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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Mol Med ; 22(6): 3159-3166, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29536648

RESUMO

Fingolimod (FTY720) is used as an immunosuppressant for multiple sclerosis. Numerous studies indicated its neuroprotective effects in stroke. However, the mechanism remains to be elucidated. This study was intended to investigate the mechanisms of phosphorylated FTY720 (pFTY720), which was the principle active molecule in regulating astrocyte-mediated inflammatory responses induced by oxygen-glucose deprivation (OGD). Results demonstrated that pFTY720 could protect astrocytes against OGD-induced injury and inflammatory responses. It significantly decreased pro-inflammatory cytokines, including high mobility group box 1 (HMGB1) and tumour necrosis factor-α (TNF-α). Further, studies displayed that pFTY720 could prevent up-regulation of Toll-like receptor 2 (TLR2), phosphorylation of phosphoinositide 3-kinase (PI3K) and nuclear translocation of nuclear factor kappa B (NFκB) p65 subunit caused by OGD. Sphingosine-1-phosphate receptor 3 (S1PR3) knockdown could reverse the above change. Moreover, administration of TLR2/4 blocker abolished the protective effects of pFTY720. Taken together, this study reveals that pFTY720 depends on S1PR3 to protect astrocytes against OGD-induced neuroinflammation, due to inhibiting TLR2/4-PI3K-NFκB signalling pathway.


Assuntos
Cloridrato de Fingolimode/farmacologia , Inflamação/tratamento farmacológico , Receptores de Lisoesfingolipídeo/genética , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Animais , Astrócitos/efeitos dos fármacos , Carência Cultural , Citocinas/genética , Modelos Animais de Doenças , Cloridrato de Fingolimode/química , Proteína HMGB1/genética , Humanos , Imunossupressores/química , Imunossupressores/farmacologia , Inflamação/genética , Inflamação/patologia , NF-kappa B/genética , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Cultura Primária de Células , Ratos , Receptores de Lisoesfingolipídeo/química , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato , Fator de Necrose Tumoral alfa/genética
2.
J Neuroinflammation ; 13(1): 60, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26961366

RESUMO

BACKGROUND: It is generally recognized that the inflammatory reaction in glia is one of the important pathological factors in brain ischemic injury. Our previous study has revealed that opening ATP-sensitive potassium (K-ATP) channels could attenuate glial inflammation induced by ischemic stroke. However, the detailed mechanisms are not well known. METHODS: Primary cultured astrocytes separated from C57BL/6 mice were subjected to oxygen-glucose deprivation (OGD); cellular injuries were determined via observing the changes of cellular morphology and cell viability. MicroRNA (miR) and messenger RNA (mRNA) level was validated by real-time PCR. The interaction between microRNA and the target was confirmed via dual luciferase reporter gene assay. Expressions of proteins and inflammatory cytokines were respectively assessed by western blotting and enzyme-linked immunosorbent assay. RESULTS: OGD resulted in astrocytic damage, which was prevented by K-ATP channel opener nicorandil. Notably, we found that OGD significantly downregulated miR-7 and upregulated Herpud2. Our further study proved that miR-7 targeted Herpud2 3'UTR, which encoded endoplasmic reticulum (ER) stress protein-HERP2. Correspondingly, our results showed that OGD increased the levels of ER stress proteins along with significant elevations of pro-inflammatory cytokines, including tumor necrosis factor α (TNF-α) and interleukin 1ß (IL-1ß). Pretreatment with nicorandil could remarkably upregulate miR-7, depress the ER-related protein expressions including glucose-regulated protein 78 (GRP78), C/EBP-homologous protein (CHOP), and Caspase-12, and thereby attenuate inflammatory responses and astrocytic damages. CONCLUSIONS: These findings demonstrate that opening K-ATP channels protects astrocytes against OGD-mediated neuroinflammation. Potentially, miR-7-targeted ER stress acts as a key molecular brake on neuroinflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Glucose/deficiência , Inflamação/tratamento farmacológico , MicroRNAs/fisiologia , Nicorandil/farmacologia , Canais de Potássio/agonistas , Animais , Astrócitos/ultraestrutura , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sobrevivência Celular , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/efeitos dos fármacos , Cultura Primária de Células , Proteínas Repressoras/metabolismo
3.
J Cell Mol Med ; 19(8): 1949-55, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25858687

RESUMO

Paclitaxel is generally used to treat cancers in clinic as an inhibitor of cell division. However, the acquired resistance in tumours limits its clinical efficacy. Therefore, the aim of this study was to detect whether co-treatment with lentinan enhanced the anti-cancer effects of paclitaxel in A549 cells. We found that the combination of paclitaxel and lentinan resulted in a significantly stronger inhibition on A549 cell proliferation than paclitaxel treatment alone. Co-treatment with paclitaxel and lentinan enhanced cell apoptosis rate by inducing caspase-3 activation. Furthermore, co-treatment with paclitaxel and lentinan significantly triggered reactive oxygen species (ROS) production, and increased thioredoxin-interacting protein (TXNIP) expression. Moreover, co-treatment with paclitaxel and lentinan enhanced TXNIP-NLRP3 interaction, and activated NLRP3 inflammasome whereat interleukin-1ß levels were increased and cell apoptosis was induced. In addition, combination of paclitaxel and lentinan could activate apoptosis signal regulating kinase-1 (ASK1)/p38 mitogen-activated protein kinase (MAPK) signal which also contributed to cell apoptosis. Taken together, co-treatment with paclitaxel and lentinan exerts synergistic apoptotic effects in A549 cells through inducing ROS production, and activating NLRP3 inflammasome and ASK1/p38 MAPK signal pathway.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Lentinano/farmacologia , Paclitaxel/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Humanos , MAP Quinase Quinase Quinase 5 , Modelos Biológicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Clin Exp Pharmacol Physiol ; 42(7): 766-71, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25998857

RESUMO

It has been reported that the novel ATP-sensitive potassium (K-ATP) channel opener iptakalim (IPT) decreases ischemic neuronal damage in rats. However, the mechanisms underlying neuroprotection are still to be fully elucidated. The results of this study showed that mice with ischemia induced by middle cerebral artery occlusion exhibited higher mortality and more neurological deficits, as well as larger infarct volume, compared with sham mice. Moreover, it was found that ischemia activated astrocytes surrounding CA1 neurons with an increased expression of D-serine, induced greater microglial activation accompanied by higher tumor necrosis factor alpha (TNF-α) production, and caused higher expressions of matrix metalloproteinase 9 (MMP-9) in the endothelial cells of mice. Pretreatment with IPT significantly attenuated the neurological deficits and decreased the infarct volume in mice. IPT treatment could decrease MMP-9 secretion, inhibit astrocytic activation with decreasing D-serine and elevating connexin43 expression. Microglial activation was also inhibited and TNF-α production was decreased by IPT. Taken together, a K-ATP channel opener may improve the function of neurovascular unit and protect against ischemic injury. These findings suggest that targeting K-ATP channels provides a promising therapeutic approach for stroke.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Propilaminas/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais KATP/metabolismo , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Life Sci ; 150: 8-14, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26920629

RESUMO

AIMS: Memantine is a low-moderate affinity and uncompetitive N-methyl-d-aspartate receptor (NMDAR) antagonist, which is also a potential neuroprotectant in acute ischemic stroke for its particular action profiles. The present study was to reveal the mechanisms involved in the neuroprotection of memantine. MAIN METHODS: We used a mouse model of permanent focal cerebral ischemia via middle cerebral artery occlusion to verify our hypothesis. 2,3,5-Triphenyltetrazolium chloride staining was used to compare infarct size. The amount of astrocytes and the somal volume of the microglia cell body were analyzed by immunohistochemistry and stereological estimates. Western blotting was used to determine the protein expressions. KEY FINDINGS: Memantine prevented cerebral ischemia-induced brain infarct and neuronal injury, and reduced oxygen-glucose deprivation-induced cortical neuronal apoptosis. Moreover, memantine reduced the amount of the damaged astrocytes and over activated microglia after 24h of ischemia. In the early phase of ischemia, higher production of MMP-9 was observed, and thereby collagen IV was dramatically disrupted. Meanwhile, the post-synaptic density protein 95(PSD-95) was also severely cleavaged. Memantine decreased MMP-9 secretion, prevented the degradation of collagen IV in mouse brain. PSD-95 cleavage was also inhibited by memantine. SIGNIFICANCE: These results suggested that memantine exerted neuroprotection effects in acute ischemic brain damage, partially via improving the functions of neurovascular unit. Taking all these findings together, we consider that memantine might be a promising protective agent against ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/fisiopatologia , Veias Cerebrais/inervação , Memantina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Colágeno Tipo IV/metabolismo , Proteína 4 Homóloga a Disks-Large , Glucose/deficiência , Guanilato Quinases/metabolismo , Hipóxia Encefálica/patologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Cultura Primária de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA