Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Genet ; 53: 263-288, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31518519

RESUMEN

Advances in human genetics have implicated a growing number of genes in neurodegenerative diseases, providing insight into pathological processes. For Alzheimer disease in particular, genome-wide association studies and gene expression studies have emphasized the pathogenic contributions from microglial cells and motivated studies of microglial function/dysfunction. Here, we summarize recent genetic evidence for microglial involvement in neurodegenerative disease with a focus on Alzheimer disease, for which the evidence is most compelling. To provide context for these genetic discoveries, we discuss how microglia influence brain development and homeostasis, how microglial characteristics change in disease, and which microglial activities likely influence the course of neurodegeneration. In all, we aim to synthesize varied aspects of microglial biology and highlight microglia as possible targets for therapeutic interventions in neurodegenerative disease.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Microglía/patología , Microglía/fisiología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/fisiología , Sistema Nervioso Central/metabolismo , Vía Clásica del Complemento/fisiología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Homeostasis , Humanos , Macrófagos/fisiología , Placa Amiloide/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo
2.
Cell ; 140(4): 567-78, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178748

RESUMEN

The molecular mechanisms regulating the ubiquitin proteasome system (UPS) at synapses are poorly understood. We report that CaMKIIalpha-an abundant postsynaptic protein kinase-mediates the activity-dependent recruitment of proteasomes to dendritic spines in hippocampal neurons. CaMKIIalpha is biochemically associated with proteasomes in the brain. CaMKIIalpha translocation to synapses is required for activity-induced proteasome accumulation in spines, and is sufficient to redistribute proteasomes to postsynaptic sites. CaMKIIalpha autophosphorylation enhances its binding to proteasomes and promotes proteasome recruitment to spines. In addition to this structural role, CaMKIIalpha stimulates proteasome activity by phosphorylating proteasome subunit Rpt6 on Serine 120. However, CaMKIIalpha translocation, but not its kinase activity, is required for activity-dependent degradation of polyubiquitinated proteins in spines. Our findings reveal a scaffolding role of postsynaptic CaMKIIalpha in activity-dependent proteasome redistribution, which is commensurate with the great abundance of CaMKIIalpha in synapses.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Espinas Dendríticas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Encéfalo/citología , Hipocampo/citología , Neuronas/citología , Fosforilación , Transporte de Proteínas , Ratas , Sinapsis
3.
Cell ; 141(5): 859-71, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20510932

RESUMEN

NMDA receptor-dependent synaptic modifications, such as long-term potentiation (LTP) and long-term depression (LTD), are essential for brain development and function. LTD occurs mainly by the removal of AMPA receptors from the postsynaptic membrane, but the underlying molecular mechanisms remain unclear. Here, we show that activation of caspase-3 via mitochondria is required for LTD and AMPA receptor internalization in hippocampal neurons. LTD and AMPA receptor internalization are blocked by peptide inhibitors of caspase-3 and -9. In hippocampal slices from caspase-3 knockout mice, LTD is abolished whereas LTP remains normal. LTD is also prevented by overexpression of the anti-apoptotic proteins XIAP or Bcl-xL, and by a mutant Akt1 protein that is resistant to caspase-3 proteolysis. NMDA receptor stimulation that induces LTD transiently activates caspase-3 in dendrites, without causing cell death. These data indicate an unexpected causal link between the molecular mechanisms of apoptosis and LTD.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Hipocampo/metabolismo , Depresión Sináptica a Largo Plazo , Receptores AMPA/metabolismo , Animales , Células Cultivadas , Citocromos c/metabolismo , Hipocampo/citología , Potenciación a Largo Plazo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína bcl-X/metabolismo
4.
Glia ; 71(5): 1247-1258, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36625077

RESUMEN

Disability in multiple sclerosis (MS) is driven in part by the failure of remyelination and progressive neurodegeneration. Microglia, and specifically triggering receptor expressed on myeloid cells 2 (TREM2), a factor highly expressed in microglia, have been shown to play an important role in remyelination. Here, using a focal demyelination model in the brain, we demonstrate that demyelination is persistent in TREM2 knockout mice, lasting more than 6 weeks after lysolecithin injection and resulting in substantial neurodegeneration. We also find that TREM2 knockout mice exhibit an altered glial response following demyelination. TREM2 knockout microglia demonstrate defects in migration and phagocytosis of myelin debris. In addition, human monocyte-derived macrophages from subjects with a TREM2 mutation prevalent in human disease also show a defect in myelin debris phagocytosis. Together, we highlight the central role of TREM2 signaling in remyelination and neuroprotection. These findings provide insights into how chronic demyelination might lead to axonal damage and could help identify novel neuroprotective therapeutic targets for MS.


Asunto(s)
Esclerosis Múltiple , Remielinización , Animales , Ratones , Humanos , Microglía/fisiología , Neuroprotección , Esclerosis Múltiple/tratamiento farmacológico , Vaina de Mielina , Ratones Noqueados , Ratones Endogámicos C57BL , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
5.
J Neurosci ; 40(5): 958-973, 2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31831521

RESUMEN

Cortical circuit activity is shaped by the parvalbumin (PV) and somatostatin (SST) interneurons that inhibit principal excitatory (EXC) neurons and the vasoactive intestinal peptide (VIP) interneurons that suppress activation of other interneurons. To understand the molecular-genetic basis of functional specialization and identify potential drug targets specific to each neuron subtype, we performed a genome wide assessment of both gene expression and splicing across EXC, PV, SST and VIP neurons from male and female mouse brains. These results reveal numerous examples where neuron subtype-specific gene expression, as well as splice-isoform usage, can explain functional differences between neuron subtypes, including in presynaptic plasticity, postsynaptic receptor function, and synaptic connectivity specification. We provide a searchable web resource for exploring differential mRNA expression and splice form usage between excitatory, PV, SST, and VIP neurons (http://research-pub.gene.com/NeuronSubtypeTranscriptomes). This resource, combining a unique new dataset and novel application of analysis methods to multiple relevant datasets, identifies numerous potential drug targets for manipulating circuit function, reveals neuron subtype-specific roles for disease-linked genes, and is useful for understanding gene expression changes observed in human patient brains.SIGNIFICANCE STATEMENT Understanding the basis of functional specialization of neuron subtypes and identifying drug targets for manipulating circuit function requires comprehensive information on cell-type-specific transcriptional profiles. We sorted excitatory neurons and key inhibitory neuron subtypes from mouse brains and assessed differential mRNA expression. We used a genome-wide analysis which not only examined differential gene expression levels but could also detect differences in splice isoform usage. This analysis reveals numerous examples of neuron subtype-specific isoform usage with functional importance, identifies potential drug targets, and provides insight into the neuron subtypes involved in psychiatric disease. We also apply our analysis to two other relevant datasets for comparison, and provide a searchable website for convenient access to the resource.


Asunto(s)
Corteza Cerebral/metabolismo , Interneuronas/metabolismo , Neuronas/metabolismo , Transcriptoma , Animales , Células Cultivadas , Femenino , Hipocampo/metabolismo , Masculino , Ratones Transgénicos , Parvalbúminas/metabolismo , ARN Mensajero/metabolismo , Somatostatina/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
6.
J Neurosci ; 40(9): 1956-1974, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31980586

RESUMEN

TREM2 is an Alzheimer's disease (AD) risk gene expressed in microglia. To study the role of Trem2 in a mouse model of ß-amyloidosis, we compared PS2APP transgenic mice versus PS2APP mice lacking Trem2 (PS2APP;Trem2ko) at ages ranging from 4 to 22 months. Microgliosis was impaired in PS2APP;Trem2ko mice, with Trem2-deficient microglia showing compromised expression of proliferation/Wnt-related genes and marked accumulation of ApoE. Plaque abundance was elevated in PS2APP;Trem2ko females at 6-7 months; but by 12 or 19-22 months of age, it was notably diminished in female and male PS2APP;Trem2ko mice, respectively. Across all ages, plaque morphology was more diffuse in PS2APP;Trem2ko brains, and the Aß42:Aß40 ratio was elevated. The amount of soluble, fibrillar Aß oligomers also increased in PS2APP;Trem2ko hippocampi. Associated with these changes, axonal dystrophy was exacerbated from 6 to 7 months onward in PS2APP;Trem2ko mice, notwithstanding the reduced plaque load at later ages. PS2APP;Trem2ko mice also exhibited more dendritic spine loss around plaque and more neurofilament light chain in CSF. Thus, aggravated neuritic dystrophy is a more consistent outcome of Trem2 deficiency than amyloid plaque load, suggesting that the microglial packing of Aß into dense plaque is an important neuroprotective activity.SIGNIFICANCE STATEMENT Genetic studies indicate that TREM2 gene mutations confer increased Alzheimer's disease (AD) risk. We studied the effects of Trem2 deletion in the PS2APP mouse AD model, in which overproduction of Aß peptide leads to amyloid plaque formation and associated neuritic dystrophy. Interestingly, neuritic dystrophies were intensified in the brains of Trem2-deficient mice, despite these mice displaying reduced plaque accumulation at later ages (12-22 months). Microglial clustering around plaques was impaired, plaques were more diffuse, and the Aß42:Aß40 ratio and amount of soluble, fibrillar Aß oligomers were elevated in Trem2-deficient brains. These results suggest that the Trem2-dependent compaction of Aß into dense plaques is a protective microglial activity, limiting the exposure of neurons to toxic Aß species.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Axones/patología , Espinas Dendríticas/patología , Glicoproteínas de Membrana/genética , Fragmentos de Péptidos/metabolismo , Placa Amiloide/genética , Receptores Inmunológicos/genética , Factor Trefoil-1/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Neuritas/patología , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Placa Amiloide/patología
7.
J Neurosci ; 39(24): 4636-4656, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-30948477

RESUMEN

In addition to amyloid-ß plaques and tau tangles, mitochondrial dysfunction is implicated in the pathology of Alzheimer's disease (AD). Neurons heavily rely on mitochondrial function, and deficits in brain energy metabolism are detected early in AD; however, direct human genetic evidence for mitochondrial involvement in AD pathogenesis is limited. We analyzed whole-exome sequencing data of 4549 AD cases and 3332 age-matched controls and discovered that rare protein altering variants in the gene pentatricopeptide repeat-containing protein 1 (PTCD1) show a trend for enrichment in cases compared with controls. We show here that PTCD1 is required for normal mitochondrial rRNA levels, proper assembly of the mitochondrial ribosome and hence for mitochondrial translation and assembly of the electron transport chain. Loss of PTCD1 function impairs oxidative phosphorylation and forces cells to rely on glycolysis for energy production. Cells expressing the AD-linked variant of PTCD1 fail to sustain energy production under increased metabolic stress. In neurons, reduced PTCD1 expression leads to lower ATP levels and impacts spontaneous synaptic activity. Thus, our study uncovers a possible link between a protein required for mitochondrial function and energy metabolism and AD risk.SIGNIFICANCE STATEMENT Mitochondria are the main source of cellular energy and mitochondrial dysfunction is implicated in the pathology of Alzheimer's disease (AD) and other neurodegenerative disorders. Here, we identify a variant in the gene PTCD1 that is enriched in AD patients and demonstrate that PTCD1 is required for ATP generation through oxidative phosphorylation. PTCD1 regulates the level of 16S rRNA, the backbone of the mitoribosome, and is essential for mitochondrial translation and assembly of the electron transport chain. Cells expressing the AD-associated variant fail to maintain adequate ATP production during metabolic stress, and reduced PTCD1 activity disrupts neuronal energy homeostasis and dampens spontaneous transmission. Our work provides a mechanistic link between a protein required for mitochondrial function and genetic AD risk.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosforilación Oxidativa , Adenosina Trifosfato/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético/genética , Técnicas de Inactivación de Genes , Variación Genética , Glucólisis/genética , Células HeLa , Humanos , Estrés Oxidativo , Ribosomas/metabolismo , Estrés Fisiológico/genética
8.
Nature ; 510(7505): 370-5, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24896179

RESUMEN

Cells maintain healthy mitochondria by degrading damaged mitochondria through mitophagy; defective mitophagy is linked to Parkinson's disease. Here we report that USP30, a deubiquitinase localized to mitochondria, antagonizes mitophagy driven by the ubiquitin ligase parkin (also known as PARK2) and protein kinase PINK1, which are encoded by two genes associated with Parkinson's disease. Parkin ubiquitinates and tags damaged mitochondria for clearance. Overexpression of USP30 removes ubiquitin attached by parkin onto damaged mitochondria and blocks parkin's ability to drive mitophagy, whereas reducing USP30 activity enhances mitochondrial degradation in neurons. Global ubiquitination site profiling identified multiple mitochondrial substrates oppositely regulated by parkin and USP30. Knockdown of USP30 rescues the defective mitophagy caused by pathogenic mutations in parkin and improves mitochondrial integrity in parkin- or PINK1-deficient flies. Knockdown of USP30 in dopaminergic neurons protects flies against paraquat toxicity in vivo, ameliorating defects in dopamine levels, motor function and organismal survival. Thus USP30 inhibition is potentially beneficial for Parkinson's disease by promoting mitochondrial clearance and quality control.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Mitofagia/fisiología , Tioléster Hidrolasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Masculino , Proteínas Mitocondriales/genética , Neuronas/metabolismo , Enfermedad de Parkinson/fisiopatología , Proteínas Quinasas/metabolismo , Ratas , Tioléster Hidrolasas/genética , Ubiquitina-Proteína Ligasas/genética , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación
9.
J Neurosci ; 38(23): 5289-5301, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29735556

RESUMEN

Mutations or altered protein levels of SHANK3 are implicated in neurodevelopmental disorders such as Phelan-McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre et al., 2014). Loss of SHANK3 in mouse models results in decreased synapse density and reduction in the levels of multiple synaptic proteins (Jiang and Ehlers, 2013). The family of SHANK scaffolding molecules are among the most heavily ubiquitinated proteins at the postsynaptic density. The ubiquitin-dependent proteasome degradation of SHANK is regulated by synaptic activity and may contribute to activity-dependent synaptic remodeling (Ehlers, 2003; Shin et al., 2012). However, the identity of the specific deubiquitinating enzymes and E3 ligases that regulate SHANK ubiquitination at synapses are unknown. Here we identify USP8/UBPY as a deubiquitinating enzyme that regulates SHANK3 and SHANK1 ubiquitination and protein levels. In primary rat neurons, USP8 enhances SHANK3 and SHANK1 protein levels via deubiquitination and increases dendritic spine density. Additionally, USP8 is essential for changes in SHANK3 protein levels following synaptic activity modulation. These data identify USP8 as a key modulator of SHANK3 downstream of synaptic activity.SIGNIFICANCE STATEMENT Precise regulation of the protein levels of the postsynaptic scaffolding protein SHANK3 is essential for proper neurodevelopment. Mutations of SHANK3 have been identified in Phelan-McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre et al., 2014). In this research, we identify USP8 as a key enzyme that regulates SHANK3 protein levels in neurons. USP8 acts to deubiquitinate SHANK3, which prevents its proteasomal-mediated degradation and enhances overall dendritic spine stability. In the future, the modulation of USP8 deubiquitinating activity could potentially be used to titrate the protein levels of SHANK3 to ameliorate disease.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Animales , Endopeptidasas , Complejos de Clasificación Endosomal Requeridos para el Transporte , Células HEK293 , Humanos , Ratas , Ratas Sprague-Dawley , Ubiquitina Tiolesterasa , Ubiquitinación
10.
J Neurosci ; 36(38): 9962-75, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27656033

RESUMEN

UNLABELLED: After traumatic brain injury (TBI), neurons surviving the initial insult can undergo chronic (secondary) degeneration via poorly understood mechanisms, resulting in long-term cognitive impairment. Although a neuroinflammatory response is promptly activated after TBI, it is unknown whether it has a significant role in chronic phases of TBI (>1 year after injury). Using a closed-head injury model of TBI in mice, we showed by MRI scans that TBI caused substantial degeneration at the lesion site within a few weeks and these did not expand significantly thereafter. However, chronic alterations in neurons were observed, with reduced dendritic spine density lasting >1 year after injury. In parallel, we found a long-lasting inflammatory response throughout the entire brain. Deletion of one allele of CX3CR1, a chemokine receptor, limited infiltration of peripheral immune cells and largely prevented the chronic degeneration of the injured brain and provided a better functional recovery in female, but not male, mice. Therefore, targeting persistent neuroinflammation presents a new therapeutic option to reduce chronic neurodegeneration. SIGNIFICANCE STATEMENT: Traumatic brain injury (TBI) often causes chronic neurological problems including epilepsy, neuropsychiatric disorders, and dementia through unknown mechanisms. Our study demonstrates that inflammatory cells invading the brain lead to secondary brain damage. Sex-specific amelioration of chronic neuroinflammation rescues the brain degeneration and results in improved motor functions. Therefore, this study pinpoints an effective therapeutic approach to preventing secondary complications after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Inflamación/etiología , Degeneración Nerviosa , Recuperación de la Función/fisiología , Animales , Encéfalo/patología , Receptor 1 de Quimiocinas CX3C , Proteínas de Unión al Calcio/metabolismo , Enfermedad Crónica , Espinas Dendríticas/inmunología , Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Actividad Motora , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Desempeño Psicomotor/fisiología , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Factores de Tiempo
11.
J Neurosci ; 35(5): 2118-32, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25653368

RESUMEN

The ability to attend to relevant stimuli and to adapt dynamically as demands change is a core aspect of cognition, and one that is impaired in several neuropsychiatric diseases, including attention deficit/hyperactivity disorder. However, the cellular and molecular mechanisms underlying such cognitive adaptability are poorly understood. We found that deletion of the caspase-3 gene, encoding an apoptosis protease with newly discovered roles in neural plasticity, disrupts attention in mice while preserving multiple learning and memory capabilities. Attention-related deficits include distractibility, impulsivity, behavioral rigidity, and reduced habituation to novel stimuli. Excess exploratory activity in Casp3(-/-) mice was correlated with enhanced novelty-induced activity in the dentate gyrus, which may be related to our findings that caspase-3 is required for homeostatic synaptic plasticity in vitro and homeostatic expression of AMPA receptors in vivo in response to chronic or repeated stimuli. These results suggest an important role for caspase-3 in synaptic suppression of irrelevant stimuli.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Atención , Caspasa 3/deficiencia , Homeostasis , Sinapsis/fisiología , Animales , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Caspasa 3/genética , Giro Dentado/metabolismo , Giro Dentado/fisiología , Eliminación de Gen , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal
12.
J Neurosci ; 34(5): 1672-88, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24478350

RESUMEN

Synapse loss occurs normally during development and pathologically during neurodegenerative disease. Long-term depression, a proposed physiological correlate of synapse elimination, requires caspase-3 and the mitochondrial pathway of apoptosis. Here, we show that caspase-3 activity is essential--and can act locally within neurons--for regulation of spine density and dendrite morphology. By photostimulation of Mito-KillerRed, we induced caspase-3 activity in defined dendritic regions of cultured neurons. Within the photostimulated region, local elimination of dendritic spines and dendrite retraction occurred in a caspase-3-dependent manner without inducing cell death. However, pharmacological inhibition of inhibitor of apoptosis proteins or proteasome function led to neuronal death, suggesting that caspase activation is spatially restricted by these "molecular brakes" on apoptosis. Caspase-3 knock-out mice have increased spine density and altered miniature EPSCs, confirming a physiological involvement of caspase-3 in the regulation of spines in vivo.


Asunto(s)
Caspasa 3/metabolismo , Dendritas/ultraestructura , Espinas Dendríticas/fisiología , Neuronas/citología , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Factores de Edad , Animales , Caspasa 3/genética , Caspasa 9/metabolismo , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Dendritas/fisiología , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/enzimología , Sinapsis/inmunología , Factores de Tiempo
13.
J Neurosci ; 34(46): 15327-39, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25392500

RESUMEN

Neuronal gene expression is modulated by activity via calcium-permeable receptors such as NMDA receptors (NMDARs). While gene expression changes downstream of evoked NMDAR activity have been well studied, much less is known about gene expression changes that occur under conditions of basal neuronal activity. In mouse dissociated hippocampal neuronal cultures, we found that a broad NMDAR antagonist, AP5, induced robust gene expression changes under basal activity, but subtype-specific antagonists did not. While some of the gene expression changes are also known to be downstream of stimulated NMDAR activity, others appear specific to basal NMDAR activity. The genes altered by AP5 treatment of basal cultures were enriched for pathways related to class IIa histone deacetylases (HDACs), apoptosis, and synapse-related signaling. Specifically, AP5 altered the expression of all three class IIa HDACs that are highly expressed in the brain, HDAC4, HDAC5, and HDAC9, and also induced nuclear accumulation of HDAC4. HDAC4 knockdown abolished a subset of the gene expression changes induced by AP5, and led to neuronal death under long-term tetrodotoxin or AP5 treatment in rat hippocampal organotypic slice cultures. These data suggest that basal, but not evoked, NMDAR activity regulates gene expression in part through HDAC4, and, that HDAC4 has neuroprotective functions under conditions of low NMDAR activity.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histona Desacetilasas/fisiología , Neuronas/citología , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiología , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Masculino , Ratones , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Ratas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Tetrodotoxina/toxicidad , Valina/análogos & derivados , Valina/farmacología
14.
Proc Natl Acad Sci U S A ; 109(42): 17093-8, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23027931

RESUMEN

The memory of fear extinction is context dependent: fear that is suppressed in one context readily renews in another. Understanding of the underlying neuronal circuits is, therefore, of considerable clinical relevance for anxiety disorders. Prefrontal cortical and hippocampal inputs to the amygdala have recently been shown to regulate the retrieval of fear memories, but the cellular organization of these projections remains unclear. By using anterograde tracing in a transgenic rat in which neurons express a dendritically-targeted PSD-95:Venus fusion protein under the control of a c-fos promoter, we found that, during the retrieval of extinction memory, the dominant input to active neurons in the lateral amygdala was from the infralimbic cortex, whereas the retrieval of fear memory was associated with greater hippocampal and prelimbic inputs. This pattern of retrieval-related afferent input was absent in the central nucleus of the amygdala. Our data show functional anatomy of neural circuits regulating fear and extinction, providing a framework for therapeutic manipulations of these circuits.


Asunto(s)
Amígdala del Cerebelo/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Sistema Límbico/fisiología , Memoria/fisiología , Vías Nerviosas/anatomía & histología , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Varianza , Animales , Proteínas Bacterianas/metabolismo , Condicionamiento Psicológico , Cartilla de ADN/genética , Homólogo 4 de la Proteína Discs Large , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Luminiscentes/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Vías Nerviosas/fisiología , Ratas , Ratas Transgénicas , Grabación en Video
15.
J Neurosci ; 33(8): 3612-23, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23426688

RESUMEN

Synaptic transmission depends on the matching and alignment of presynaptically released transmitters and postsynaptic neurotransmitter receptors. Neuroligin (NL) and Neurexin (Nrxn) proteins are trans-synaptic adhesion molecules that are important in validation and maturation of specific synapses. NL isoforms NL1 and NL2 have specific functional roles in excitatory and inhibitory synapses, respectively, but the molecular basis behind this distinction is still unclear. We show here that the extracellular domain of NL2 confers its unique ability to enhance inhibitory synaptic function when overexpressed in rat hippocampal pyramidal neurons, whereas NL1 normally only promotes excitatory synapses. This specificity is conferred by presynaptic Nrxn isoforms, as NL1 can also induce functional inhibitory synapse connections when the presynaptic interneurons ectopically express an Nrxn isoform that binds to NL1. Our results indicate that trans-synaptic interaction with differentially expressed presynaptic Nrxns underlies the distinct functions of NL1 and NL2, and is sufficient to induce functional inhibitory synapse formation.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Interneuronas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural/fisiología , Receptores de Superficie Celular/metabolismo , Sinapsis/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/fisiología , Células Cultivadas , Femenino , Células HEK293 , Hipocampo/metabolismo , Hipocampo/fisiología , Humanos , Interneuronas/fisiología , Masculino , Proteínas del Tejido Nervioso/fisiología , Técnicas de Cultivo de Órganos , Terminales Presinápticos/metabolismo , Terminales Presinápticos/fisiología , Unión Proteica/fisiología , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Células Piramidales/fisiología , Ratas , Receptores de Superficie Celular/fisiología , Sinapsis/metabolismo
16.
J Neurosci ; 33(29): 12122-35, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864697

RESUMEN

Activity of glycogen synthase kinase-3ß (GSK-3ß) is required for long-term depression (LTD) via molecular mechanisms that are incompletely understood. Here, we report that PSD-95, a major scaffold protein of the postsynaptic density (PSD) that promotes synaptic strength, is phosphorylated on threonine-19 (T19) by GSK-3ß. In cultured rat hippocampal neurons, phosphorylation of T19 increases rapidly with chemical LTD and is attenuated by pharmacologic or genetic suppression of GSK-3ß. In organotypic rat hippocampal slices, we find that a nonphosphorylatable PSD-95 mutant (T19A) tagged with photoactivatable green fluorescent protein (PAGFP) shows enhanced stability in dendritic spines versus wild-type PSD-95, whereas the phosphomimetic mutant (PSD-95-T19D) is more readily dispersed. Further, overexpression of PSD-95-T19A, but not WT-PSD-95, impairs AMPA receptor internalization and the induction of LTD. These data indicate that phosphorylation on T19 by GSK-3ß destabilizes PSD-95 within the PSD and is a critical step for AMPA receptor mobilization and LTD.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Treonina/metabolismo , Animales , Homólogo 4 de la Proteína Discs Large , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Indoles/farmacología , Cloruro de Litio/farmacología , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Maleimidas/farmacología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Receptores AMPA/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sinapsis/efectos de los fármacos
18.
Nat Rev Drug Discov ; 23(1): 23-42, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38012296

RESUMEN

Synapse dysfunction and loss are hallmarks of neurodegenerative diseases that correlate with cognitive decline. However, the mechanisms and therapeutic strategies to prevent or reverse synaptic damage remain elusive. In this Review, we discuss recent advances in understanding the molecular and cellular pathways that impair synapses in neurodegenerative diseases, including the effects of protein aggregation and neuroinflammation. We also highlight emerging therapeutic approaches that aim to restore synaptic function and integrity, such as enhancing synaptic plasticity, preventing synaptotoxicity, modulating neuronal network activity and targeting immune signalling. We discuss the preclinical and clinical evidence for each strategy, as well as the challenges and opportunities for developing effective synapse-targeting therapeutics for neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Sinapsis/metabolismo , Transducción de Señal , Plasticidad Neuronal
19.
Sci Rep ; 14(1): 2798, 2024 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307912

RESUMEN

Human genetic studies have revealed rare missense and protein-truncating variants in GRIN2A, encoding for the GluN2A subunit of the NMDA receptors, that confer significant risk for schizophrenia (SCZ). Mutations in GRIN2A are also associated with epilepsy and developmental delay/intellectual disability (DD/ID). However, it remains enigmatic how alterations to the same protein can result in diverse clinical phenotypes. Here, we performed functional characterization of human GluN1/GluN2A heteromeric NMDA receptors that contain SCZ-linked GluN2A variants, and compared them to NMDA receptors with GluN2A variants associated with epilepsy or DD/ID. Our findings demonstrate that SCZ-associated GRIN2A variants were predominantly loss-of-function (LoF), whereas epilepsy and DD/ID-associated variants resulted in both gain- and loss-of-function phenotypes. We additionally show that M653I and S809R, LoF GRIN2A variants associated with DD/ID, exert a dominant-negative effect when co-expressed with a wild-type GluN2A, whereas E58Ter and Y698C, SCZ-linked LoF variants, and A727T, an epilepsy-linked LoF variant, do not. These data offer a potential mechanism by which SCZ/epilepsy and DD/ID-linked variants can cause different effects on receptor function and therefore result in divergent pathological outcomes.


Asunto(s)
Epilepsia , Trastornos del Neurodesarrollo , Esquizofrenia , Humanos , Epilepsia/genética , Mutación , Trastornos del Neurodesarrollo/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/genética
20.
PLoS Biol ; 8(1): e1000283, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20098723

RESUMEN

The endosomal pathway in neuronal dendrites is essential for membrane receptor trafficking and proper synaptic function and plasticity. However, the molecular mechanisms that organize specific endocytic trafficking routes are poorly understood. Here, we identify GRIP-associated protein-1 (GRASP-1) as a neuron-specific effector of Rab4 and key component of the molecular machinery that coordinates recycling endosome maturation in dendrites. We show that GRASP-1 is necessary for AMPA receptor recycling, maintenance of spine morphology, and synaptic plasticity. At the molecular level, GRASP-1 segregates Rab4 from EEA1/Neep21/Rab5-positive early endosomal membranes and coordinates the coupling to Rab11-labelled recycling endosomes by interacting with the endosomal SNARE syntaxin 13. We propose that GRASP-1 connects early and late recycling endosomal compartments by forming a molecular bridge between Rab-specific membrane domains and the endosomal SNARE machinery. The data uncover a new mechanism to achieve specificity and directionality in neuronal membrane receptor trafficking.


Asunto(s)
Dendritas/metabolismo , Endosomas/metabolismo , Proteínas de Unión al GTP rab4/metabolismo , Animales , Transporte Biológico , Células COS , Proteínas Portadoras/análisis , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Chlorocebus aethiops , Dendritas/ultraestructura , Escherichia coli/genética , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/fisiología , Ratones , Plasticidad Neuronal , Proteínas Qa-SNARE/metabolismo , Ratas , Receptores de Glutamato/metabolismo , Porcinos , Proteínas de Unión al GTP rab4/análisis , Proteínas de Unión al GTP rab4/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA