Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Neurosci ; 26(12): 2081-2089, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37996529

RESUMO

It is generally thought that under basal conditions, neurons produce ATP mainly through mitochondrial oxidative phosphorylation (OXPHOS), and glycolytic activity only predominates when neurons are activated and need to meet higher energy demands. However, it remains unknown whether there are differences in glucose metabolism between neuronal somata and axon terminals. Here, we demonstrated that neuronal somata perform higher levels of aerobic glycolysis and lower levels of OXPHOS than terminals, both during basal and activated states. We found that the glycolytic enzyme pyruvate kinase 2 (PKM2) is localized predominantly in the somata rather than in the terminals. Deletion of Pkm2 in mice results in a switch from aerobic glycolysis to OXPHOS in neuronal somata, leading to oxidative damage and progressive loss of dopaminergic neurons. Our findings update the conventional view that neurons uniformly use OXPHOS under basal conditions and highlight the important role of somatic aerobic glycolysis in maintaining antioxidant capacity.


Assuntos
Glicólise , Fosforilação Oxidativa , Animais , Camundongos , Fosfotransferases/metabolismo , Estresse Oxidativo , Glucose/metabolismo
3.
Cell Rep ; 41(4): 111532, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36288697

RESUMO

The function and regulation of different heterogeneous reactive states of astrocytes in depression remain unclear. Here, we demonstrate that neurotoxic reactive (A1-like) astrocytes are strongly induced, prior to behavioral impairments and dendritic atrophy, in depression-like mice. More interestingly, global or microglia-specific knockout of Nod-like receptor protein 3 (Nlrp3) markedly mitigates A1-like astrocyte induction, whereas astrocyte-specific Nlrp3 depletion is ineffective. Microglial Nlrp3 ablation also alleviates the neuronal dysfunction induced by A1-like astrocytes both in vitro and in vivo. We further show that in microglia the NF-κB pathway activates the NLRP3 inflammasome which in turn activates caspase-1 to induce the secretion of A1 inductors, leading to the production of A1-like astrocytes. Altogether, this study reveals the function of microglial NLRP3 inflammasome in the induction of neurotoxic astrocytes via activating neuroinflammatory caspase-1 pathway in response to chronic stress and suggests a potential therapeutic strategy for depression.


Assuntos
Inflamassomos , Síndromes Neurotóxicas , Animais , Camundongos , Inflamassomos/metabolismo , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Astrócitos/metabolismo , NF-kappa B/metabolismo , Depressão , Caspase 1/metabolismo
4.
J Neuroinflammation ; 19(1): 23, 2022 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-35093099

RESUMO

BACKGROUND: Fluoxetine, a selective serotonin reuptake inhibitor, has been reported to directly bind with 5-HT2B receptor (5-HT2BR), but the precise mechanisms, whereby fluoxetine confers the anti-depressive actions via 5-HT2BR is not fully understood. Although neuroinflammation-induced A1 astrocytes are involved in neurodegenerative diseases, the role of A1 astrocyte in the pathogenesis and treatment of major depressive disorder (MDD) remains unclear. METHODS: Mice were subjected to chronic mild stress (CMS) for 6 weeks and subsequently treated with fluoxetine for 4 weeks. The depressive-like and anxiety-like behaviors and the activation of A1 reactive astrocyte in hippocampus and cortex of mice were measured. Primary astrocytes were stimulated with A1 cocktail (tumor necrosis factor (TNF)-α, interleukin (IL)-1α and C1q), activated (LPS) microglia-conditioned medium (MCM) or IL-6 for 24 h and the expression of A1-special and A2-special markers were determined using RT-qPCR and western blot. The role of 5-HT2BR in the effects of fluoxetine on A1 reactive astrocyte was measured using 5-HT2BR inhibitor and siRNA in vitro and AAVs in vivo. The functions of downstream signaling Gq protein and ß-arrestins in the effects of fluoxetine on the activation of A1 astrocyte were determined using pharmacological inhibitor and genetic knockout, respectively. RESULTS: In this study, we found that fluoxetine inhibited the activation of A1 reactive astrocyte and reduced the abnormal behaviors in CMS mice, as well as ameliorated A1 astrocyte reactivity under three different stimulators in primary astrocytes. We also showed that astrocytic 5-HT2BR was required in the inhibitory effects of fluoxetine on A1 reactive astrocyte in MDD in vivo and in vitro. We further found that the functions of fluoxetine in the activation of A1 astrocyte were independent of either Gq protein or ß-arrestin1 in vitro. ß-arrestin2 pathway was the downstream signaling of astrocytic 5-HT2BR mediated the inhibitory effects of fluoxetine on A1 astrocyte reactivity in primary astrocytes and CMS mice, as well as the improved roles of fluoxetine in behavioral impairments of CMS mice. CONCLUSIONS: These data demonstrate that fluoxetine restricts reactive A1 astrocyte via astrocytic 5-HT2BR/ß-arrestin2 pathway in a mouse model of MDD and provide a novel therapeutic avenue for MDD.


Assuntos
Transtorno Depressivo Maior , Fluoxetina , Animais , Astrócitos/metabolismo , Transtorno Depressivo Maior/metabolismo , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Camundongos , Serotonina/metabolismo , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
5.
JCI Insight ; 6(23)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34877938

RESUMO

Emerging evidence suggests that astrocyte loss is one of the most important pathological features in the hippocampus of patients with major depressive disorder (MDD) and depressive mice. Pyroptosis is a recently discovered form of programmed cell death depending on Caspase-gasdermin D (Casp-GSDMD), which is involved in multiple neuropsychiatric diseases. However, the involvement of pyroptosis in the onset of MDD and glial pathological injury remains obscure. Here, we observed that depressive mice showed astrocytic pyroptosis, which was responsible for astrocyte loss, and selective serotonin reuptake inhibitor (SSRI) treatment could attenuate the pyroptosis induced by the chronic mild stress (CMS) model. Genetic KO of GSDMD, Casp-1, and astrocytic NOD-like receptor protein 3 (NLRP3) inflammasome in mice alleviated depression-like behaviors and inhibited the pyroptosis-associated protein expression. In contrast, overexpression of astrocytic GSDMD-N-terminal domain (GSDMD-N) in the hippocampus could abolish the improvement of behavioral alterations in GSDMD-deficient mice. This work illustrates that targeting the NLRP3/Casp-1/GSDMD-mediated pyroptosis may provide potential therapeutic benefits to stress-related astrocyte loss in the pathogenesis of depression.


Assuntos
Astrócitos/metabolismo , Caspase 1/metabolismo , Depressão/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
6.
Redox Biol ; 47: 102168, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34673451

RESUMO

Kir6.2, a pore-forming subunit of the ATP-sensitive potassium (KATP) channels, regulates the functions of metabolically active tissues and acts as an ideal therapeutic target for multiple diseases. Previous studies have been conducted on peripheral kir6.2, but its precise physiological roles in the central nervous system (CNS) have rarely been revealed. In the current study, we evaluated the neurophenotypes and neuroethology of kir6.2 knockout (kir6.2-/-) mice. We demonstrated the beneficial effects of kir6.2 on maintaining the morphology of mesencephalic neurons and controlling the motor coordination of mice. The mechanisms underlying the abnormal neurological features of kir6.2 deficiency were analyzed by RNA sequencing (RNA-seq). Pm20d1, a gene encoding PM20D1 secretase that promotes the generation of endogenous mitochondria uncouplers in vivo, was dramatically upregulated in the midbrain of kir6.2-/- mice. Further investigations verified that PM20D1-induced increase of N-acyl amino acids (N-AAAs) from circulating fatty acids and amino acids promoted mitochondrial impairments and cut down the ATP generation, which mediated the morphological defects of the mesencephalic neurons and thus led to the behavioral impairments of kir6.2 knockout mice. This study is the first evidence to demonstrate the roles of kir6.2 in the morphological maintenance of neurite and motor coordination control of mice, which extends our understanding of kir6.2/KATP channels in regulating the neurophysiological function.


Assuntos
Amidoidrolases/metabolismo , Canais KATP , Neuritos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Trifosfato de Adenosina , Animais , Camundongos , Camundongos Knockout , Mitocôndrias/genética
7.
J Biol Chem ; 296: 100517, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33676895

RESUMO

G protein-coupled receptors (GPCRs) are important modulators of synaptic functions. A fundamental but poorly addressed question in neurobiology is how targeted GPCR trafficking is achieved. Rab GTPases are the master regulators of vesicle-mediated membrane trafficking, but their functions in the synaptic presentation of newly synthesized GPCRs are virtually unknown. Here, we investigate the role of Rab43, via dominant-negative inhibition and CRISPR-Cas9-mediated KO, in the export trafficking of α2-adrenergic receptor (α2-AR) and muscarinic acetylcholine receptor (mAChR) in primary neurons and cells. We demonstrate that Rab43 differentially regulates the overall surface expression of endogenous α2-AR and mAChR, as well as their signaling, in primary neurons. In parallel, Rab43 exerts distinct effects on the dendritic and postsynaptic transport of specific α2B-AR and M3 mAChR subtypes. More interestingly, the selective actions of Rab43 toward α2B-AR and M3 mAChR are neuronal cell specific and dictated by direct interaction. These data reveal novel, neuron-specific functions for Rab43 in the dendritic and postsynaptic targeting and sorting of GPCRs and imply multiple forward delivery routes for different GPCRs in neurons. Overall, this study provides important insights into regulatory mechanisms of GPCR anterograde traffic to the functional destination in neurons.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transmissão Sináptica , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Células HEK293 , Humanos , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Proteínas rab de Ligação ao GTP/genética
8.
Cell Death Differ ; 28(6): 1822-1836, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33686256

RESUMO

Although ß-arrestins (ARRBs) regulate diverse physiological and pathophysiological processes, their functions and regulation in Parkinson's disease (PD) remain poorly defined. In this study, we show that the expression of ß-arrestin 1 (ARRB1) and ß-arrestin 2 (ARRB2) is reciprocally regulated in PD mouse models, particularly in microglia. ARRB1 ablation ameliorates, whereas ARRB2 knockout aggravates, the pathological features of PD, including dopaminergic neuron loss, neuroinflammation and microglia activation in vivo, and microglia-mediated neuron damage in vitro. We also demonstrate that ARRB1 and ARRB2 produce adverse effects on inflammation and activation of the inflammatory STAT1 and NF-κB pathways in primary cultures of microglia and macrophages and that two ARRBs competitively interact with the activated form of p65, a component of the NF-κB pathway. We further find that ARRB1 and ARRB2 differentially regulate the expression of nitrogen permease regulator-like 3 (Nprl3), a functionally poorly characterized protein, as revealed by RNA sequencing, and that in the gain- and loss-of-function studies, Nprl3 mediates the functions of both ARRBs in microglia inflammatory responses. Collectively, these data demonstrate that two closely related ARRBs exert opposite functions in microglia-mediated inflammation and the pathogenesis of PD which are mediated at least in part through Nprl3 and provide novel insights into the understanding of the functional divergence of ARRBs in PD.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Inflamação/genética , Microglia/metabolismo , Doença de Parkinson/genética , beta-Arrestina 1/metabolismo , beta-Arrestina 2/metabolismo , Idoso , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Doença de Parkinson/patologia , Transdução de Sinais
9.
Front Pharmacol ; 11: 618992, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584303

RESUMO

Glia-mediated inflammatory processes are crucial in the pathogenesis of Parkinson's disease (PD). As the most abundant cells of the brain and active participants in neuroinflammatory responses, astrocytes largely propagate inflammatory signals and amplify neuronal loss. Hence, intensive control of astrocytic activation is necessary to prevent neurodegeneration. In this study, we report that the astrocytic kir6.2, as a abnormal response after inflammatory stimuli, promotes the reactivity of A1 neurotoxic astrocytes. Using kir6.2 knockout (KO) mice, we find reversal effects of kir6.2 deficiency on A1-like astrocyte activation and death of dopaminergic neurons in lipopolysaccharide (LPS)-induced mouse models for PD. Further in vitro experiments show that aberrant kir6.2 expression induced by inflammatory irritants in astrocytes mediates the dynamin-related protein 1 (Drp1)-dependent excessive mitochondrial fragmentation and results in mitochondrial malfunctions. By deleting kir6.2, astrocytic activation is reduced and astrocytes-derived neuronal injury is prevented. We therefore conclude that astrocytic kir6.2 can potentially elucidate the pathology of PD and promote the development of therapeutic strategies for PD.

10.
J Neuroinflammation ; 15(1): 210, 2018 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-30029608

RESUMO

BACKGROUND: Alzheimer's disease is characterized by progressive accumulation of ß-amyloid (Aß)-containing amyloid plaques, and microglia play a critical role in internalization and degradation of Aß. Our previous research confirmed that Nogo-66 binding to Nogo receptors (NgR) expressed on microglia inhibits cell adhesion and migration in vitro. METHODS: The adhesion and migration of microglia isolated from WT and APP/PS1 mice from different ages were measured by adhesion assays and transwells. After NEP1-40 (a competitive antagonist of Nogo/NgR pathway) was intracerebroventricularly administered via mini-osmotic pumps for 2 months in APP/PS1 transgenic mice, microglial recruitment toward Aß deposits and CD36 expression were determined. RESULTS: In this paper, we found that aging led to a reduction of microglia adhesion and migration to fAß1-42 in WT and APP/PS1 mice. The adhesion and migration of microglia to fAß1-42 were downregulated by the Nogo, which was mediated by NgR, and the increased inhibitory effects of the Nogo could be observed in aged mice. Moreover, Rho GTPases contributed to the effects of the Nogo on adhesion and migration of microglia to fAß1-42 by regulating cytoskeleton arrangement. Furthermore, blocking the Nogo/NgR pathway enhanced recruitment of microglia toward Aß deposits and expression of CD36 in APP/PS1 mice. CONCLUSION: Taken together, Nogo/NgR pathway could take part in Aß pathology in AD by modulating microglial adhesion and migration to Aß and the Nogo/NgR pathway might be an important target for treating AD.


Assuntos
Envelhecimento , Peptídeos beta-Amiloides/farmacologia , Adesão Celular/efeitos dos fármacos , Microglia/efeitos dos fármacos , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Fragmentos de Peptídeos/farmacologia , Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Adesão Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Mielina/farmacologia , Presenilina-1/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
11.
Biochem Pharmacol ; 150: 245-255, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29447945

RESUMO

ß2-Aderenergic receptor (ß2AR) agonist, Salmeterol exhibits anti-inflammatory activities. However, the inhibitory effects of Salmeterol on inflammasome activation are elusive and the underlying mechanisms need to be explored. In this study, we established inflammatory model in primary bone marrow-derived macrophages (BMDM) from C57BL/6J mice and ß-arrestin2 knockout (ß-arrestin2-/-) mice in vitro. In vivo study by LPS intraperitoneally (i.p.) in C57BL/6J mice was carried out to ascertain its roles in systemic inflammation. We found that Salmeterol (10-10 M-10-7 M) prevented the cleavage of caspase-1 and the activation of NLRP3 inflammasome, reduced the release of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in vitro. Blockade of adenosine3',5'cyclic monophosphate (cAMP)/protein kinase A (PKA) pathway with cAMP or PKA inhibitors inhibited anti-inflammatory effects of Salmeterol only at 10-7 M. Depletion of ß-arrestin2 compromised the inhibitory effects of Salmeterol at both 10-10 M and 10-7 M. Salmeterol increased the interaction of ß-arrestin2 and NLRP3. In vivo study showed that Salmeterol decreased the serum concentrations of pro-inflammatory cytokines IL-1ß and TNF-α, blocked cleavage of caspase-1 and release of IL-1ß in BMDM. These findings imply that Salmeterol at low concentrations (10-10 M-10-7 M) shows anti-inflammatory effect via inhibiting NLRP3 inflammasome. The underlying mechanisms is dosage-dependent: Salmeterol at 10-10 M shows anti-inflammatory effects through ß-arrestin2 pathway, and 10-7 M Salmeterol inhibits inflammation via both classical G-protein coupled receptor (GPCR)/cAMP pathway and ß-arrestin2 pathway. These results provide new ideas for the future treatment of systemic inflammation and other inflammatory diseases.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Inflamassomos/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Xinafoato de Salmeterol/farmacologia , Animais , Relação Dose-Resposta a Droga , Inflamassomos/metabolismo , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
12.
Mol Neurobiol ; 54(6): 4217-4231, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27335029

RESUMO

Disability including deficiency in sensorimotor and cognition functions is a dominant consequence after stroke. Evidence suggests that serine proteases play an important role in the physiology and pathology of the brain. Previous studies reported that nafamostat mesilate (NM), a synthetic serine protease inhibitor, attenuates neuronal damage in the acute phase after stroke. However, its efficacy in the chronic phase and the mechanism underlying its beneficial effect are not fully known. Here, we have studied whether NM improves long-term functional recovery after ischemic stroke. Experimental ischemic stroke was induced by transient middle cerebral artery occlusion (tMCAO). NM treatment attenuated the brain infarct volume and the loss of body weight and improved the recovery of sensorimotor and cognitive functions. One month after tMCAO, neuronal axons and dendrites were preserved in the NM group, accompanied by increasedsynaptic proteins and structures in the ipsilateral hippocampus. The expression of brain-derived neurotrophic factor, nerve growth factor and neurotrophin-3 was increased in the contralateral sensorimotor cortex and ipsilateral hippocampus by the administration of NM. Furthermore, NM activated tyrosine receptor kinase B (TrkB), extracellular signal-regulated kinas1/2(ERK1/2) and cAMP-response element binding protein (CREB) and inhibited the activity of Cyclin-dependent Kinase 5 (Cdk5) in the contralateral sensorimotor cortex and ipsilateral hippocampus. These results demonstrated that NM treatment could improve neurological outcome and axonal regeneration, which might be correlated with down-regulating Cdk5 activity and up-regulating TrkB-ERK1/2-CREB pathway.


Assuntos
Axônios/patologia , Guanidinas/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia , Animais , Axônios/efeitos dos fármacos , Benzamidinas , Peso Corporal/efeitos dos fármacos , Infarto Encefálico/complicações , Infarto Encefálico/patologia , Cognição/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Guanidinas/farmacologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Masculino , Modelos Neurológicos , Fatores de Crescimento Neural/metabolismo , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/complicações , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Resultado do Tratamento
13.
Brain Behav Immun ; 56: 230-45, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27033633

RESUMO

Inflammation plays an important role in stroke pathology, making it a promising target for stroke intervention. Nafamostat mesilate (NM), a wide-spectrum serine protease inhibitor, is commonly used for treating inflammatory diseases, such as pancreatitis. However, its effect on neuroinflammation after stroke was unknown. Hence, the effects of NM on the inflammatory response post stroke were characterized. After transient middle cerebral artery occlusion (tMCAO) in rats, NM reduced the infarct size, improved behavioral functions, decreased the expression of proinflammatory mediators (TNF-α, IL-1ß, iNOS and COX-2) in a time-dependent manner and promoted the expression of different anti-inflammatory factors (CD206, TGF-ß, IL-10 and IL-4) at different time points. Furthermore, NM could inhibit the expression of proinflammatory mediators and promote anti-inflammatory mediators expression in rat primary microglia following exposure to thrombin combined with oxygen-glucose deprivation (OGD). The immune-modulatory effect of NM might be partly due to its inhibition of the NF-κB signaling pathway and inflammasome activation after tMCAO. In addition, NM significantly inhibited the infiltration of macrophage, neutrophil and T lymphocytes, which was partly mediated by the inhibition of monocyte chemotactic protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Taken together, our results indicated that NM can provide long-term protection of the brain against tMCAO by modulating a broad components of the inflammatory response.


Assuntos
Comportamento Animal/efeitos dos fármacos , Guanidinas/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Inflamação/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Animais , Benzamidinas , Modelos Animais de Doenças , Guanidinas/administração & dosagem , Inflamassomos/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Inibidores de Serina Proteinase/administração & dosagem , Trombina/efeitos dos fármacos
14.
J Neuroinflammation ; 13(1): 56, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26939570

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by extracellular ß-amyloid (Aß) plaques, neurofibrillary tangles (NFTs), and microglia-dominated neuroinflammation. The Nogo/NgR signal pathway is involved in AD pathological features, but the detailed mechanism needs further investigation. Our previous studies have confirmed that the activation of NgR on microglia by Nogo promotes the expression of proinflammatory cytokines and inhibits cell adhesion and migration behaviors. In the present study, we investigated the effects of Nogo/NgR signaling pathway on the pathological features of AD and possible mechanisms. METHODS: After NEP1-40 (a competitive antagonist of Nogo/NgR pathway) was intracerebroventricularly administered via mini-osmotic pumps for 2 months in amyloid precursor protein (APP)/PS1 transgenic mice, plaque load, tau phosphorylation, and inflammatory responses were determined. After primary mouse neurons were exposed to the conditioned medium from BV-2 microglia stimulated by Nogo, the production of Aß and phosphorylation of tau was quantified by ELISA and western blot. RESULTS: Inhibition of the Nogo/NgR signaling pathway ameliorated pathological features including amyloid plaques and phosphorylated levels of tau in APP/PS1 mice. In addition, after treatment with the conditioned medium from BV-2 microglia stimulated by Nogo, Aß production and tau phosphorylation in cultured neurons were increased. The conditioned medium also increased the expression of APP, its amyloidogenic processing, and the activity of GSK3ß in neurons. The conditioned medium was also proinflammatory medium, and the blockage of the Nogo/NgR pathway improved the neuroinflammatory environment in APP/PS1 mice. CONCLUSIONS: Taken together, the neuroinflammation mediated by Nogo/NgR pathway in microglia could directly take part in the pathological process of AD by influencing the amyloidogenesis and tau phosphorylation. These results contribute to a better understanding of AD pathogenesis and could offer a new therapeutic option for delaying the progression of AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Microglia/efeitos dos fármacos , Proteínas da Mielina/farmacologia , Proteínas Nogo/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Placa Amiloide/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Meios de Cultivo Condicionados , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
15.
Neuropharmacology ; 105: 398-410, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26861077

RESUMO

Serine proteases, such as thrombin, are contributors to the disruption of the blood-brain barrier (BBB) and exacerbate brain damage during ischemic stroke, for which the current clinical therapy remains unsatisfactory. However, the effect of nafamostat mesilate (NM), a synthetic serine protease inhibitor, on BBB disruption following cerebral ischemia is unknown. Here, we investigated the in vivo effect of NM on BBB integrity in rats subjected to transient middle cerebral artery occlusion (MCAO) and explored the possible mechanism in an in vitro BBB model comprising rat brain microvascular endothelial cells and astrocytes after oxygen and glucose deprivation (OGD) in the presence of thrombin. The results showed that NM treatment remarkably attenuated transient MCAO-induced brain infarcts, brain oedema and motor dysfunction in addition to BBB disruption, which might be related to changes in tight junction protein expression and localization. Meanwhile, NM preserved BBB integrity and alleviated the changes in tight junction protein expression and localization and cytoskeleton rearrangement in rat brain microvascular endothelial cells via thrombin inhibition. Our findings suggest that NM treatment can preserve BBB integrity through the inhibition of thrombin, which might be correlated with the regulation of PKCα/RhoA/MLC2 pathway components.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/prevenção & controle , Guanidinas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Animais , Astrócitos/efeitos dos fármacos , Benzamidinas , Edema Encefálico/prevenção & controle , Células Endoteliais/efeitos dos fármacos , Glucose/deficiência , Hipóxia/prevenção & controle , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Transtornos dos Movimentos/prevenção & controle , Ratos , Ratos Sprague-Dawley , Trombina/farmacologia , Proteínas de Junções Íntimas/antagonistas & inibidores
16.
J Biol Chem ; 290(48): 28901-14, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26472924

RESUMO

Microglia have been proposed to play a pivotal role in the inflammation response of the CNS by expressing a range of proinflammatory enzymes and cytokines under pathological stimulus. Our previous study has confirmed that Nogo receptor (NgR), an axon outgrowth inhibition receptor, is also expressed on microglia and regulates cell adhesion and migration behavior in vitro. In the present study, we further investigated the proinflammatory effects and possible mechanisms of Nogo on microglia in vitro. In this study, Nogo peptide, Nogo-P4, a 25-amino acid core inhibitory peptide sequence of Nogo-66, was used. We found that Nogo-P4 was able to induce the expression of inducible nitric-oxide synthase and cyclooxygenase-2 and the release of proinflammatory cytokines, including IL-1ß, TNF-α, NO, and prostaglandin E2 in microglia, which could be reversed by NEP1-40 (Nogo-66(1-40) antagonist peptide), phosphatidylinositol-specificphospholipase C, or NgR siRNA treatment. After Nogo-P4 stimulated microglia, the phosphorylation levels of NF-κB and STAT3 were increased obviously, which further mediated microglia expressing proinflammatory factors induced by Nogo-P4. Taken together, we concluded that Nogo peptide could directly take part in CNS inflammatory process by influencing the expression of proinflammatory factors in microglia, which were related to the NF-κB and STAT3 signal pathways. Besides neurite outgrowth restriction, the Nogo/NgR signal might be involved in multiple processes in various inflammation-associated CNS diseases.


Assuntos
Microglia/metabolismo , Proteínas da Mielina/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Microglia/patologia , Proteínas da Mielina/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Proteínas Nogo , Receptor Nogo 1 , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Acta Pharmacol Sin ; 34(1): 119-28, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23202804

RESUMO

AIM: To explore whether the synthetic cannabinoid receptor agonist WIN55,212-2 could protect oligodendrocyte precursor cells (OPCs) in stroke penumbra, thereby providing neuroprotection following permanent focal cerebral ischemia in rats. METHODS: Adult male SD rats were subjected to permanent middle cerebral artery occlusion (p-MCAO). The animals were administered WIN55,212-2 at 2 h, and sacrificed at 24 h after the ischemic insult. The infarct volumes and brain swelling were assessed. The expression of cannabinoid receptor type 1 (CB1) in the stroke penumbra was examined using Western blot assay. The pathological changes and proliferation of neural glial antigen 2-positive OPCs (NG2(+) cells) in the stroke penumbra were studied using immunohistochemistry staining. RESULTS: p-MCAO significantly increased the expression of CB1 within the stroke penumbra with the highest level appearing at 2 h following the ischemic insult. Administration of WIN55,212-2 (9 mg/kg, iv) significantly attenuated the brain swelling, and reduced the infarct volume as well as the number of tau-immunoreactive NG2(+) cells (tau-1(+)/NG2(+) cells) in the stroke penumbra. Moreover, WIN55,212-2 significantly promoted the proliferation of NG2(+) cells in the stroke penumbra and in the ipsilateral subventricular zone at 24 h following the ischemic insult. Administration of the selective CB1 antagonist rimonabant (1 mg/kg, iv) partially blocked the effects caused by WIN55,212-2. CONCLUSION: Tau-1 is expressed in NG2(+) cells following permanent focal cerebral ischemic injury. Treatment with WIN55,212-2 reduces the number of tau-1(+)/NG2(+) cells and promotes NG2(+) cell proliferation in the stroke penumbra, which are mediated partially via CB1 and may contribute to its neuroprotective effects.


Assuntos
Benzoxazinas/uso terapêutico , Encéfalo/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Morfolinas/uso terapêutico , Naftalenos/uso terapêutico , Animais , Anticorpos Monoclonais/análise , Encéfalo/patologia , Proliferação de Células/efeitos dos fármacos , Infarto da Artéria Cerebral Média/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/análise
18.
Brain Res ; 1491: 225-35, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23148948

RESUMO

In stroke, a common cause of neurological disability in adults is that the myelin sheaths are lost through the injury or death of mature oligodendrocytes, and the failure of remyelination may be often due to insufficient proliferation and differentiation of oligodendroglial progenitors. In the current study, we used middle cerebral artery occlusion (MCAO) to induced transient focal cerebral ischemia, and found that WIN55, 212-2 augmented actively proliferating oligodendrocytes measured by CC1 immunoreactive cells within the peri-infarct areas. To establish whether these effects were associated with changes in myelin formation, we analyzed the expression of myelin basic protein (MBP) and myelin ultrastructure. We found that WIN55, 212-2 showed more extensive remyelination than vehicle at 14 days post injection (dpi). The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) signaling pathway may be involved in OPCs differentiation. To determine the regulatory effect of WIN55, 212-2 post-treatment on phospho-ERK 1/2 (p-ERK 1/2) after ischemia/reperfusion, Western blot analysis was performed. We found that WIN55, 212-2 regulated the phosphorylation level of the ERK 1/2 to promote OPCs survival and differentiation. Notably, cannabinoid receptor 1 is coupled to the activation of the ERK cascade. Following rimonabant combined treatment, the effect of WIN55, 212-2 on regulating the phosphorylation level of the ERK 1/2 was reversed, and the effect of accelerated myelin formation was partially inhibited. Together, we first found that WIN55, 212-2 promoted OPCs differentiation and remyelination through regulation of the level of the p-ERK 1/2 via cannabinoid receptor 1.


Assuntos
Benzoxazinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Doenças Desmielinizantes/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Morfolinas/farmacologia , Bainha de Mielina/efeitos dos fármacos , Naftalenos/farmacologia , Oligodendroglia/efeitos dos fármacos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Animais , Antimetabólitos , Western Blotting , Isquemia Encefálica/patologia , Bromodesoxiuridina , Antagonistas de Receptores de Canabinoides/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Doenças Desmielinizantes/etiologia , Imuno-Histoquímica , Indicadores e Reagentes , Masculino , Microscopia Eletrônica , Proteína Básica da Mielina/biossíntese , Oligodendroglia/ultraestrutura , Fosforilação , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Rimonabanto , Acidente Vascular Cerebral/complicações
19.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 42(6): 615-9, 2013 11.
Artigo em Chinês | MEDLINE | ID: mdl-24421225

RESUMO

OBJECTIVE: To explore the synergistic antidepressant effect of quercetin and hyperforin (HF, extracted from Hypericum perforatum). METHODS: Male ICR mice were divided into nine groups:blank control, positive control (Paroxetine, 10 mg/kg), quercetin groups (A: 5 mg/kg, B: 10 mg/kg, C: 20 mg/kg), Hypericum perforatum extract (HF 10 mg/kg),combination groups (A: quercetin 2.5 mg/kg + HF 5 mg/kg,B:quercetin 5 mg/kg + HF 5 mg/kg,C: quercetin 10 mg/kg + HF 5 mg/kg). All drugs were administered intragastrically. Reserpine reversal tests were used to compare the reversal effects of drugs on body temperature decline, eyelid ptosis and akinesia. Tail suspension test was used to compare immobility time in each group. RESULTS: Combination group B showed no significant difference (P>0.05) compared with combination group C in reserpine reversal tests and tail suspension test. However, its body temperature reversal effect was significantly higher (P<0.01) than that of quercetin group B, and its effect in shortening immobility time was stronger than that of HF 10 mg/kg group (P<0.05) and quercetin group B (P<0.01). CONCLUSION: The combination of quercetin and Hypericum perforatum extract in certain ratio has significant synergistic antidepressant effect in ICR mice.


Assuntos
Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Hypericum/química , Quercetina/farmacologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos ICR , Extratos Vegetais/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA