Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 74: 105721, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31255881

RESUMO

Glucocorticoids (GCs) exposure has deleterious alteration on the structure and function in hippocampal neurons. NADPH oxidase 2 (NOX2) is a major contributor to oxidative stress in neurological diseases, and NLRP1 inflammasome can be activated in response to oxidative stress. We hypothesize that inhibition of NOX2-mediated NLRP1 inflammasome activation may protect against chronic GCs exposure-induced neuronal injury. In this study, the lentivirus with NLRP1-siRNA was injected into the hippocampus of male mice which were then treated with dexamethasone (DEX, 5 mg/kg) for 28 d. The data indicated that NLRP1-siRNA treatment down-regulated the NLRP1 expression and significantly improved the exploratory behavior and spatial memory deficits in open field tests and Morris water maze which were deteriorated by chronic DEX treatment in mice. Additionally, inhibition of NLRP1 expression significantly alleviated neuronal degeneration and increased MAP2 expression in the hippocampus in mice. Meanwhile, the results showed that DEX exposure increased NOX2, p22phox and p47phox expression in hippocampus tissue in mice. We further examined the effect of tempol (ROS scavenger) and apocynin (NOX inhibitor) treatment on NLRP1 inflammasome activation in chronic DEX-treated hippocampal neurons. The results revealed that the tempol (50 µM) and apocynin (50 µM) treatment significantly decreased generation of ROS, expression of NOX2 and NLRP1-related protein in DEX-treated hippocampal neurons. These data indicate that NOX2-mediated NLRP1 activation involves in chronic GCs exposure-induced neuronal injury and inhibition of NOX2-NLRP1 signaling pathway protects against GCs-induced neuronal damage.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Dexametasona/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Exposição Ambiental/efeitos adversos , Hipocampo/patologia , Neurônios/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/etiologia , Animais , Proteínas Reguladoras de Apoptose/genética , Comportamento Animal , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , NADPH Oxidase 2/metabolismo , Neurônios/patologia , Neuroproteção , RNA Interferente Pequeno/genética , Transdução de Sinais
2.
Int Immunopharmacol ; 69: 60-70, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30677569

RESUMO

Oxidative stress and inflammation are closely related to neuron ageing. NADPH oxidase 2 (NOX2) is a major source of reactive oxygen species (ROS) generation in brain. The nucleotide-binding oligomerisation domain (NOD)-like receptor protein 1 (NLRP1) inflammasome is responsible for the formation of proinflammatory molecules in neurons. We hypothesize that NOX2-derived ROS accumulation mediates activation of NLRP1 inflammasome, which is involved in age-related neuronal damage. In the present study, we investigated the changes of NOX2-NLRP1 signaling pathway in primary hippocampal neurons cultured for different time (6, 9 and 12 days, d). Meanwhile, we further examined the effect of ROS inhibitor and NLRP1-siRNA on neuronal senescence. The results showed that, compared with 6 d group, the neuronal apoptosis and ß-Galactosidase (ß-Gal) expression were significantly increased, and the microtubule-associated protein 2 (MAP2) expression significantly decreased in primary hippocampal neurons cultured for 12 d. In addition, the results also showed that the production of ROS, the expressions of NOX2 and NLRP1 inflammasome were significantly increased with the prolongation of culture time in hippocampal neurons. Moreover, the NOX inhibitor (apocynin) and ROS scavenger (tempol) significantly decreased ROS production and alleviated neuronal damage. Meanwhile, the tempol and apocynin treatment significantly decreased the expression of NLRP1 inflammasome in hippocampal neurons. Furthermore, the NLRP1-siRNA and caspase-1 inhibitor treatment also alleviated neuronal damage. These results suggest that NOX2-derived ROS generation may induce brain inflammation via NLRP-1 inflammasome activation and lead to age-related neuronal damage. The NADPH oxidase and NLRP1 inflammasome may be important therapeutic targets for age-related neuronal damage.


Assuntos
Hipocampo/patologia , Inflamação/metabolismo , NADPH Oxidase 2/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Animais , Apoptose , Células Cultivadas , Senescência Celular , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
3.
Int J Mol Med ; 43(2): 717-726, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30535505

RESUMO

Oxidative stress and neuroinflammation are important in the pathogenesis of ageing and age­related neurodegenerative diseases, including Alzheimer's disease. NADPH oxidase 2 (NOX2) is a major source of reactive oxygen species (ROS) in the brain. The nucleotide­binding oligomerisation domain (NOD)­like receptor protein 1 (NLRP1) inflammasome is responsible for the formation of pro­inflammatory molecules in neurons. Whether the NOX2­NLRP1 inflammasome signalling pathway is involved in neuronal ageing and age­related damage remains to be elucidated. Ginsenoside Rg1 (Rg1) is a steroidal saponin found in ginseng. In the present study, the primary hippocampal neurons were treated with H2O2 (200 µM) and Rg1 (1, 5 and 10 µM) for 24 h to investigate the protective effects and mechanisms of Rg1 on H2O2­induced hippocampal neuron damage, which mimics age­related damage. The results showed that H2O2 treatment significantly increased ROS production and upregulated the expression of NOX2 and the NLRP1 inflammasome, and led to neuronal senescence and damage to hippocampal neurons. Rg1 decreased ROS production, reducing the expression of NOX2 and the NLRP1 inflammasome in H2O2­treated hippocampal neurons. Furthermore, Rg1 and tempol treatment significantly decreased neuronal apoptosis and the expression of ß­galactosidase, and alleviated the neuronal senescence and damage induced by H2O2. The present study indicates that Rg1 may reduce NOX2­mediated ROS generation, inhibit NLRP1 inflammasome activation, and inhibit neuronal senescence and damage.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Ginsenosídeos/farmacologia , Hipocampo/patologia , Inflamassomos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Caspase 1/metabolismo , Células Cultivadas , Medicamentos de Ervas Chinesas/farmacologia , Peróxido de Hidrogênio , Inflamassomos/efeitos dos fármacos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , NADPH Oxidase 2/antagonistas & inibidores , Proteínas NLR/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , beta-Galactosidase/biossíntese
4.
J Neuroinflammation ; 14(1): 139, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28732502

RESUMO

BACKGROUND: Neuroinflammation mediated by NLRP1 (nucleotide-binding oligomerization domain (NOD)-like receptor protein 1) inflammasome plays an important role in many neurological diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). Our previous studies showed that chronic glucocorticoid (GC) exposure increased brain inflammation via NLRP1 inflammasome and induce neurodegeneration. However, little is known about the mechanism of chronic GC exposure on NLRP1 inflammasome activation in hippocampal neurons. METHODS: Hippocampal neurons damage was assessed by LDH kit and Hoechst 33258 staining. The expression of microtubule-associated protein 2 (MAP2), inflammasome complex protein (NLRP1, ASC and caspase-1), inflammatory cytokines (IL-1ß), and large-conductance Ca2+ and voltage-activated K+ channel (BK channels) protein was detected by Western blot. The inflammatory cytokines (IL-1ß and IL-18) were examined by ELISA kit. The mRNA levels of NLRP1, IL-1ß, and BK were detected by real-time PCR. BK channel currents were recorded by whole-cell patch-clamp technology. Measurement of [K+]i was performed by ion-selective electrode (ISE) technology. RESULTS: Chronic dexamethasone (DEX) treatment significantly increased LDH release and neuronal apoptosis and decreased expression of MAP2. The mechanistic studies revealed that chronic DEX exposure significantly increased the expression of NLRP1, ASC, caspase-1, IL-1ß, L-18, and BK protein and NLRP1, IL-1ß and BK mRNA levels in hippocampal neurons. Further studies showed that DEX exposure results in the increase of BK channel currents, with the subsequent K+ efflux and a low concentration of intracellular K+, which involved in activation of NLRP1 inflammasome. Moreover, these effects of chronic DEX exposure could be blocked by specific BK channel inhibitor iberiotoxin (IbTx). CONCLUSION: Our findings suggest that chronic GC exposure may increase neuroinflammation via activation of BK-NLRP1 signal pathway and promote hippocampal neurons damage, which may be involved in the development and progression of AD.


Assuntos
Lesões Encefálicas/induzido quimicamente , Dexametasona/toxicidade , Hipocampo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinaptotagminas/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Lesões Encefálicas/patologia , Células Cultivadas , Modelos Animais de Doenças , Glucocorticoides/toxicidade , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Peptídeos/toxicidade , Ratos , Sinaptotagminas/genética
5.
Int Immunopharmacol ; 49: 222-230, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28605710

RESUMO

Neuroinflammation mediated by NLRP-1 inflammasome plays an important role in the pathogenesis of neurodegeneration diseases such as Alzheimer's disease (AD). Chronic glucocorticoids (GCs) exposure has deleterious effect on the structure and function of neurons and was found to be correlated with development and progression of AD. We hypothesize that chronic glucocorticoids may down-regulate the expression of glucocorticoids receptor (GR) and activate NLRP-1 inflammasome in hippocampal neurons, which may promote neuroinflammation and induce neuronal injury. The present results showed that chronic DEX exposure significantly increased LDH release and apoptosis, decreased MAP2 and GR expression in hippocampal neurons. DEX (5µΜ) exposure for 3d significantly increased the expression of NLRP-1, ASC, caspase-1 and IL-1ß in the hippocampal neurons and the release of IL-1ß and IL-18 in the supernatants. Moreover, DEX (1, 5µΜ) treatment for 3d significantly increased the expression of NF-κB in hippocampal neurons. The GR antagonist, mifepristone (RU486), had protective effects on chronic DEX induced hippocampal neurons injury and NLRP1 inflammasome activation. The results suggest that chronic GCs exposure can decrease GR expression and increase neuroinflammation via NLRP1 inflammasome and promote hippocampal neurons degeneration, which may play an important role in the progression and development of AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/epidemiologia , Proteínas Reguladoras de Apoptose/metabolismo , Dexametasona/efeitos adversos , Hipocampo/patologia , Inflamassomos/metabolismo , Efeitos Adversos de Longa Duração/epidemiologia , Neurônios/patologia , Doença de Alzheimer/etiologia , Animais , Morte Celular , Células Cultivadas , Dexametasona/uso terapêutico , Humanos , Mifepristona/farmacologia , NF-kappa B/metabolismo , Proteínas NLR , Inflamação Neurogênica/etiologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...