Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(10): 113183, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37777962

RESUMEN

Recent developments in genome sequencing have expanded the knowledge of genetic factors associated with late-onset Alzheimer's disease (AD). Among them, genetic variant ε4 of the APOE gene (APOE4) confers the greatest disease risk. Dysregulated glucose metabolism is an early pathological feature of AD. Using isogenic ApoE3 and ApoE4 astrocytes derived from human induced pluripotent stem cells, we find that ApoE4 increases glycolytic activity but impairs mitochondrial respiration in astrocytes. Ultrastructural and autophagy flux analyses show that ApoE4-induced cholesterol accumulation impairs lysosome-dependent removal of damaged mitochondria. Acute treatment with cholesterol-depleting agents restores autophagic activity, mitochondrial dynamics, and associated proteomes, and extended treatment rescues mitochondrial respiration in ApoE4 astrocytes. Taken together, our study provides a direct link between ApoE4-induced lysosomal cholesterol accumulation and abnormal oxidative phosphorylation.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Astrocitos/metabolismo , Fosforilación Oxidativa , Células Cultivadas , Células Madre Pluripotentes Inducidas/metabolismo , Apolipoproteína E3/metabolismo , Colesterol/metabolismo , Enfermedad de Alzheimer/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo
2.
Sensors (Basel) ; 22(21)2022 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-36366175

RESUMEN

With the advancement of sensors, image and video processing have developed for use in the visual sensing area. Among them, video super-resolution (VSR) aims to reconstruct high-resolution sequences from low-resolution sequences. To use consecutive contexts within a low-resolution sequence, VSR learns the spatial and temporal characteristics of multiple frames of the low-resolution sequence. As one of the convolutional neural network-based VSR methods, we propose a deformable convolution-based alignment network (DCAN) to generate scaled high-resolution sequences with quadruple the size of the low-resolution sequences. The proposed method consists of a feature extraction block, two different alignment blocks that use deformable convolution, and an up-sampling block. Experimental results show that the proposed DCAN achieved better performances in both the peak signal-to-noise ratio and structural similarity index measure than the compared methods. The proposed DCAN significantly reduces the network complexities, such as the number of network parameters, the total memory, and the inference speed, compared with the latest method.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Procesamiento de Imagen Asistido por Computador/métodos , Relación Señal-Ruido
3.
Sensors (Basel) ; 22(15)2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35957313

RESUMEN

Despite advanced machine learning methods, the implementation of emotion recognition systems based on real-world video content remains challenging. Videos may contain data such as images, audio, and text. However, the application of multimodal models using two or more types of data to real-world video media (CCTV, illegally filmed content, etc.) lacking sound or subtitles is difficult. Although facial expressions in image sequences can be utilized in emotion recognition, the diverse identities of individuals in real-world content limits computational models of relationships between facial expressions. This study proposed a transformation model which employed a video vision transformer to focus on facial expression sequences in videos. It effectively understood and extracted facial expression information from the identities of individuals, instead of fusing multimodal models. The design entailed capture of higher-quality facial expression information through mixed-token embedding facial expression sequences augmented via various methods into a single data representation, and comprised two modules: spatial and temporal encoders. Further, temporal position embedding, focusing on relationships between video frames, was proposed and subsequently applied to the temporal encoder module. The performance of the proposed algorithm was compared with that of conventional methods on two emotion recognition datasets of video content, with results demonstrating its superiority.


Asunto(s)
Reconocimiento Facial , Algoritmos , Cara , Expresión Facial , Humanos , Aprendizaje Automático
4.
FASEB J ; 35(10): e21894, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34460995

RESUMEN

Neuromyelitis optica (NMO) is an autoimmune disease that primarily targets astrocytes. Autoantibodies (NMO-IgG) against the water channel protein, aquaporin 4 (AQP4), are a serologic marker in NMO patients, and they are known to be responsible for the pathophysiology of the disease. In the brain, AQP4 is mainly expressed in astrocytes, especially at the end-feet, where they form the blood-brain barrier. Following the interaction between NMO-IgG and AQP4 in astrocytes, rapid AQP4 endocytosis initiates pathogenesis. However, the cellular and molecular mechanisms of astrocyte destruction by autoantibodies remain largely elusive. We established an in vitro human astrocyte model system using induced pluripotent stem cells (iPSCs) technology in combination with NMO patient-derived serum and IgG to elucidate the cellular and functional changes caused by NMO-IgG. Herein, we observed that NMO-IgG induces structural alterations in mitochondria and their association with the endoplasmic reticulum (ER) and lysosomes at the ultrastructural level, which potentially leads to impaired mitochondrial functions and dynamics. Indeed, human astrocytes display impaired mitochondrial bioenergetics and autophagy activity in the presence of NMO-IgG. We further demonstrated NMO-IgG-driven ER membrane deformation into a multilamellar structure in human astrocytes. Together, we show that NMO-IgG rearranges cellular organelles and alter their functions and that our in vitro system using human iPSCs offers previously unavailable experimental opportunities to study the pathophysiological mechanisms of NMO in human astrocytes or conduct large-scale screening for potential therapeutic compounds targeting astrocytic abnormalities in patients with NMO.


Asunto(s)
Astrocitos/inmunología , Autoanticuerpos/inmunología , Retículo Endoplásmico/inmunología , Inmunoglobulina G/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Mitocondrias/inmunología , Neuromielitis Óptica/inmunología , Acuaporina 4/inmunología , Humanos
5.
Stem Cell Reports ; 16(9): 2128-2137, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34450034

RESUMEN

The ε4 allele of APOE-encoding apolipoprotein (ApoE) is one of the strongest genetic risk factors for Alzheimer's disease (AD). One of the overarching questions is whether and how this astrocyte-enriched risk factor initiates AD-associated pathology in neurons such as amyloid-ß (Aß) accumulation. Here, we generate neurons and astrocytes from isogenic human induced pluripotent stem cells (hiPSCs) carrying either APOE ε3 or APOE ε4 allele and investigate the effect of astrocytic ApoE4 on neuronal Aß production. Secretory factors in conditioned media from ApoE4 astrocytes significantly increased amyloid precursor protein (APP) levels and Aß secretion in neurons. We further found that increased cholesterol secretion from ApoE4 astrocytes was necessary and sufficient to induce the formation of lipid rafts that potentially provide a physical platform for APP localization and facilitate its processing. Our study reveals the contribution of ApoE4 astrocytes to amyloidosis in neurons by expanding lipid rafts and facilitating Aß production through an oversupply of cholesterol.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Apolipoproteína E4/genética , Astrocitos/metabolismo , Colesterol/metabolismo , Microdominios de Membrana/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteína E4/metabolismo , Biomarcadores , Comunicación Celular , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Espacio Extracelular/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/efectos de los fármacos
7.
Mol Neurobiol ; 58(6): 2608-2619, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33479841

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disease that affects more than 30 million people worldwide. Despite growing knowledge of AD pathophysiology, a complete understanding of the pathogenic mechanisms underpinning AD is lacking, and there is currently no cure for AD. Extant literature suggests that AD is a polygenic and multifactorial disease underscored by complex and dynamic pathogenic mechanisms. Despite extensive research and clinical trials, there has been a dearth of novel drugs for AD treatment on the market since memantine in 2003. This lack of therapeutic success has directed the entire research community to approach the disease from a different angle. In this review, we discuss growing evidence for the close link between altered glucose metabolism and AD pathogenesis by exploring how genetic risk factors for AD are associated with dysfunctional glucose metabolism. We also discuss modification of genes responsible for metabolic pathways implicated in AD pathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Predisposición Genética a la Enfermedad , Glucosa/metabolismo , Animales , Glucólisis/genética , Humanos , Factores de Riesgo
8.
Mol Cells ; 42(11): 739-746, 2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31711277

RESUMEN

Significant knowledge about the pathophysiology of Alzheimer's disease (AD) has been gained in the last century; however, the understanding of its causes of onset remains limited. Late-onset AD is observed in about 95% of patients, and APOE4-encoding apolipoprotein E4 (ApoE4) is strongly associated with these cases. As an apolipoprotein, the function of ApoE in brain cholesterol transport has been extensively studied and widely appreciated. Development of new technologies such as human-induced pluripotent stem cells (hiPSCs) and CRISPR-Cas9 genome editing tools have enabled us to develop human brain model systems in vitro and readily manipulate genomic information. In the context of these advances, recent studies provide strong evidence that abnormal cholesterol metabolism by ApoE4 could be linked to AD-associated pathology. In this review, we discuss novel discoveries in brain cholesterol dysregulation by ApoE4. We further elaborate cell type-specific roles in cholesterol regulation of four major brain cell types, neurons, astrocytes, microglia, and oligodendrocytes, and how its dysregulation can be linked to AD pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Colesterol/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Encéfalo/citología , Humanos , Microglía/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(44): 11316-11321, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30327343

RESUMEN

Astrocytes are important regulators of neural circuit function and behavior in the healthy and diseased nervous system. We screened for molecules in Drosophila astrocytes that modulate neuronal hyperexcitability and identified multiple components of focal adhesion complexes (FAs). Depletion of astrocytic Tensin, ß-integrin, Talin, focal adhesion kinase (FAK), or matrix metalloproteinase 1 (Mmp1), resulted in enhanced behavioral recovery from genetic or pharmacologically induced seizure. Overexpression of Mmp1, predicted to activate FA signaling, led to a reciprocal enhancement of seizure severity. Blockade of FA-signaling molecules in astrocytes at basal levels of CNS excitability resulted in reduced astrocytic coverage of the synaptic neuropil and expression of the excitatory amino acid transporter EAAT1. However, induction of hyperexcitability after depletion of FA-signaling components resulted in enhanced astrocyte coverage and an approximately twofold increase in EAAT1 levels. Our work identifies FA-signaling molecules as important regulators of astrocyte outgrowth and EAAT1 expression under normal physiological conditions. Paradoxically, in the context of hyperexcitability, this pathway negatively regulates astrocytic process outgrowth and EAAT1 expression, and their blockade leading to enhanced recovery from seizure.


Asunto(s)
Astrocitos/metabolismo , Adhesiones Focales/metabolismo , Glutamatos/metabolismo , Animales , Transporte Biológico/fisiología , Drosophila/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Neuronas/metabolismo , Convulsiones/metabolismo
10.
J Neurosci ; 37(41): 9917-9924, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912154

RESUMEN

Increased p25, a proteolytic fragment of the regulatory subunit p35, is known to induce aberrant activity of cyclin-dependent kinase 5 (Cdk5), which is associated with neurodegenerative disorders, including Alzheimer's disease. Previously, we showed that replacing endogenous p35 with the noncleavable mutant p35 (Δp35) attenuated amyloidosis and improved cognitive function in a familial Alzheimer's disease mouse model. Here, to address the role of p25/Cdk5 in tauopathy, we generated double-transgenic mice by crossing mice overexpressing mutant human tau (P301S) with Δp35KI mice. We observed significant reduction of phosphorylated tau and its seeding activity in the brain of double transgenic mice compared with the P301S mice. Furthermore, synaptic loss and impaired LTP at hippocampal CA3 region of P301S mice were attenuated by blocking p25 generation. To further validate the role of p25/Cdk5 in tauopathy, we used frontotemporal dementia patient-derived induced pluripotent stem cells (iPSCs) carrying the Tau P301L mutation and generated P301L:Δp35KI isogenic iPSC lines using CRISPR/Cas9 genome editing. We created cerebral organoids from the isogenic iPSCs and found that blockade of p25 generation reduced levels of phosphorylated tau and increased expression of synaptophysin. Together, these data demonstrate a crucial role for p25/Cdk5 in mediating tau-associated pathology and suggest that inhibition of this kinase can remedy neurodegenerative processes in the presence of pathogenic tau mutation.SIGNIFICANCE STATEMENT Accumulation of p25 results in aberrant Cdk5 activation and induction of numerous pathological phenotypes, such as neuroinflammation, synaptic loss, Aß accumulation, and tau hyperphosphorylation. However, it was not clear whether p25/Cdk5 activity is necessary for the progression of these pathological changes. We recently developed the Δp35KI transgenic mouse that is deficient in p25 generation and Cdk5 hyperactivation. In this study, we used this mouse model to elucidate the role of p25/Cdk5 in FTD mutant tau-mediated pathology. We also used a frontotemporal dementia patient-derived induced pluripotent stem cell carrying the Tau P301L mutation and generated isogenic lines in which p35 is replaced with noncleavable mutant Δp35. Our data suggest that p25/Cdk5 plays an important role in tauopathy in both mouse and human model systems.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Demencia Frontotemporal/genética , Fosfotransferasas/genética , Células Madre Pluripotentes , Tauopatías/genética , Animales , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Demencia Frontotemporal/prevención & control , Humanos , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Fibras Musgosas del Hipocampo/patología , Fosforilación , Fosfotransferasas/antagonistas & inhibidores , Trasplante de Células Madre , Sinapsis/patología , Sinaptofisina/genética , Tauopatías/prevención & control
12.
Cell Rep ; 13(5): 933-43, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26565907

RESUMEN

Lateral diffusion in the membrane and endosomal trafficking both contribute to the addition and removal of AMPA receptors (AMPARs) at postsynaptic sites. However, the spatial coordination between these mechanisms has remained unclear, because little is known about the dynamics of AMPAR-containing endosomes. In addition, how the positioning of AMPAR-containing endosomes affects synapse organization and functioning has never been directly explored. Here, we used live-cell imaging in hippocampal neuron cultures to show that intracellular AMPARs are transported in Rab11-positive recycling endosomes, which frequently enter dendritic spines and depend on the microtubule and actin cytoskeleton. By using chemically induced dimerization systems to recruit kinesin (KIF1C) or myosin (MyosinV/VI) motors to Rab11-positive recycling endosomes, we controlled their trafficking and found that induced removal of recycling endosomes from spines decreases surface AMPAR expression and PSD-95 clusters at synapses. Our data suggest a mechanistic link between endosome positioning and postsynaptic structure and composition.


Asunto(s)
Endosomas/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Femenino , Cinesinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miosinas/metabolismo , Ratas , Receptores AMPA/genética , Sinapsis/ultraestructura , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
13.
Cell ; 161(7): 1592-605, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26052046

RESUMEN

Neuronal activity causes the rapid expression of immediate early genes that are crucial for experience-driven changes to synapses, learning, and memory. Here, using both molecular and genome-wide next-generation sequencing methods, we report that neuronal activity stimulation triggers the formation of DNA double strand breaks (DSBs) in the promoters of a subset of early-response genes, including Fos, Npas4, and Egr1. Generation of targeted DNA DSBs within Fos and Npas4 promoters is sufficient to induce their expression even in the absence of an external stimulus. Activity-dependent DSB formation is likely mediated by the type II topoisomerase, Topoisomerase IIß (Topo IIß), and knockdown of Topo IIß attenuates both DSB formation and early-response gene expression following neuronal stimulation. Our results suggest that DSB formation is a physiological event that rapidly resolves topological constraints to early-response gene expression in neurons.


Asunto(s)
Roturas del ADN de Doble Cadena , Neuronas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factor de Unión a CCCTC , ADN-Topoisomerasas de Tipo II/análisis , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Etopósido/farmacología , Regulación de la Expresión Génica , Genes fos , Estudio de Asociación del Genoma Completo , Ratones , Proteínas Represoras/metabolismo , Transcriptoma/efectos de los fármacos
14.
Nat Neurosci ; 18(7): 1008-16, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26005852

RESUMEN

Noncoding variants in the human MIR137 gene locus increase schizophrenia risk with genome-wide significance. However, the functional consequence of these risk alleles is unknown. Here we examined induced human neurons harboring the minor alleles of four disease-associated single nucleotide polymorphisms in MIR137. We observed increased MIR137 levels compared to those in major allele-carrying cells. microRNA-137 gain of function caused downregulation of the presynaptic target genes complexin-1 (Cplx1), Nsf and synaptotagmin-1 (Syt1), leading to impaired vesicle release. In vivo, miR-137 gain of function resulted in changes in synaptic vesicle pool distribution, impaired induction of mossy fiber long-term potentiation and deficits in hippocampus-dependent learning and memory. By sequestering endogenous miR-137, we were able to ameliorate the synaptic phenotypes. Moreover, reinstatement of Syt1 expression partially restored synaptic plasticity, demonstrating the importance of Syt1 as a miR-137 target. Our data provide new insight into the mechanism by which miR-137 dysregulation can impair synaptic plasticity in the hippocampus.


Asunto(s)
Regulación de la Expresión Génica/genética , MicroARNs/metabolismo , Fibras Musgosas del Hipocampo/metabolismo , Plasticidad Neuronal/genética , Esquizofrenia/genética , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Alelos , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Fibroblastos , Sitios Genéticos , Células HEK293 , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo , Ratones , Ratones Endogámicos C57BL , Proteínas Sensibles a N-Etilmaleimida/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Polimorfismo de Nucleótido Simple , Sinaptotagmina I/metabolismo
15.
Proc Natl Acad Sci U S A ; 112(23): 7291-6, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-25995364

RESUMEN

Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.


Asunto(s)
Complejo Nuclear Basolateral/fisiopatología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/fisiopatología , Trastornos de la Memoria/fisiopatología , Animales , Complejo Nuclear Basolateral/efectos de la radiación , Hipocampo/efectos de la radiación , Luz , Ratones , Estrés Fisiológico
16.
Eur J Neurosci ; 41(8): 1025-35, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25816842

RESUMEN

The Shank genes (SHANK1, 2, 3) encode scaffold proteins highly enriched in postsynaptic densities where they regulate synaptic structure in spiny neurons. Mutations in human Shank genes are linked to autism spectrum disorder and schizophrenia. Shank1 mutant mice exhibit intriguing cognitive phenotypes reminiscent of individuals with autism spectrum disorder. However, the molecular mechanisms leading to the human pathophysiological phenotypes and mouse behaviors have not been elucidated. In this study it is shown that Shank1 protein is highly localized in parvalbumin-expressing (PV+) fast-spiking inhibitory interneurons in the hippocampus. Importantly, a lack of Shank1 in hippocampal CA1 PV+ neurons reduced excitatory synaptic inputs and inhibitory synaptic outputs to pyramidal neurons. Furthermore, it is demonstrated that hippocampal CA1 pyramidal neurons in Shank1 mutant mice exhibit a shift in the excitatory and inhibitory balance (E-I balance), a pathophysiological hallmark of autism spectrum disorder. The mutant mice also exhibit lower expression of gephyrin (a scaffold component of inhibitory synapses), supporting the dysregulation of E-I balance in the hippocampus. These results suggest that Shank1 scaffold in PV+ interneurons regulates excitatory synaptic strength and participates in the maintenance of E-I balance in excitatory neurons.


Asunto(s)
Región CA1 Hipocampal/fisiología , Neuronas GABAérgicas/fisiología , Proteínas del Tejido Nervioso/fisiología , Células Piramidales/fisiología , Transmisión Sináptica , Animales , Región CA1 Hipocampal/metabolismo , Proteínas Portadoras/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Potenciales de la Membrana , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural , Parvalbúminas/metabolismo , Densidad Postsináptica/metabolismo , Células Piramidales/metabolismo
17.
Mol Brain ; 8: 7, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25645137

RESUMEN

BACKGROUND: Activation of G protein coupled receptor (GPCR) in astrocytes leads to Ca(2+)-dependent glutamate release via Bestrophin 1 (Best1) channel. Whether receptor-mediated glutamate release from astrocytes can regulate synaptic plasticity remains to be fully understood. RESULTS: We show here that Best1-mediated astrocytic glutamate activates the synaptic N-methyl-D-aspartate receptor (NMDAR) and modulates NMDAR-dependent synaptic plasticity. Our data show that activation of the protease-activated receptor 1 (PAR1) in hippocampal CA1 astrocytes elevates the glutamate concentration at Schaffer collateral-CA1 (SC-CA1) synapses, resulting in activation of GluN2A-containing NMDARs and NMDAR-dependent potentiation of synaptic responses. Furthermore, the threshold for inducing NMDAR-dependent long-term potentiation (LTP) is lowered when astrocytic glutamate release accompanied LTP induction, suggesting that astrocytic glutamate is significant in modulating synaptic plasticity. CONCLUSIONS: Our results provide direct evidence for the physiological importance of channel-mediated astrocytic glutamate in modulating neural circuit functions.


Asunto(s)
Astrocitos/metabolismo , Proteínas del Ojo/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Canales Iónicos/metabolismo , Plasticidad Neuronal , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Bestrofinas , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/ultraestructura , Hipocampo/ultraestructura , Humanos , Potenciación a Largo Plazo , Ratones , Modelos Biológicos , Receptor PAR-1/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
18.
Neuroepigenetics ; 1: 34-40, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25705589

RESUMEN

An experience-dependent postnatal increase in GABAergic inhibition in the visual cortex is important for the closure of a critical period of enhanced synaptic plasticity. Although maturation of the subclass of Parvalbumin (Pv)-expressing GABAergic interneurons is known to contribute to critical period closure, the role of epigenetics on cortical inhibition and synaptic plasticity has not been explored. The transcription regulator, histone deacetylase 2 (HDAC2), has been shown to modulate synaptic plasticity and learning processes in hippocampal excitatory neurons. We found that genetic deletion of HDAC2 specifically from Pv-interneurons reduces inhibitory input in the visual cortex of adult mice, and coincides with enhanced long-term depression (LTD) that is more typical of young mice. These findings show that HDAC2 loss in Pv-interneurons leads to a delayed closure of the critical period in the visual cortex and supports the hypothesis that HDAC2 is a key negative regulator of synaptic plasticity in the adult brain.

19.
Proc Natl Acad Sci U S A ; 111(32): 11852-7, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25071187

RESUMEN

Understanding the mechanisms by which long-term memories are formed and stored in the brain represents a central aim of neuroscience. Prevailing theory suggests that long-term memory encoding involves early plasticity within hippocampal circuits, whereas reorganization of the neocortex is thought to occur weeks to months later to subserve remote memory storage. Here we report that long-term memory encoding can elicit early transcriptional, structural, and functional remodeling of the neocortex. Parallel studies using genome-wide RNA sequencing, ultrastructural imaging, and whole-cell recording in wild-type mice suggest that contextual fear conditioning initiates a transcriptional program in the medial prefrontal cortex (mPFC) that is accompanied by rapid expansion of the synaptic active zone and postsynaptic density, enhanced dendritic spine plasticity, and increased synaptic efficacy. To address the real-time contribution of the mPFC to long-term memory encoding, we performed temporally precise optogenetic inhibition of excitatory mPFC neurons during contextual fear conditioning. Using this approach, we found that real-time inhibition of the mPFC inhibited activation of the entorhinal-hippocampal circuit and impaired the formation of long-term associative memory. These findings suggest that encoding of long-term episodic memory is associated with early remodeling of neocortical circuits, identify the prefrontal cortex as a critical regulator of encoding-induced hippocampal activation and long-term memory formation, and have important implications for understanding memory processing in healthy and diseased brain states.


Asunto(s)
Memoria Episódica , Neocórtex/fisiología , Animales , Condicionamiento Psicológico/fisiología , Corteza Entorrinal/fisiología , Miedo/fisiología , Hipocampo/fisiología , Masculino , Memoria a Largo Plazo/fisiología , Ratones , Microscopía Electrónica de Transmisión , Potenciales Postsinápticos Miniatura/fisiología , Neocórtex/ultraestructura , Plasticidad Neuronal/fisiología , Optogenética , Corteza Prefrontal/fisiología , Corteza Prefrontal/ultraestructura , Transcriptoma
20.
Cell ; 157(2): 486-498, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725413

RESUMEN

Cyclin-dependent kinase 5 regulates numerous neuronal functions with its activator, p35. Under neurotoxic conditions, p35 undergoes proteolytic cleavage to liberate p25, which has been implicated in various neurodegenerative diseases. Here, we show that p25 is generated following neuronal activity under physiological conditions in a GluN2B- and CaMKIIα-dependent manner. Moreover, we developed a knockin mouse model in which endogenous p35 is replaced with a calpain-resistant mutant p35 (Δp35KI) to prevent p25 generation. The Δp35KI mice exhibit impaired long-term depression and defective memory extinction, likely mediated through persistent GluA1 phosphorylation at Ser845. Finally, crossing the Δp35KI mice with the 5XFAD mouse model of Alzheimer's disease (AD) resulted in an amelioration of ß-amyloid (Aß)-induced synaptic depression and cognitive impairment. Together, these results reveal a physiological role of p25 production in synaptic plasticity and memory and provide new insights into the function of p25 in Aß-associated neurotoxicity and AD-like pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Calpaína/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cognición , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Endocitosis , Técnicas de Sustitución del Gen , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Ratones , Proteínas del Tejido Nervioso/genética , Fosfotransferasas , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...