Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 113: 104-123, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37393058

RESUMO

Social novelty is a cognitive process that is essential for animals to interact strategically with conspecifics based on their prior experiences. The commensal microbiome in the gut modulates social behavior through various routes, including microbe-derived metabolite signaling. Short-chain fatty acids (SCFAs), metabolites derived from bacterial fermentation in the gastrointestinal tract, have been previously shown to impact host behavior. Herein, we demonstrate that the delivery of SCFAs directly into the brain disrupts social novelty through distinct neuronal populations. We are the first to observe that infusion of SCFAs into the lateral ventricle disrupted social novelty in microbiome-depleted mice without affecting brain inflammatory responses. The deficit in social novelty can be recapitulated by activating calcium/calmodulin-dependent protein kinase II (CaMKII)-labeled neurons in the bed nucleus of the stria terminalis (BNST). Conversely, chemogenetic silencing of the CaMKII-labeled neurons and pharmacological inhibition of fatty acid oxidation in the BNST reversed the SCFAs-induced deficit in social novelty. Our findings suggest that microbial metabolites impact social novelty through a distinct neuron population in the BNST.


Assuntos
Núcleos Septais , Camundongos , Animais , Núcleos Septais/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Neurônios/metabolismo , Transdução de Sinais , Comportamento Social
2.
J Pain ; 24(11): 1915-1930, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37271352

RESUMO

The complexity and diversity of pain signaling have led to obstacles for prominent treatments due to mechanisms that are not yet fully understood. Among adenosine triphosphate (ATP) receptors, P2×7 differs in many respects from P2×1-6, it plays a significant role in various inflammatory pain, but whether it plays a role in noninflammatory pain has not been widely discussed. In this study, we utilized major neuropharmacological methods to record the effects of manipulating P2×7 during nociceptive signal transmission in the thalamocingulate circuits. Our results show that regardless of the specific cell type distribution of P2×7 in the central nervous system (CNS), it participates directly in the generated nociceptive transmission, which indicates its apparent functional existence in the major pain transmission path, the thalamocingulate circuits. Activation of P2×7 may facilitate transmission velocity along the thalamocingulate projection as well as neuron firings and synaptic vesicle release in anterior cingulate cortical neurons. Targeting thalamic P2×7 affects glutamate and ATP secretion during nociceptive signal transmission. PERSPECTIVE: The observations in this study provide evidence that the ATP receptor P2×7 presents in the central ascending pain path and plays a modulatory role during nociceptive transmission, which could contribute new insights for many antinociceptive applications.


Assuntos
Nociceptividade , Dor , Humanos , Dor/metabolismo , Neurônios/metabolismo , Glutamatos/metabolismo , Glutamatos/farmacologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Receptores Purinérgicos P2X7/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34544873

RESUMO

The biological mechanisms underpinning learning are unclear. Mounting evidence has suggested that adult hippocampal neurogenesis is involved although a causal relationship has not been well defined. Here, using high-resolution genetic mapping of adult neurogenesis, combined with sequencing information, we identify follistatin (Fst) and demonstrate its involvement in learning and adult neurogenesis. We confirmed that brain-specific Fst knockout (KO) mice exhibited decreased hippocampal neurogenesis and demonstrated that FST is critical for learning. Fst KO mice exhibit deficits in spatial learning, working memory, and long-term potentiation (LTP). In contrast, hippocampal overexpression of Fst in KO mice reversed these impairments. By utilizing RNA sequencing and chromatin immunoprecipitation, we identified Asic4 as a target gene regulated by FST and show that Asic4 plays a critical role in learning deficits caused by Fst deletion. Long-term overexpression of hippocampal Fst in C57BL/6 wild-type mice alleviates age-related decline in cognition, neurogenesis, and LTP. Collectively, our study reveals the functions for FST in adult neurogenesis and learning behaviors.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Folistatina/fisiologia , Hipocampo/metabolismo , Neurogênese , Plasticidade Neuronal , Aprendizagem Espacial/fisiologia , Canais Iônicos Sensíveis a Ácido/genética , Animais , Cognição , Feminino , Potenciação de Longa Duração , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sinapses/fisiologia
4.
Acta Neuropathol Commun ; 9(1): 112, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158119

RESUMO

Tau pathology is instrumental in the gradual loss of neuronal functions and cognitive decline in tauopathies, including Alzheimer's disease (AD). Earlier reports showed that adenosine metabolism is abnormal in the brain of AD patients while consequences remained ill-defined. Herein, we aimed at investigating whether manipulation of adenosine tone would impact Tau pathology, associated molecular alterations and subsequent neurodegeneration. We demonstrated that treatment with an inhibitor (J4) of equilibrative nucleoside transporter 1 (ENT1) exerted beneficial effects in a mouse model of Tauopathy. Treatment with J4 not only reduced Tau hyperphosphorylation but also rescued memory deficits, mitochondrial dysfunction, synaptic loss, and abnormal expression of immune-related gene signatures. These beneficial effects were particularly ascribed to the ability of J4 to suppress the overactivation of AMPK (an energy reduction sensor), suggesting that normalization of energy dysfunction mitigates neuronal dysfunctions in Tauopathy. Collectively, these data highlight that targeting adenosine metabolism is a novel strategy for tauopathies.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Transportador Equilibrativo 1 de Nucleosídeo/antagonistas & inibidores , Tauopatias/metabolismo , Tauopatias/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos
5.
Transl Psychiatry ; 11(1): 162, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723225

RESUMO

Nerve growth factor (NGF) gene therapy has been used in clinical trials of Alzheimer's disease. Understanding the underlying mechanisms of how NGF influences memory may help develop new strategies for treatment. Both NGF and the cholinergic system play important roles in learning and memory. NGF is essential for maintaining cholinergic innervation of the hippocampus, but it is unclear whether the supportive effect of NGF on learning and memory is specifically dependent upon intact hippocampal cholinergic innervation. Here we characterize the behavior and hippocampal measurements of volume, neurogenesis, long-term potentiation, and cholinergic innervation, in brain-specific Ngf-deficient mice. Our results show that knockout mice exhibit increased anxiety, impaired spatial learning and memory, decreased adult hippocampal volume, neurogenesis, short-term potentiation, and cholinergic innervation. Overexpression of Ngf in the hippocampus of Ngf gene knockout mice rescued spatial memory and partially restored cholinergic innervations, but not anxiety. Selective depletion of hippocampal cholinergic innervation resulted in impaired spatial memory. However, Ngf overexpression in the hippocampus failed to rescue spatial memory in mice with hippocampal-selective cholinergic fiber depletion. In conclusion, we demonstrate the impact of Ngf deficiency in the brain and provide evidence that the effect of NGF on spatial memory is reliant on intact cholinergic innervations in the hippocampus. These results suggest that adequate cholinergic targeting may be a critical requirement for successful use of NGF gene therapy of Alzheimer's disease.


Assuntos
Fator de Crescimento Neural , Memória Espacial , Animais , Colinérgicos , Hipocampo , Potenciação de Longa Duração , Camundongos
6.
Front Cell Neurosci ; 13: 86, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30930747

RESUMO

Stereotypic and/or repetitive behavior is one of the major symptoms of autism spectrum disorder (ASD). Increase of self-grooming behavior is a behavioral phenotype commonly observed in the mouse models for ASD. Previously, we have shown that knockout of acid-sensing ion channel 3 (ASIC3) led to the increased self-grooming behavior in resident-intruder test. Given the facts that ASIC3 is mainly expressed in the peripheral dorsal root ganglion (DRG) and conditional knockout of ASIC3 in the proprioceptors induced proprioception deficits. We speculate a hypothesis that stereotypic phenotype related to ASD, pararalled with striatal dysfunction, might be caused by proprioception defect in the peripheral sensory neuron origin. Herein, we investigate in depth whether and how ASIC3 is involved in the regulation of self-grooming behavior. First, we observed that Asic3 null mutant mice exhibited increased self-grooming in social interaction during juvenile stage. Similarly, they displayed increased self-grooming behavior in a novel cage in the absence of cagemate. To further understand the mechanism by which ASIC3 affects grooming behavior, we analyzed neurochemical, neuropathological and electrophysiological features in the dorsal striatum of Asic3 null mutant mice. Knockout of Asic3 increased dopamine (DA) activity and phospho-ERK immunoreactivities in the dorsal striatum. Furthermore, we detected a lower paired-pulse ratio (PPR) and impaired long-term potentiation (LTP) in corticostriatal circuits in Asic3 null mutant mice as compared with wild-type (WT) littermates. Moreover, knockout of Asic3 altered the medial spiny neurons in the striatum with defects in presynaptic function and decrease of dendritic spines. Lastly, genetic ablation of Asic3 specifically in parvalbumin-positive (PV+) cells resulted in the increase of self-grooming behavior in mice. These findings suggest knockout of Asic3 in the PV+ neurons alters grooming behavior by co-opting corticostriatal circuits.

7.
EBioMedicine ; 40: 675-684, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30639418

RESUMO

BACKGROUND: Mutations in PKD1 or PKD2 gene lead to autosomal dominant polycystic kidney disease (ADPKD). The mechanism of ADPKD progression and its link to increased cardiovascular mortality is still elusive. METHODS: We differentiated ADPKD patient induced pluripotent stem cells (iPSCs) to cardiomyocytes (CMs). The electrophysiological properties at the cellular level were analyzed by calcium imaging and whole cell patch clamping. FINDINGS: The ADPKD patient iPSC-CMs had decreased sarcoplasmic reticulum calcium content compared with Control-CMs. Spontaneous action potential of the PKD2 mutation line-derived CMs demonstrated slower beating rate and longer action potential duration. The PKD1 mutation line-derived CMs showed a comparable dose-dependent shortening of phase II repolarization with the Control-CMs, but a significant increase in beating frequency in response to L-type calcium channel blocker. The PKD1-mutant iPSC-CMs also showed a relatively unstable baseline as a greater percentage of cells exhibited delayed afterdepolarizations (DADs). Both the ADPKD patient iPSC-CMs showed more ß-adrenergic agonist-elicited DADs compared with Control-CMs. INTERPRETATION: Characterization of ADPKD patient iPSC-CMs provides new insights into the increased clinical risk of arrhythmias, and the results enable disease modeling and drug screening for cardiac manifestations of ADPKD. FUND: Ministry of Science and Technology, National Health Research Institutes, Academia Sinica Program for Technology Supporting Platform Axis Scheme, Thematic Research Program and Summit Research Program, and Kaohsiung Medical University Hospital, Taiwan.


Assuntos
Cardiopatias/etiologia , Cardiopatias/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Rim Policístico Autossômico Dominante/complicações , Agonistas Adrenérgicos beta/farmacologia , Biomarcadores , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Diferenciação Celular , Linhagem Celular , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Expressão Gênica , Cardiopatias/diagnóstico , Humanos , Imagem Molecular , Mutação , Fenótipo , Rim Policístico Autossômico Dominante/genética , Retículo Sarcoplasmático/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
8.
Mol Neurobiol ; 55(12): 8936-8952, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29616397

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and synaptic dysfunction. Adenosine is an important homeostatic modulator that controls the bioenergetic network in the brain through regulating receptor-evoked signaling pathways, bioenergetic machineries, and epigenetic-mediated gene regulation. Equilibrative nucleoside transporter 1 (ENT1) is a major adenosine transporter that recycles adenosine from the extracellular space. In the present study, we report that a small adenosine analogue (designated J4) that inhibited ENT1 prevented the decline in spatial memory in an AD mouse model (APP/PS1). Electrophysiological and biochemical analyses further demonstrated that chronic treatment with J4 normalized the impaired basal synaptic transmission and long-term potentiation (LTP) at Schaffer collateral synapses as well as the aberrant expression of synaptic proteins (e.g., NR2A and NR2B), abnormal neuronal plasticity-related signaling pathways (e.g., PKA and GSK3ß), and detrimental elevation in astrocytic A2AR expression in the hippocampus and cortex of APP/PS1 mice. In conclusion, our findings suggest that modulation of adenosine homeostasis by J4 is beneficial in a mouse model of AD. Our study provides a potential therapeutic strategy to delay the progression of AD.


Assuntos
Adenosina/uso terapêutico , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Transportador Equilibrativo 1 de Nucleosídeo/antagonistas & inibidores , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/fisiopatologia , Plasticidade Neuronal , Presenilina-1/metabolismo , Adenosina/farmacologia , Doença de Alzheimer/patologia , Animais , Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/prevenção & controle , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Camundongos Transgênicos , Plasticidade Neuronal/efeitos dos fármacos , Placa Amiloide/patologia , Placa Amiloide/fisiopatologia , Receptor A2A de Adenosina/metabolismo , Transmissão Sináptica/efeitos dos fármacos
9.
Pain ; 159(5): 907-918, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29672451

RESUMO

Chronic pain can be initiated by one or more acute stimulations to sensitize neurons into the primed state. In the primed state, the basal nociceptive thresholds of the animal are normal, but, in response to another hyperalgesic stimulus, the animal develops enhanced and prolonged hyperalgesia. The exact mechanism of how primed state is formed is not completely understood. Here, we showed that spinal protein kinase C (PKC)/extracellular signal-regulated kinase (ERK) signal pathway is required for neuronal plasticity change, hyperalgesic priming formation, and the development of chronic hyperalgesia using acid-induced muscle pain model in mice. We discovered that phosphorylated extracellular signal-regulated kinase-positive neurons in the amygdala, spinal cord, and dorsal root ganglion were significantly increased after first acid injection. Inhibition of the phosphorylated extracellular signal-regulated kinase activity intrathecally, but not intracerebroventricularly or intramuscularly before first acid injection, prevented the development of chronic pain induced by second acid injection, which suggests that hyperalgesic priming signal is stored at spinal cord level. Furthermore, intrathecal injection of PKC but not protein kinase A blocker prevented the development of chronic pain, and PKC agonist was sufficient to induce prolonged hyperalgesia response after acid injection. We also found that mammalian target of rapamycin-dependent protein synthesis was required for the priming establishment. To test whether hyperalgesic priming leads to synaptic plasticity change, we recorded field excitatory postsynaptic potentials from spinal cord slices and found enhanced long-term potentiation in mice that received one acid injection. This long-term potentiation enhancement was prevented by inhibition of extracellular signal-regulated kinase. These findings show that the activation of PKC/ERK signal pathway and downstream protein synthesis is required for hyperalgesic priming and the consolidation of pain singling.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hiperalgesia/metabolismo , Mialgia/fisiopatologia , Proteínas Quinases/metabolismo , Ácidos/toxicidade , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mialgia/induzido quimicamente , Mialgia/patologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Medição da Dor , Sirolimo/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Estilbamidinas/metabolismo
10.
Front Mol Neurosci ; 10: 217, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28744198

RESUMO

Galectin-3, a member of the galectin protein family, has been found to regulate cell proliferation, inhibit apoptosis and promote inflammatory responses. Galectin-3 is also expressed in the adult rat hippocampus, but its role in learning and memory function is not known. Here, we found that contextual fear-conditioning training, spatial training or injection of NMDA into the rat CA1 area each dramatically decreased the level of endogenous galectin-3 expression. Overexpression of galectin-3 impaired fear memory, whereas galectin-3 knockout (KO) enhanced fear retention, spatial memory and hippocampal long-term potentiation. Galectin-3 was further found to associate with integrin α3, an association that was decreased after fear-conditioning training. Transfection of the rat CA1 area with small interfering RNA against galectin-3 facilitated fear memory and increased phosphorylated focal adhesion kinase (FAK) levels, effects that were blocked by co-transfection of the FAK phosphorylation-defective mutant Flag-FAKY397F. Notably, levels of serine-phosphorylated galectin-3 were decreased by fear conditioning training. In addition, blockade of galectin-3 phosphorylation at Ser-6 facilitated fear memory, whereas constitutive activation of galectin-3 at Ser-6 impaired fear memory. Interestingly galectin-1 plays a role in fear-memory formation similar to that of galectin-3. Collectively, our data provide the first demonstration that galectin-3 is a novel negative regulator of memory formation that exerts its effects through both extracellular and intracellular mechanisms.

11.
Hum Mol Genet ; 26(12): 2247-2257, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28369378

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by an expansion of CTG repeats in the 3' untranslated region (UTR) of the dystrophia myotonia protein kinase (DMPK) gene. Cognitive impairment associated with structural change in the brain is prevalent in DM1. How this histopathological abnormality during disease progression develops remains elusive. Nuclear accumulation of mutant DMPK mRNA containing expanded CUG RNA disrupting the cytoplasmic and nuclear activities of muscleblind-like (MBNL) protein has been implicated in DM1 neural pathogenesis. The association between MBNL dysfunction and morphological changes has not been investigated. We generated a mouse model for postnatal expression of expanded CUG RNA in the brain that recapitulates the features of the DM1 brain, including the formation of nuclear RNA and MBNL foci, learning disability, brain atrophy and misregulated alternative splicing. Characterization of the pathological abnormalities by a time-course study revealed that hippocampus-related learning and synaptic potentiation were impaired before structural changes in the brain, followed by brain atrophy associated with progressive reduction of axon and dendrite integrity. Moreover, cytoplasmic MBNL1 distribution on dendrites decreased before dendrite degeneration, whereas reduced MBNL2 expression and altered MBNL-regulated alternative splicing was evident after degeneration. These results suggest that the expression of expanded CUG RNA in the DM1 brain results in neurodegenerative processes, with reduced cytoplasmic MBNL1 as an early event response to expanded CUG RNA.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas , Processamento Alternativo , Animais , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Éxons , Humanos , Camundongos , Distrofia Miotônica/genética , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , RNA Mensageiro/metabolismo , Expansão das Repetições de Trinucleotídeos
12.
Cell Death Differ ; 24(4): 597-614, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28186506

RESUMO

Amyloid-ß (Aß) produces neurotoxicity in the brain and causes neuronal death, but the endogenous defense mechanism that is activated on Aß insult is less well known. Here we found that acute Aß increases the expression of PIAS1 and Mcl-1 via activation of MAPK/ERK, and Aß induction of PIAS1 enhances HDAC1 SUMOylation in rat hippocampus. Knockdown of PIAS1 decreases endogenous HDAC1 SUMOylation and blocks Aß induction of Mcl-1. Sumoylated HDAC1 reduces it association with CREB, increases CREB binding to the Mcl-1 promoter and mediates Aß induction of Mcl-1 expression. Transduction of SUMO-modified lenti-HDAC1 vector to the hippocampus of APP/PS1 mice rescues spatial learning and memory deficit and long-term potentiation impairment in APP/PS1 mice. It also reduces the amount of amyloid plaque and the number of apoptotic cells in CA1 area of APP/PS1 mice. Meanwhile, HDAC1 SUMOylation decreases HDAC1 binding to the neprilysin promoter. These results together reveal an important role of HDAC1 SUMOylation as a naturally occurring defense mechanism protecting against Aß toxicity and provide an alternative therapeutic strategy against AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Epigênese Genética , Histona Desacetilase 1/metabolismo , Neuroproteção/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Hipocampo/metabolismo , Histona Desacetilase 1/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Nitrilas/farmacologia , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/antagonistas & inibidores , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Sumoilação
13.
Nat Neurosci ; 19(11): 1513-1522, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27595386

RESUMO

Cortico-basal ganglia circuits are critical for speech and language and are implicated in autism spectrum disorder, in which language function can be severely affected. We demonstrate that in the mouse striatum, the gene Foxp2 negatively interacts with the synapse suppressor gene Mef2c. We present causal evidence that Mef2c inhibition by Foxp2 in neonatal mouse striatum controls synaptogenesis of corticostriatal inputs and vocalization in neonates. Mef2c suppresses corticostriatal synapse formation and striatal spinogenesis, but can itself be repressed by Foxp2 through direct DNA binding. Foxp2 deletion de-represses Mef2c, and both intrastriatal and global decrease of Mef2c rescue vocalization and striatal spinogenesis defects of Foxp2-deletion mutants. These findings suggest that Foxp2-Mef2C signaling is critical to corticostriatal circuit formation. If found in humans, such signaling defects could contribute to a range of neurologic and neuropsychiatric disorders.


Assuntos
Transtorno do Espectro Autista/genética , Fatores de Transcrição Forkhead/metabolismo , Vias Neurais/metabolismo , Proteínas Repressoras/metabolismo , Vocalização Animal/fisiologia , Animais , Gânglios da Base/metabolismo , Comunicação , Corpo Estriado/metabolismo , Aprendizagem/fisiologia , Fatores de Transcrição MEF2/genética , Camundongos Transgênicos
14.
PLoS One ; 10(12): e0144806, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26659605

RESUMO

Peroxisomal proliferator-activated receptor gamma (PPARγ) is a nuclear hormone receptor whose agonist, rosiglitazone has a neuroprotective effect to hippocampal neurons in pilocarpine-induced seizures. Hippocampal slice preparations treated in Mg2+ free medium can induce ictal and interictal-like epileptiform discharges, which is regarded as an in vitro model of N-methyl-D-aspartate (NMDA) receptor-mediated temporal lobe epilepsy (TLE). We applied rosiglitazone in hippocampal slices treated in Mg2+ free medium. The effects of rosiglitazone on hippocampal CA1-Schaffer collateral synaptic transmission were tested. We also examined the neuroprotective effect of rosiglitazone toward NMDA excitotoxicity on cultured hippocampal slices. Application of 10 µM rosiglitazone significantly suppressed amplitude and frequency of epileptiform discharges in CA1 neurons. Pretreatment with the PPARγ antagonist GW9662 did not block the effect of rosiglitazone on suppressing discharge frequency, but reverse the effect on suppressing discharge amplitude. Application of rosiglitazone suppressed synaptic transmission in the CA1-Schaffer collateral pathway. By miniature excitatory-potential synaptic current (mEPSC) analysis, rosiglitazone significantly suppressed presynaptic neurotransmitter release. This phenomenon can be reversed by pretreating PPARγ antagonist GW9662. Also, rosiglitazone protected cultured hippocampal slices from NMDA-induced excitotoxicity. The protective effect of 10 µM rosiglitazone was partially antagonized by concomitant high dose GW9662 treatment, indicating that this effect is partially mediated by PPARγ receptors. In conclusion, rosiglitazone suppressed NMDA receptor-mediated epileptiform discharges by inhibition of presynaptic neurotransmitter release. Rosiglitazone protected hippocampal slice from NMDA excitotoxicity partially by PPARγ activation. We suggest that rosiglitazone could be a potential agent to treat patients with TLE.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Tiazolidinedionas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Anilidas/farmacologia , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Meios de Cultura/química , Meios de Cultura/farmacologia , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/patologia , Regulação da Expressão Gênica , Magnésio/farmacologia , Microtomia , Modelos Biológicos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/antagonistas & inibidores , PPAR gama/antagonistas & inibidores , PPAR gama/genética , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Rosiglitazona , Convulsões/tratamento farmacológico , Convulsões/genética , Convulsões/metabolismo , Convulsões/patologia , Transmissão Sináptica/efeitos dos fármacos , Tiazolidinedionas/antagonistas & inibidores , Técnicas de Cultura de Tecidos
15.
Front Cell Neurosci ; 8: 87, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24744698

RESUMO

Innate immune responses have been shown to influence brain development and function. Dysregulation of innate immunity is significantly associated with psychiatric disorders such as autism spectrum disorders and schizophrenia, which are well-known neurodevelopmental disorders. Recent studies have revealed that critical players of the innate immune response are expressed in neuronal tissues and regulate neuronal function and activity. For example, Sarm1, a negative regulator that acts downstream of Toll-like receptor (TLR) 3 and 4, is predominantly expressed in neurons. We have previously shown that Sarm1 regulates neuronal morphogenesis and the expression of inflammatory cytokines in the brain, which then affects learning ability, cognitive flexibility, and social interaction. Because impaired neuronal morphogenesis and dysregulation of cytokine expression may disrupt neuronal activity, we investigated whether Sarm1 knockdown affects the synaptic responses of neurons. We here show that reduced Sarm1 expression impairs metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD) formation but enhances N-methyl-D-aspartate receptor (NMDAR)-dependent long-term potentiation production in hippocampal CA1 neurons. The expression levels of post-synaptic proteins, including NR2a, NR1, Shank1 and Shank3, are also altered in Sarm1 knockdown mice, suggesting a role for Sarm1 in the maintenance of synaptic homeostasis. The addition of a positive allosteric modulator of mGluR5, CDPPB, ameliorates the LTD defects in slice recording and the behavioral deficits in social interaction and associative memory. These results suggest an important role for mGluR5 signaling in the function of Sarm1. In conclusion, our study demonstrates a role for Sarm1 in the regulation of synaptic plasticity. Through these mechanisms, Sarm1 knockdown results in the impairment of associative memory and social interactions in mice.

16.
PLoS One ; 8(12): e83394, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386192

RESUMO

Hippocampal neurons play a critical role in learning and memory; however, the effects of environmental mechanical forces on neurite extension and associated underlying mechanisms are largely unexplored, possibly due to difficulties in maintaining central nervous system neurons. Neuron adhesion, neurite length, and mechanotransduction are mainly influenced by the extracellular matrix (ECM), which is often associated with structural scaffolding. In this study, we investigated the relationship between substrate stiffness and hippocampal neurite outgrowth by controlling the ratios of polydimethylsiloxane (PDMS) base to curing agent to create substrates of varying stiffness. Immunostaining results demonstrated that hippocampal neurons have longer neurite elongation in 35:1 PDMS substrate compared those grown on 15:1 PDMS, indicating that soft substrates provide a more optimal stiffness for hippocampal neurons. Additionally, we discovered that pPKCα expression was higher in the 15:1 and 35:1 PDMS groups than in the poly-L-lysine-coated glass group. However, when we used a fibronectin (FN) coating, we found that pFAKy397 and pFAKy925 expression were higher in glass group than in the 15:1 or 35: 1 PDMS groups, but pPKCα and pERK1/2 expression were higher in the 35:1 PDMS group than in the glass group. These results support the hypothesis that environmental stiffness influences hippocampal neurite outgrowth and underlying signaling pathways.


Assuntos
Hipocampo/citologia , Hipocampo/metabolismo , Neuritos/metabolismo , Células Piramidais/crescimento & desenvolvimento , Células Piramidais/metabolismo , Animais , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Cultura Primária de Células , Proteína Quinase C-alfa/metabolismo
17.
Acta Neuropathol ; 124(2): 231-45, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22760527

RESUMO

Impairment of learning and memory is a significant pathological feature of many neurodegenerative diseases including FTLD-TDP. Appropriate regulation and fine tuning of spinogenesis of the dendrites, which is an integral part of the learning/memory program of the mammalian brain, are essential for the normal function of the hippocampal neurons. TDP-43 is a nucleic acid-binding protein implicated in multi-cellular functions and in the pathogenesis of a range of neurodegenerative diseases including FTLD-TDP and ALS. We have combined the use of single-cell dye injection, shRNA knockdown, plasmid rescue, immunofluorescence staining, Western blot analysis and patch clamp electrophysiological measurement of primary mouse hippocampal neurons in culture to study the functional role of TDP-43 in mammalian spinogenesis. We found that depletion of TDP-43 leads to an increase in the number of protrusions/spines as well as the percentage of matured spines among the protrusions. Significantly, the knockdown of TDP-43 also increases the level of Rac1 and its activated form GTP-Rac1, a known positive regulator of spinogenesis. Clustering of the AMPA receptors on the dendritic surface and neuronal firing are also induced by depletion of TDP-43. Furthermore, use of an inhibitor of Rac1 activation negatively regulated spinogenesis of control hippocampal neurons as well as TDP-43-depleted hippocampal neurons. Mechanistically, RT-PCR assay and cycloheximide chase experiments have indicated that increases in Rac1 protein upon TDP-43 depletion is regulated at the translational level. These data together establish that TDP-43 is an upstream regulator of spinogenesis in part through its action on the Rac1 â†’ GTP-Rac1 â†’ AMPAR pathway. This study provides the first evidence connecting TDP-43 with the GTP-Rac1 â†’ AMPAR regulatory pathway of spinogenesis. It establishes that mis-metabolism of TDP-43, as occurs in neurodegenerative diseases with TDP-43 proteinopathies, e.g., FTLD-TDP, would alter its homeostatic cellular concentration, thus leading to impairment of hippocampal plasticity.


Assuntos
Proteínas de Ligação a DNA/genética , Espinhas Dendríticas/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Biossíntese de Proteínas/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Espinhas Dendríticas/metabolismo , Hipocampo/citologia , Camundongos , Neurônios/citologia , RNA Interferente Pequeno , Transdução de Sinais/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
Commun Integr Biol ; 4(4): 394-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21966555

RESUMO

Chronic pain is characterized by post-injury pain hypersensitivity. Current evidence suggests that it might result from altered neuronal excitability and/or synaptic functions in pain-related pathways and brain areas, an effect known as central sensitization. Increased activity of extracellular signal-regulated kinase (ERK) has been well-demonstrated in the dorsal horn of the spinal cord in chronic pain animal models. Recently, increased ERK activity has also been identified in two supraspinal areas, the central amygdala and the paraventricular thalamic nucleus anterior. Our recent work on the capsular central amygdala has shown that this increased ERK activity can enhance synaptic transmission, which might account for central sensitization and behavior hypersensitivity in animals receiving noxious stimuli.

19.
J Neurophysiol ; 105(6): 2715-28, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21430282

RESUMO

Noradrenergic (NAergic) A7 neurons that project axonal terminals to the dorsal horn of the spinal cord to modulate nociceptive signaling are suggested to receive tonic inhibition from local GABAergic interneurons, which are under the regulation of descending analgesic pathways. In support of this argument, we presently report GABA(B) receptor (GABA(B)R)-mediated tonic inhibition of NAergic A7 neurons. Bath application of baclofen induced an outward current (I(Bac)) in NAergic A7 neurons that was blocked by CGP 54626, a GABA(B)R blocker. The I(Bac) was reversed at about -99 mV, displayed inward rectification, and was blocked by Ba(2+) or Tertipian-Q, showing it was mediated by G protein-activated inward-rectifying K(+) (GIRK) channels. Single-cell RT-PCR results suggested that GIRK1/3 heterotetramers might dominate functional GIRK channels in NAergic A7 neurons. Under conditions in which GABA(A) and glycine receptors were blocked, bath application of GABA inhibited the spontaneous firing of NAergic A7 neurons in a dose-dependent manner. Interestingly, CGP 54626 application not only blocked the effect of GABA but also increased the firing rate to 126.9% of the control level, showing that GABA(B)Rs were constitutively active at an ambient GABA concentration of 2.8 µM and inhibited NAergic A7 neurons. GABA(B)Rs were also found at presynaptic excitatory and inhibitory axonal terminals in the A7 area. Pharmacological activation of these GABA(B)Rs inhibited the release of neurotransmitters. No physiological role was found for GABA(B)Rs on excitatory terminals, whereas those on the inhibitory terminals were found to exert autoregulatory control of GABA release.


Assuntos
Inibição Neural/fisiologia , Neurônios/fisiologia , Norepinefrina/metabolismo , Ponte/citologia , Receptores de GABA-B/metabolismo , Análise de Variância , Animais , Anisóis/farmacologia , Baclofeno/farmacologia , Bário/farmacologia , Venenos de Abelha/farmacologia , Dopamina beta-Hidroxilase/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Antagonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Morfolinas/farmacologia , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ácidos Nipecóticos/farmacologia , Compostos Organofosforados/farmacologia , Oximas/farmacologia , Técnicas de Patch-Clamp/métodos , Bloqueadores dos Canais de Potássio/farmacologia , Cloreto de Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/farmacologia
20.
J Neurosci ; 31(6): 2258-70, 2011 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21307262

RESUMO

Application of phorbol 12,13-diacetate (PDA) caused marked enhancement of synaptic transmission of nociceptive parabrachio-amygdaloid (PBA) input onto neurons of the capsular central amygdaloid (CeAC) nucleus. The potentiation of PBA-CeAC EPSCs by PDA involved a presynaptic protein kinase C (PKC)-dependent component and a postsynaptic PKC-extracellular-regulated kinase (ERK)-dependent component. NMDA glutamatergic receptor (NMDAR)-dependent long-term potentiation (LTP) of PBA-CeAC EPSCs, which was also dependent on the PKC-ERK signaling pathway, was induced by tetanus stimulation at 100 Hz. In slices from mice subjected to acid-induced muscle pain (AIMP), phosphorylated ERK levels in the CeAC increased, and PBA-CeAC synaptic transmission was postsynaptically enhanced. The enhanced PBA-CeAC synaptic transmission in AIMP mice shared common mechanisms with the postsynaptic potentiation effect of PDA and induction of NMDAR-dependent LTP by high-frequency stimulation in normal slices, both of which required ERK activation. Since the CeAC plays an important role in the emotionality of pain, enhanced synaptic function of nociceptive (PBA) inputs onto CeAC neurons might partially account for the supraspinal mechanisms underlying central sensitization.


Assuntos
Tonsila do Cerebelo/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Potenciação de Longa Duração/fisiologia , Dor/patologia , Células Receptoras Sensoriais/fisiologia , Transmissão Sináptica/fisiologia , Ácidos/efeitos adversos , Vias Aferentes/fisiologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/fisiopatologia , Dor/induzido quimicamente , Medição da Dor , Técnicas de Patch-Clamp/métodos , Ésteres de Forbol/farmacologia , Fosforilação , Células Receptoras Sensoriais/citologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Fatores de Tempo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...