience network is unknown. Here, we address this question using a mouse model expressing the entire human tau gene with an FTD-associated mutation (V337M). Mutant, but not wild-type, human tau transgenic mice had aging-dependent repetitive and disinhibited behaviors, with synaptic deficits selectively in the ventral striatum and insula. There, mutant tau depleted PSD-95, resulting in smaller postsynaptic densities and impaired synaptic localization of NMDA receptors (NMDARs). In the ventral striatum, decreased NMDAR-mediated transmission reduced striatal neuron firing. Pharmacologically enhancing NMDAR function with the NMDAR co-agonist cycloserine reversed electrophysiological and behavioral deficits. These results indicate that NMDAR hypofunction critically contributes to FTD-associated behavioral and electrophysiological alterations and that this process can be therapeutically targeted by a Food and Drug Administration-approved drug.


Asunto(s)
Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/fisiopatología , Receptores de N-Metil-D-Aspartato/fisiología , Proteínas tau/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Envejecimiento/psicología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cicloserina/farmacología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Agonistas de Aminoácidos Excitadores/farmacología , Agonistas de Aminoácidos Excitadores/uso terapéutico , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Demencia Frontotemporal/tratamiento farmacológico , Guanilato-Quinasas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mutación , Neuronas/fisiología , Densidad Postsináptica/genética , Densidad Postsináptica/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA