Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Res ; 158: 104863, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32407957

RESUMO

Neural stem cell (NSC) neuronal differentiation requires a metabolic shift towards oxidative phosphorylation. We now show that a branched-chain amino acids-driven, persistent metabolic shift toward energy metabolism is required for full neuronal maturation. We increased energy metabolism of differentiating neurons derived both from murine NSCs and human induced pluripotent stem cells (iPSCs) by supplementing the cell culture medium with a mixture composed of branched-chain amino acids, essential amino acids, TCA cycle precursors and co-factors. We found that treated differentiating neuronal cells with enhanced energy metabolism increased: i) total dendritic length; ii) the mean number of branches and iii) the number and maturation of the dendritic spines. Furthermore, neuronal spines in treated neurons appeared more stable with stubby and mushroom phenotype and with increased expression of molecules involved in synapse formation. Treated neurons modified their mitochondrial dynamics increasing the mitochondrial fusion and, consistently with the increase of cellular ATP content, they activated cellular mTORC1 dependent p70S6 K1 anabolism. Global transcriptomic analysis further revealed that treated neurons induce Nrf2 mediated gene expression. This was correlated with a functional increase in the Reactive Oxygen Species (ROS) scavenging mechanisms. In conclusion, persistent branched-chain amino acids-driven metabolic shift toward energy metabolism enhanced neuronal differentiation and antioxidant defences. These findings offer new opportunities to pharmacologically modulate NSC neuronal differentiation and to develop effective strategies for treating neurodegenerative diseases.


Assuntos
Aminoácidos de Cadeia Ramificada/farmacologia , Diferenciação Celular/fisiologia , Metabolismo Energético/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/ultraestrutura , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sinapses/genética , Sinapses/fisiologia , Sinapses/ultraestrutura , Transcriptoma
2.
Artigo em Inglês | MEDLINE | ID: mdl-31715283

RESUMO

Genetic risk for schizophrenia is due to the joint effect of multiple genes acting mainly at two different processes, prenatal/perinatal neurodevelopment and adolescence/early adulthood synapse maturation. Identification of important genes at the second process is of relevance for early intervention. The aim of this work was to identify gene co-expression modules with altered expression in schizophrenia during adolescence/early adulthood. To this goal, we predicted frontal cortex gene expression in one discovery sample, the largest GWAS of schizophrenia from the Psychiatric Genomics Consortium, using S-prediXcan, and in one target sample, consisting of 625 schizophrenic patients and 819 controls from Spain, using prediXcan. Prediction models were trained on GTEx frontal cortex expression dataset. In parallel, we identified brain co-expression modules from BrainSpan using WGCNA. Then, we estimated polygenic risk scores based on predicted expression (PE-PRS) for each co-expression module in the target sample, based on PE-PRS model from the discovery sample. This analysis led to the identification of a module with mainly adolescence/adulthood expression whose PE-PRS was significantly associated with schizophrenia. The module was significantly enriched in synaptic processes. Several hub genes at this module are drugabble, according to the drug-gene interaction database, and/or involved in synaptic transmission, such as the voltage-gated ion channels SCN2B and KCNAB2, the calcium calmodulin kinases CAMK2A and CAMK1G, or genes involved in synaptic vesicle cycle, such as DNM1, or SYNGR1. Therefore, identification of this module may be the first step in patient stratification based on biology, as well as in drug design and drug repurposing efforts.


Assuntos
Regulação da Expressão Gênica , Esquizofrenia/genética , Adolescente , Antipsicóticos/química , Antipsicóticos/farmacologia , Desenho de Fármacos , Feminino , Lobo Frontal/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Herança Multifatorial , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Espanha , Sinapses/genética , Transcriptoma , Adulto Jovem
3.
Hum Mol Genet ; 28(22): 3842-3852, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31625566

RESUMO

Ubiquitin E3 ligase 3A (UBE3A) encodes an E3 ubiquitin ligase whose loss from the maternal allele causes the neurodevelopmental disorder Angelman syndrome (AS). Previous studies of UBE3A function have not examined full Ube3a deletion in mouse, the complexity of imprinted gene networks in brain nor the molecular basis of systems-level cognitive dysfunctions in AS. We therefore utilized a systems biology approach to elucidate how UBE3A loss impacts the early postnatal brain in a novel CRISPR/Cas9-engineered rat Angelman model of a complete Ube3a deletion. Strand-specific transcriptome analysis of offspring from maternally or paternally inherited Ube3a deletions revealed the expected parental expression patterns of Ube3a sense and antisense transcripts by postnatal day 2 (P2) in hypothalamus and day 9 (P9) in cortex, compared to wild-type littermates. The dependency of genome-wide effects on parent-of-origin, Ube3a genotype and time (P2 and P9) was investigated through transcriptome (RNA sequencing of cortex and hypothalamus) and methylome (whole-genome bisulfite sequencing of hypothalamus). Weighted gene co-expression and co-methylation network analyses identified co-regulated networks in maternally inherited Ube3a deletion offspring enriched in postnatal developmental processes including Wnt signaling, synaptic regulation, neuronal and glial functions, epigenetic regulation, ubiquitin, circadian entrainment and splicing. Furthermore, we showed that loss of the paternal Ube3a antisense transcript resulted in both unique and overlapping dysregulated gene pathways with maternal loss, predominantly at the level of differential methylation. Together, these results provide a holistic examination of the molecular impacts of UBE3A loss in brain, supporting the existence of interactive epigenetic networks between maternal and paternal transcripts at the Ube3a locus.


Assuntos
Impressão Genômica , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Epigênese Genética , Feminino , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes/genética , Hipotálamo/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Sinapses/genética , Sinapses/metabolismo , Biologia de Sistemas , Transcriptoma , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt
4.
Neurosci Lett ; 709: 134374, 2019 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-31310785

RESUMO

Sensory cortex topographic maps consist of organized arrays of thalamocortical afferents (TCAs) that project into distinct areas of the cortex. Formation of topographic maps in sensory cortices is a prerequisite for functional maturation of the neocortex. Studies have shown that the formation of topographic maps and the maturation of thalamocortical synapses in the somatosensory cortex depend on the cyclic adenosine 5'-monophosphate-(cAMP)-protein kinase A (PKA) signaling pathway. AKAP5 is a scaffold protein (also called AKAP79 in humans or AKAP150 in rodents; AKAP79/150) that serves as a signaling hub that links cAMP and PKA signaling. Whether AKAP5 plays a role in topographic map formation and the maturation of thalamocortical synapses during development of the somatosensory cortex is still unknown. Here, we generated cortex-specific AKAP5-knockout mice (CxAKAP5KO) to examine its roles in somatosensory cortex development. We found that CxAKAP5KO mice displayed impaired cortical barrel maps. Electrophysiological recordings showed that the AMPA/NMDA ratio was reduced, and silent synapses were increased in thalamocortical synapses of CxAKAP5KO mice during postnatal development. Morphological analysis of layer IV cortical neurons demonstrated that dendritic refinement of these neurons was abnormal. These results indicate that AKAP5 is necessary for both topographic map formation and maturation of thalamocortical synapses as well as morphological development of cortical neurons in the somatosensory cortex.


Assuntos
Proteínas de Ancoragem à Quinase A/biossíntese , Neocórtex/metabolismo , Córtex Somatossensorial/metabolismo , Sinapses/metabolismo , Tálamo/metabolismo , Proteínas de Ancoragem à Quinase A/deficiência , Proteínas de Ancoragem à Quinase A/genética , Animais , Expressão Gênica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neocórtex/citologia , Vias Neurais/citologia , Vias Neurais/metabolismo , Córtex Somatossensorial/citologia , Sinapses/genética , Tálamo/citologia
5.
Biochem Biophys Res Commun ; 509(2): 429-434, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30594389

RESUMO

Appropriate synapse formation during development is necessary for normal brain function, and synapse impairment is often associated with brain dysfunction. Brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) are key factors in regulating synaptic development. We previously reported that BDNF/NT-3 secretion was enhanced by calcium-dependent activator protein for secretion 2 (CADPS2). Although BDNF/NT-3 and CADPS2 are co-expressed in various brain regions, the effect of Cadps2-deficiency on brain region-specific BDNF/NT-3 levels and synaptic development remains elusive. Here, we show developmental changes of BDNF/NT-3 levels and we assess disruption of excitatory/inhibitory synapses in multiple brain regions (cerebellum, hypothalamus, striatum, hippocampus, parietal cortex and prefrontal cortex) of Cadps2 knockout (KO) mice compared with wild-type (WT) mice. Compared with WT, BDNF levels in KO mice were reduced in young/adult hippocampus, but increased in young hypothalamus, while NT-3 levels were reduced in adult cerebellum and young hippocampus, but increased in adult parietal cortex. Immunofluorescence of vGluT1, an excitatory synapse marker, and vGAT, an inhibitory synapse marker, in adult KO showed that vGluT1 was higher in the cerebellum and parietal cortex but lower in the hippocampus, whereas vGAT was lower in the hippocampus and parietal cortex compared with WT. Immunolabeling for both vGluT1 and vGAT was increased in the parietal cortex but vGAT was decreased in the cerebellum in adult KO compared with WT. These data suggest that CADPS2-mediated secretion of BDNF/NT-3 may be involved in development and maturation of synapses and in the balance between inhibitory and excitatory synapses.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Proteínas de Ligação ao Cálcio/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurotrofina 3/genética , Sinapses/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/deficiência , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Corpo Estriado/citologia , Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/metabolismo , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurônios/citologia , Neurotrofina 3/metabolismo , Especificidade de Órgãos , Lobo Parietal/citologia , Lobo Parietal/crescimento & desenvolvimento , Lobo Parietal/metabolismo , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/metabolismo , Sinapses/classificação , Sinapses/metabolismo , Transmissão Sináptica/genética , Proteína Vesicular 1 de Transporte de Glutamato/genética , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
6.
Mol Autism ; 8: 34, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28702161

RESUMO

Autism spectrum disorder (ASD) is a group of complex neurodevelopmental disorders with diverse clinical manifestations and symptoms. In the last 10 years, there have been significant advances in understanding the genetic basis for ASD, critically supported through the establishment of ASD bio-collections and application in research. Here, we summarise a selection of major ASD bio-collections and their associated findings. Collectively, these include mapping ASD candidate genes, assessing the nature and frequency of gene mutations and their association with ASD clinical subgroups, insights into related molecular pathways such as the synapses, chromatin remodelling, transcription and ASD-related brain regions. We also briefly review emerging studies on the use of induced pluripotent stem cells (iPSCs) to potentially model ASD in culture. These provide deeper insight into ASD progression during development and could generate human cell models for drug screening. Finally, we provide perspectives concerning the utilities of ASD bio-collections and limitations, and highlight considerations in setting up a new bio-collection for ASD research.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Encéfalo , Montagem e Desmontagem da Cromatina/genética , Mapeamento Cromossômico , Sinapses , Transcrição Gênica/genética , Transtorno do Espectro Autista/tratamento farmacológico , Encéfalo/metabolismo , Encéfalo/patologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Sinapses/genética , Sinapses/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(7): 1684-1689, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28143929

RESUMO

Experience-driven synaptic plasticity is believed to underlie adaptive behavior by rearranging the way neuronal circuits process information. We have previously discovered that O-GlcNAc transferase (OGT), an enzyme that modifies protein function by attaching ß-N-acetylglucosamine (GlcNAc) to serine and threonine residues of intracellular proteins (O-GlcNAc), regulates food intake by modulating excitatory synaptic function in neurons in the hypothalamus. However, how OGT regulates excitatory synapse function is largely unknown. Here we demonstrate that OGT is enriched in the postsynaptic density of excitatory synapses. In the postsynaptic density, O-GlcNAcylation on multiple proteins increased upon neuronal stimulation. Knockout of the OGT gene decreased the synaptic expression of the AMPA receptor GluA2 and GluA3 subunits, but not the GluA1 subunit. The number of opposed excitatory presynaptic terminals was sharply reduced upon postsynaptic knockout of OGT. There were also fewer and less mature dendritic spines on OGT knockout neurons. These data identify OGT as a molecular mechanism that regulates synapse maturity.


Assuntos
Hipotálamo/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Células Cultivadas , Espinhas Dendríticas/metabolismo , Potenciais Pós-Sinápticos Excitadores/genética , Hipotálamo/citologia , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , Plasticidade Neuronal/genética , Terminações Pré-Sinápticas/metabolismo , Ratos , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Sinapses/genética , Transmissão Sináptica/genética
8.
J Investig Med ; 64(8): 1220-1234, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27521081

RESUMO

The purpose of our study was to investigate the protective effects of a natural product-'curcumin'- in Alzheimer's disease (AD)-like neurons. Although much research has been done in AD, very little has been reported on the effects of curcumin on mitochondrial biogenesis, dynamics, function and synaptic activities. Therefore, the present study investigated the protective effects against amyloid ß (Aß) induced mitochondrial and synaptic toxicities. Using human neuroblastoma (SHSY5Y) cells, curcumin and Aß, we studied the protective effects of curcumin against Aß. Further, we also studied preventive (curcumin+Aß) and intervention (Aß+curcumin) effects of curcumin against Aß in SHSY5Y cells. Using real time RT-PCR, immunoblotting and immunofluorescence analysis, we measured mRNA and protein levels of mitochondrial dynamics, mitochondrial biogenesis and synaptic genes. We also assessed mitochondrial function by measuring hydrogen peroxide, lipid peroxidation, cytochrome oxidase activity and mitochondrial ATP. Cell viability was studied using the MTT assay. Aß was found to impair mitochondrial dynamics, reduce mitochondrial biogenesis and decrease synaptic activity and mitochondrial function. In contrast, curcumin enhanced mitochondrial fusion activity and reduced fission machinery, and increased biogenesis and synaptic proteins. Mitochondrial function and cell viability were elevated in curcumin treated cells. Interestingly, curcumin pre- and post-treated cells incubated with Aß showed reduced mitochondrial dysfunction, and maintained cell viability and mitochondrial dynamics, mitochondrial biogenesis and synaptic activity. Further, the protective effects of curcumin were stronger in pretreated SHSY5Y cells than in post-treated cells, indicating that curcumin works better in prevention than treatment in AD-like neurons. Our findings suggest that curcumin is a promising drug molecule to treat AD patients.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/toxicidade , Produtos Biológicos/uso terapêutico , Curcumina/uso terapêutico , Mitocôndrias/patologia , Fármacos Neuroprotetores/uso terapêutico , Sinapses/patologia , Doença de Alzheimer/patologia , Produtos Biológicos/farmacologia , Linhagem Celular Tumoral , Curcumina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Dinâmica Mitocondrial/genética , Fármacos Neuroprotetores/farmacologia , Biogênese de Organelas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/genética , Sinapses/metabolismo
9.
Neuron ; 86(3): 768-82, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25913856

RESUMO

Corticothalamic neurons provide massive input to the thalamus. This top-down projection may allow the cortex to regulate sensory processing by modulating the excitability of thalamic cells. Layer 6 corticothalamic neurons monosynaptically excite thalamocortical cells, but also indirectly inhibit them by driving inhibitory cells of the thalamic reticular nucleus. Whether corticothalamic activity generally suppresses or excites the thalamus remains unclear. Here we show that the corticothalamic influence is dynamic, with the excitatory-inhibitory balance shifting in an activity-dependent fashion. During low-frequency activity, corticothalamic effects are mainly suppressive, whereas higher-frequency activity (even a short bout of gamma frequency oscillations) converts the corticothalamic influence to enhancement. The mechanism of this switching depends on distinct forms of short-term synaptic plasticity across multiple corticothalamic circuit components. Our results reveal an activity-dependent mechanism by which corticothalamic neurons can bidirectionally switch the excitability and sensory throughput of the thalamus, possibly to meet changing behavioral demands.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Vias Neurais/fisiologia , Córtex Somatossensorial/fisiologia , Sinapses/fisiologia , Tálamo/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Channelrhodopsins , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Optogenética , Picrotoxina/farmacologia , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo , Sinapses/genética , Valina/análogos & derivados , Valina/farmacologia , Ácido gama-Aminobutírico/farmacologia
10.
Mov Disord ; 29(13): 1655-65, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25195914

RESUMO

Broad-spectrum muscarinic receptor antagonists have represented the first available treatment for different movement disorders such as dystonia. However, the specificity of these drugs and their mechanism of action is not entirely clear. We performed a systematic analysis of the effects of anticholinergic drugs on short- and long-term plasticity recorded from striatal medium spiny neurons from DYT1 dystonia knock-in (Tor1a(+/Δgag) ) mice heterozygous for ΔE-torsinA and their controls (Tor1a(+/+) mice). Antagonists were chosen that had previously been proposed to be selective for muscarinic receptor subtypes and included pirenzepine, trihexyphenydil, biperiden, orphenadrine, and a novel selective M1 antagonist, VU0255035. Tor1a(+/Δgag) mice exhibited a significant impairment of corticostriatal synaptic plasticity. Anticholinergics had no significant effects on intrinsic membrane properties and on short-term plasticity of striatal neurons. However, they exhibited a differential ability to restore the corticostriatal plasticity deficits. A complete rescue of both long-term depression (LTD) and synaptic depotentiation (SD) was obtained by applying the M1 -preferring antagonists pirenzepine and trihexyphenidyl as well as VU0255035. Conversely, the nonselective antagonist orphenadrine produced only a partial rescue of synaptic plasticity, whereas biperiden and ethopropazine failed to restore plasticity. The selectivity for M1 receptors was further demonstrated by their ability to counteract the M1 -dependent potentiation of N-methyl-d-aspartate (NMDA) current recorded from striatal neurons. Our study demonstrates that selective M1 muscarinic receptor antagonism offsets synaptic plasticity deficits in the striatum of mice with the DYT1 dystonia mutation, providing a potential mechanistic rationale for the development of improved antimuscarinic therapies for this movement disorder.


Assuntos
Antagonistas Colinérgicos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Chaperonas Moleculares/genética , Sinapses/efeitos dos fármacos , Animais , Biofísica , Corpo Estriado/citologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Mutação/genética , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Sinapses/genética , Tálamo/citologia
11.
J Neurosci ; 34(21): 7137-47, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24849349

RESUMO

GABA-A receptors (GABA-ARs) are typically expressed at synaptic or nonsynaptic sites mediating phasic and tonic inhibition, respectively. These two forms of inhibition conjointly control various network oscillations. To disentangle their roles in thalamocortical rhythms, we focally deleted synaptic, γ2 subunit-containing GABA-ARs in the thalamus using viral intervention in mice. After successful removal of γ2 subunit clusters, spontaneous and evoked GABAergic synaptic currents disappeared in thalamocortical cells when the presynaptic, reticular thalamic (nRT) neurons fired in tonic mode. However, when nRT cells fired in burst mode, slow phasic GABA-AR-mediated events persisted, indicating a dynamic, burst-specific recruitment of nonsynaptic GABA-ARs. In vivo, removal of synaptic GABA-ARs reduced the firing of individual thalamocortical cells but did not abolish slow oscillations or sleep spindles. We conclude that nonsynaptic GABA-ARs are recruited in a phasic manner specifically during burst firing of nRT cells and provide sufficient GABA-AR activation to control major thalamocortical oscillations.


Assuntos
Córtex Cerebral/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Receptores de GABA-A/metabolismo , Tálamo/fisiologia , Animais , Dependovirus/genética , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas GABAérgicos/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Piridazinas/farmacologia , Receptores de GABA-A/genética , Sinapses/efeitos dos fármacos , Sinapses/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
J Neurosci ; 34(21): 7124-36, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24849348

RESUMO

Hyperphosphorylation and accumulation of tau aggregates are prominent features in tauopathies, including Alzheimer's disease, but the impact of loss of tau function on synaptic and cognitive deficits remains poorly understood. We report that old (19-20 months; OKO) but not middle-aged (8-9 months; MKO) tau knock-out mice develop Morris Water Maze (MWM) deficits and loss of hippocampal acetylated α-tubulin and excitatory synaptic proteins. Mild motor deficits and reduction in tyrosine hydroxylase (TH) in the substantia nigra were present by middle age, but did not affect MWM performance, whereas OKO mice showed MWM deficits paralleling hippocampal deficits. Deletion of tau, a microtubule-associated protein (MAP), resulted in increased levels of MAP1A, MAP1B, and MAP2 in MKO, followed by loss of MAP2 and MAP1B in OKO. Hippocampal synaptic deficits in OKO mice were partially corrected with dietary supplementation with docosahexaenoic acid (DHA) and both MWM and synaptic deficits were fully corrected by combining DHA with α-lipoic acid (ALA), which also prevented TH loss. DHA or DHA/ALA restored phosphorylated and total GSK3ß and attenuated hyperactivation of the tau C-Jun N-terminal kinases (JNKs) while increasing MAP1B, dephosphorylated (active) MAP2, and acetylated α-tubulin, suggesting improved microtubule stability and maintenance of active compensatory MAPs. Our results implicate the loss of MAP function in age-associated hippocampal deficits and identify a safe dietary intervention, rescuing both MAP function and TH in OKO mice. Therefore, in addition to microtubule-stabilizing therapeutic drugs, preserving or restoring compensatory MAP function may be a useful new prevention strategy.


Assuntos
Envelhecimento/patologia , Hipocampo/patologia , Aprendizagem em Labirinto/fisiologia , Sinapses/metabolismo , Proteínas tau/deficiência , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/efeitos dos fármacos , Deficiências da Aprendizagem/dietoterapia , Deficiências da Aprendizagem/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos dos Movimentos/dietoterapia , Transtornos dos Movimentos/etiologia , Desempenho Psicomotor/fisiologia , Tempo de Reação/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia , Sinapses/efeitos dos fármacos , Sinapses/genética , Ácido Tióctico/administração & dosagem
13.
J Neurosci ; 33(47): 18409-24, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24259566

RESUMO

Auditory neuropathy is a form of hearing loss in which cochlear inner hair cells fail to correctly encode or transmit acoustic information to the brain. Few genes have been implicated in the adult-onset form of this disease. Here we show that mice lacking the transcription factor Foxo3 have adult onset hearing loss with the hallmark characteristics of auditory neuropathy, namely, elevated auditory thresholds combined with normal outer hair cell function. Using histological techniques, we demonstrate that Foxo3-dependent hearing loss is not due to a loss of cochlear hair cells or spiral ganglion neurons, both of which normally express Foxo3. Moreover, Foxo3-knock-out (KO) inner hair cells do not display reductions in numbers of synapses. Instead, we find that there are subtle structural changes in and surrounding inner hair cells. Confocal microscopy in conjunction with 3D modeling and quantitative analysis show that synaptic localization is altered in Foxo3-KO mice and Myo7a immunoreactivity is reduced. TEM demonstrates apparent afferent degeneration. Strikingly, acoustic stimulation promotes Foxo3 nuclear localization in vivo, implying a connection between cochlear activity and synaptic function maintenance. Together, these findings support a new role for the canonical damage response factor Foxo3 in contributing to the maintenance of auditory synaptic transmission.


Assuntos
Cóclea/patologia , Fatores de Transcrição Forkhead/genética , Perda Auditiva Central/genética , Perda Auditiva Central/patologia , Mutação/genética , Sinapses/patologia , Estimulação Acústica , Fatores Etários , Oxirredutases do Álcool , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Correpressoras , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Células Ciliadas Auditivas Internas/ultraestrutura , Perda Auditiva Central/fisiopatologia , Imageamento Tridimensional , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Miosina VIIa , Miosinas/metabolismo , Fosfoproteínas/metabolismo , Receptores de AMPA/metabolismo , Sinapses/genética , Sinapses/ultraestrutura
14.
J Alzheimers Dis ; 37(1): 51-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23780662

RESUMO

Sporadic Creutzfeldt-Jakob disease (sCJD) is characterized by wide clinical and pathological variability, which is mainly influenced by the conformation of the misfolded prion protein (PrPSc) and by methionine and valine polymorphism at codon 129 of the gene encoding PrP. This heterogeneity likely implies differences in the molecular cascades that lead to the development of certain disease phenotypes. Here, we investigated synaptic proteome patterns in two most common sCJD subtypes (MM1 and VV2) using 2D DIGE and mass spectrometry. We found that 23 distinct proteins were differentially expressed in at least one sCJD subtype when compared to age-matched controls. The majority of these proteins displayed significant subtype-specific alterations, with only up-regulated glial fibrillary acidic protein and down-regulated spectrin alpha chain in both sCJD subtypes. Differentially expressed proteins found in this study are mainly involved in synaptic structure and activity, mitochondrial function, or calcium metabolism. Moreover, several of them have been already linked to the pathophysiological processes occurring in Alzheimer's disease.


Assuntos
Síndrome de Creutzfeldt-Jakob/metabolismo , Lobo Frontal/metabolismo , Proteoma/metabolismo , Sinapses/metabolismo , Idoso , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/patologia , Eletroforese em Gel Bidimensional/métodos , Feminino , Lobo Frontal/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteoma/genética , Sinapses/genética , Sinapses/patologia
15.
Neuron ; 78(5): 839-54, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23684785

RESUMO

Highly topographic organization of neural circuits exists for the regulation of various brain functions in corticobasal ganglia circuits. Although neural circuit-specific refinement during synapse development is essential for the execution of particular neural functions, the molecular and cellular mechanisms for synapse refinement are largely unknown. Here, we show that protocadherin 17 (PCDH17), one of the nonclustered δ2-protocadherin family members, is enriched along corticobasal ganglia synapses in a zone-specific manner during synaptogenesis and regulates presynaptic assembly in these synapses. PCDH17 deficiency in mice causes facilitated presynaptic vesicle accumulation and enhanced synaptic transmission efficacy in corticobasal ganglia circuits. Furthermore, PCDH17(-/-) mice exhibit antidepressant-like phenotypes that are known to be regulated by corticobasal ganglia circuits. Our findings demonstrate a critical role for PCDH17 in the synaptic development of specific corticobasal ganglia circuits and suggest the involvement of PCDH17 in such circuits in depressive behaviors.


Assuntos
Gânglios da Base/citologia , Caderinas/fisiologia , Córtex Cerebral/citologia , Neurônios/fisiologia , Terminações Pré-Sinápticas/fisiologia , Sinapses/genética , Estimulação Acústica , Animais , Animais Recém-Nascidos , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Transformada , Condicionamento Psicológico/fisiologia , Cricetinae , Cricetulus , Proteína 4 Homóloga a Disks-Large , Comportamento Exploratório , Medo/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Guanilato Quinases/metabolismo , Elevação dos Membros Posteriores/fisiologia , Humanos , Técnicas In Vitro , Macaca mulatta , Masculino , Aprendizagem em Labirinto/fisiologia , Potenciais da Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Rede Nervosa/fisiologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Protocaderinas , Natação/fisiologia , Sinapses/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica/genética , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
16.
Nat Neurosci ; 16(5): 596-604, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23563581

RESUMO

Stressful experience initiates a neuroendocrine response culminating in the release of glucocorticoid hormones into the blood. Glucocorticoids feed back to the brain, causing adaptations that prevent excessive hormone responses to subsequent challenges. How these changes occur remains unknown. We found that glucocorticoid receptor activation in rodent hypothalamic neuroendocrine neurons following in vivo stress is a metaplastic signal that allows GABA synapses to undergo activity-dependent long-term depression (LTDGABA). LTDGABA was unmasked through glucocorticoid receptor-dependent inhibition of Regulator of G protein Signaling 4 (RGS4), which amplified signaling through postsynaptic metabotropic glutamate receptors. This drove somatodendritic opioid release, resulting in a persistent retrograde suppression of synaptic transmission through presynaptic µ receptors. Together, our data provide new evidence for retrograde opioid signaling at synapses in neuroendocrine circuits and represent a potential mechanism underlying glucocorticoid contributions to stress adaptation.


Assuntos
Analgésicos Opioides/metabolismo , Retroalimentação Fisiológica/fisiologia , Glucocorticoides/metabolismo , Hipotálamo/citologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/genética , Channelrhodopsins , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurotransmissores/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/deficiência , Receptores de Glucocorticoides/metabolismo , Receptores Opioides mu/genética , Estresse Psicológico/sangue , Estresse Psicológico/patologia , Sinapses/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
17.
Biol Psychiatry ; 73(5): 454-63, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22883210

RESUMO

BACKGROUND: Degeneration of the locus coeruleus (LC), the major noradrenergic nucleus in the brain, occurs early and is ubiquitous in Alzheimer's disease (AD). Experimental lesions to the LC exacerbate AD-like neuropathology and cognitive deficits in several transgenic mouse models of AD. Because the LC contains multiple neuromodulators known to affect amyloid ß toxicity and cognitive function, the specific role of noradrenaline (NA) in AD is not well understood. METHODS: To determine the consequences of selective NA deficiency in an AD mouse model, we crossed dopamine ß-hydroxylase (DBH) knockout mice with amyloid precursor protein (APP)/presenilin-1 (PS1) mice overexpressing mutant APP and PS1. Dopamine ß-hydroxylase (-/-) mice are unable to synthesize NA but otherwise have normal LC neurons and co-transmitters. Spatial memory, hippocampal long-term potentiation, and synaptic protein levels were assessed. RESULTS: The modest impairments in spatial memory and hippocampal long-term potentiation displayed by young APP/PS1 or DBH (-/-) single mutant mice were augmented in DBH (-/-)/APP/PS1 double mutant mice. Deficits were associated with reduced levels of total calcium/calmodulin-dependent protein kinase II and N-methyl-D-aspartate receptor 2A and increased N-methyl-D-aspartate receptor 2B levels and were independent of amyloid ß accumulation. Spatial memory performance was partly improved by treatment with the NA precursor drug L-threo-dihydroxyphenylserine. CONCLUSIONS: These results indicate that early LC degeneration and subsequent NA deficiency in AD may contribute to cognitive deficits via altered levels of calcium/calmodulin-dependent protein kinase II and N-methyl-D-aspartate receptors and suggest that NA supplementation could be beneficial in early AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Cognição/fisiologia , Dopamina beta-Hidroxilase/metabolismo , Potenciação de Longa Duração/fisiologia , Aprendizagem em Labirinto/fisiologia , Presenilina-1/metabolismo , Sinapses/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Dopamina beta-Hidroxilase/genética , Comportamento Exploratório/fisiologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Norepinefrina/metabolismo , Presenilina-1/genética , Sinapses/genética , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Nat Neurosci ; 16(1): 13-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23222912

RESUMO

Despite the prevailing idea that neurogliaform cells produce a spatially unrestricted widespread inhibition, we demonstrate here that their activity attenuates thalamic-evoked feed-forward inhibition in layer IV barrel cortex but has no effect on feed-forward excitation. The result of this circuit selectivity is a dynamic regulation in the temporal window for integration of excitatory thalamic input, thus revealing a new role for neurogliaform cells in shaping sensory processing.


Assuntos
Neuroglia/fisiologia , Dinâmica não Linear , Córtex Somatossensorial/fisiologia , Sinapses/fisiologia , Tálamo/fisiologia , Potenciais de Ação/genética , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , GABAérgicos/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Inibição Neural/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neuropeptídeo Y/genética , Ácidos Nipecóticos/farmacologia , Parvalbuminas/metabolismo , Ácidos Fosfínicos/farmacologia , Propanolaminas/farmacologia , Receptores de Serotonina/genética , Sinapses/efeitos dos fármacos , Sinapses/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
19.
J Neurosci ; 32(34): 11706-15, 2012 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-22915113

RESUMO

Intraperitoneal injection of the Gram-negative bacterial endotoxin lipopolysaccharide (LPS) elicits a rapid innate immune response. While this systemic inflammatory response can be destructive, tolerable low doses of LPS render the brain transiently resistant to subsequent injuries. However, the mechanism by which microglia respond to LPS stimulation and participate in subsequent neuroprotection has not been documented. In this study, we first established a novel LPS treatment paradigm where mice were injected intraperitoneally with 1.0 mg/kg LPS for four consecutive days to globally activate CNS microglia. By using a reciprocal bone marrow transplantation procedure between wild-type and Toll-like receptor 4 (TLR4) mutant mice, we demonstrated that the presence of LPS receptor (TLR4) is not required on hematogenous immune cells but is required on cells that are not replaced by bone marrow transplantation, such as vascular endothelia and microglia, to transduce microglial activation and neuroprotection. Furthermore, we showed that activated microglia physically ensheathe cortical projection neurons, which have reduced axosomatic inhibitory synapses from the neuronal perikarya. In line with previous reports that inhibitory synapse reduction protects neurons from degeneration and injury, we show here that neuronal cell death and lesion volumes are significantly reduced in LPS-treated animals following experimental brain injury. Together, our results suggest that activated microglia participate in neuroprotection and that this neuroprotection is likely achieved through reduction of inhibitory axosomatic synapses. The therapeutic significance of these findings rests not only in identifying neuroprotective functions of microglia, but also in establishing the CNS location of TLR4 activation.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lipopolissacarídeos/administração & dosagem , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Receptor 4 Toll-Like/metabolismo , Animais , Antígenos CD/metabolismo , Apoptose/efeitos dos fármacos , Transplante de Medula Óssea , Lesões Encefálicas/patologia , Lesões Encefálicas/cirurgia , Bromodesoxiuridina/metabolismo , Proliferação de Células/efeitos dos fármacos , Sistema Nervoso Central/citologia , Sistema Nervoso Central/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Citometria de Fluxo , Imunidade Inata/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Microglia/metabolismo , Microglia/ultraestrutura , Microscopia Imunoeletrônica , Córtex Motor/patologia , Córtex Motor/ultraestrutura , Sinapses/genética , Sinapses/metabolismo , Sinapses/ultraestrutura , Receptor 4 Toll-Like/deficiência , Quimeras de Transplante
20.
Neuroscience ; 222: 215-27, 2012 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-22796079

RESUMO

Beside its role in development and maturation of synapses, brain-derived neurotrophic factor (BDNF) is suggested to play a critical role in modulation and plasticity of glutamatergic as well as GABAergic synaptic transmission. Here, we used heterozygous BDNF knockout (BDNF(+/-)) mice, which chronically lack approximately 50% of BDNF of wildtype (WT) animals, to investigate the role of BDNF in regulating synaptic transmission in the ventrobasal complex (VB) of the thalamus. Excitatory transmission was characterized at glutamatergic synapses onto relay (TC) neurons of the VB and intrathalamic inhibitory transmission was characterized at GABAergic synapses between neurons of the reticular thalamic nucleus (RTN) and TC neurons. Reduced expression of BDNF in BDNF(+/-) mice did not affect intrinsic membrane properties of TC neurons. Recordings in TC neurons, however, revealed a strong reduction in the frequency of miniature excitatory postsynaptic currents (mEPSCs) in BDNF(+/-) mice, as compared to WT littermates, whereas mEPSC amplitudes were not significantly different between genotypes. A mainly presynaptic impairment of corticothalamic excitatory synapses in BDNF(+/-) mice was also indicated by a decreased paired-pulse ratio and faster synaptic fatigue upon prolonged repetitive stimulation at 40 Hz. For miniature inhibitory postsynaptic currents (mIPSCs) recorded in TC neurons, both, frequency and amplitude showed a significant reduction in knock-out animals, concurrent with a prolonged decay time constant, whereas paired-pulse depression and synaptic fatigue of inhibitory synapses were not significantly different between WT and BDNF(+/-) mice. Spontaneous IPSCs (sIPSCs) recorded in VB neurons of BDNF(+/-) animals showed a significantly reduced frequency. However, the glutamatergic drive onto RTN neurons, as revealed by the percentage reduction in frequency of sIPSCs after application of AMPA and NMDA receptor blockers, was not significantly different. Together, the present findings suggest that a chronically reduced level of BDNF to ∼50% of WT levels in heterozygous knock-out animals, strongly attenuates glutamatergic and GABAergic synaptic transmission in thalamic circuits. We hypothesize that this impairment of excitatory and inhibitory transmission may have profound consequences for the generation of rhythmical activity in the thalamocortical network.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Córtex Cerebral/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Tálamo/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Interpretação Estatística de Dados , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Formação Reticular/fisiologia , Sinapses/genética , Transmissão Sináptica/genética , Ácido gama-Aminobutírico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA