Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neuroscience ; 437: 87-97, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31982471

RESUMO

Brain ischaemia, which can cause severe nerve injury, is a global health challenge. Long non-coding RNA (lncRNA) growth-arrest specific 5 (Gas5) has been documented to exert tumour suppressive effects in several cancers. However, its role in cerebrovascular disease still requires further investigation. Therefore, in this study, we focused on the role of lncRNA regulatory signalling related to lncRNA Gas5 in ischaemic brain injury. Middle cerebral artery occlusion (MCAO) was employed as a model of ischaemic brain injury in rats. The expression of lncRNA Gas5 and microRNA-21 (miR-21) was altered in neurons to elucidate their effects in ischaemic brain injury and to identify the interactions among lncRNA Gas5, miR-21 and Pten. The neuronal survival rate, apoptosis and the expression of phosphatidyl inositol 3-kinase (PI3K)/Akt signalling pathway-related genes were also evaluated in vitro to determine the effects of lncRNA Gas5. In the brains of rats subjected to MCAO, the expression of lncRNA Gas5 and Pten was upregulated, while miR-21 was downregulated. LncRNA Gas5 inhibited miR-21 expression, leading to elevated levels of Pten. In vitro experiments revealed that lncRNA Gas5 depletion and miR-21 elevation resulted in the suppression of neuronal apoptosis, thus promoting neuronal survival via the PI3K/Akt signalling pathway. These findings demonstrate that lncRNA Gas5 increases miR-21 and activates Pten, contributing to the development of ischaemic brain injury, supporting the silencing of lncRNA Gas5 as a possible therapeutic target for the treatment of ischaemic brain injury.


Assuntos
Lesões Encefálicas , MicroRNAs , RNA Longo não Codificante , Animais , Regulação para Baixo , MicroRNAs/genética , Fosfatidilinositol 3-Quinases , RNA Longo não Codificante/genética , Ratos
2.
J Cell Physiol ; 235(3): 2414-2428, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31517390

RESUMO

Ischemic stroke (IS), which is characterized by high morbidity, disability, and mortality, is recognized as a major cerebrovascular disease. MicroRNA-31 (miR-31) was reported to participate in the progression of brain disease. The present study was conducted in order to investigate the effect of miR-31 on oxidative stress-induced neuronal injury in IS mice with the involvement of protein kinase D1 (PKD1) and the JAK/STAT3 pathway. C57BL/6J mice were used to establish the middle cerebral artery occlusion (MCAO) model. Astrocytes were transfected with miR-31 mimic, miR-31 inhibitor, si-PKD1, or JAK-STAT3 pathway inhibitor. Following the establishment of an oxygen-glucose deprivation (OGD) model, the astrocytes were cocultured with neuronal OGD. Lower miR-31, higher PKD1 expressions, and activated JAK/STAT3 pathway were found in both the MCAO and OGD models. miR-31 could negatively target PKD1. In an MCAO model, overexpressing miR-31 and silencing PKD1 reduced neuronal injury, cerebral infarct volume, neuron loss, and oxidative stress injury, inhibited the activation of JAK/STAT3 pathway and the expressions of PKD1, interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, malondialdehyde, 4-HNE, 8-HOdG, caspase-3, and Bax, but increased the superoxide dismutase content. In the OGD model, overexpression of miR-31 and silencing of PKD1 attenuated oxidative stress-induced neuronal injury, and diminished the lactate dehydrogenase leakage and reactive oxygen species level, accompanied by elevated neuronal viability. These results indicate that miR-31 alleviates inflammatory response as well as an oxidative stress-induced neuronal injury in IS mice by downregulating PKD1 and JAK/STAT3 pathway.


Assuntos
Isquemia Encefálica/genética , AVC Isquêmico/genética , MicroRNAs/genética , Proteína Quinase C/genética , Animais , Apoptose/genética , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Glucose/metabolismo , Humanos , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/genética , Oxigênio/metabolismo , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética
3.
J Cell Physiol ; 234(2): 1477-1490, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30144062

RESUMO

Ischemic stroke, caused by the blockage of blood supply, is a major cause of death worldwide. For identifying potential candidates, we explored the effects microRNA-150 (miR-150) has on ischemic stroke and its underlying mechanism by developing a stable middle cerebral artery occlusion (MCAO) rat model. Gene expression microarray analysis was performed to screen differentially expressed genes associated with MCAO. We evaluated the expression of miR-150 and Mal and the status of ERK1/2 axis in the brain tissues of MCAO rats. Then the cerebral cortical neurons (CCNs) were obtained and introduced with elevated or suppressed miR-150 or silenced Mal to validate regulatory mechanisms for miR-150 governing Mal in vitro. The relationship between miR-150 and Mal was verified by dual luciferase reporter gene assay. Besides, cell growth and apoptosis of CCNs were detected by means of MTT assay and flow cytometry analyses. We identified Mal as a downregulated gene in MCAO, based on the microarray data of GSE16561. MiR-150 was over-expressed and negatively targeted Mal in the brain tissues obtained from MCAO rats and their CCNs. Increasing miR-150 blocked the ERK1/2 axis, resulting in an inhibited cell growth of CNNs but an enhanced apoptosis. Furthermore, MiR-150 inhibition was observed to have effects on CNNs as opposed to those inhibited by miR-150 promotion. The key findings of this study support the notion that miR-150 under-expression-mediated direct promotion of Mal protects CNN functions through the activation of the ERK1/2 axis, and underscore the concept that miR-150 may represent a novel pharmacological target for ischemic stroke intervention.


Assuntos
Apoptose , Córtex Cerebral/enzimologia , Infarto da Artéria Cerebral Média/enzimologia , MicroRNAs/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/metabolismo , Neurônios/enzimologia , Animais , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Córtex Cerebral/patologia , Modelos Animais de Doenças , Humanos , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Sistema de Sinalização das MAP Quinases , Masculino , MicroRNAs/genética , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/genética , Neurônios/patologia , Ratos Sprague-Dawley
4.
J Cell Physiol ; 234(5): 7341-7355, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30362547

RESUMO

OBJECTIVE: Ischemic stroke is known as a neurodegenerative disorder, which induces long-period tissue damage. Chemokine (C-X-C motif) ligand 8 (CXCL8) is involved in acute inflammation and tumor progression through the phosphoinositide-3-kinase/protein kinase B/nuclear factor-κB (PI3K/Akt/NF-κB)-signaling pathway. In this study, we aimed to explore the mechanism of CXCL8 in ischemic stroke in relation to the PI3K/Akt/NF-κB-signaling pathway. METHODS: Microarray-based gene expression profiling of peripheral blood mononuclear cells was used to identify ischemic stroke-related differentially expressed genes and explore role of CXCL8 in ischemic stroke. Next, the ischemic mice model was successfully established, with transfection efficiency detected. After that, deflection index, recovery of nervous system, infarct sizes, ischemia-induced apoptosis, and neuroinflammatory response in ischemic stroke were measured. At last, the content of inflammatory factors as well as the expression of CXCL8, caspase-3, caspase-9, Bad, interleukin-6 (IL-6), IL-1ß, tumor necrosis factor-α (TNF-α), Akt, PI3K, and NF-κB were determined. RESULTS: Comprehensive gene expression profiling analysis identified that CXCL8 might affect the development of ischemic stroke through regulating the PI3K/Akt/NF-κB-signaling pathway. CXCL8 silencing significantly reduced deflection index and infarct size, improved neurological function, and suppressed neuroglial cell loss and apoptosis index. In addition, glial fibrillary acidic portein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA-1) expressions were decreased following CXCL8 suppression, suggesting CXCL8 affected neuroglial activation. Importantly, we also found that CXCL8 silencing activated neuroglial cell and suppressed inflammatory cytokine production in ischemic stroke mice. CONCLUSION: Taken together, these findings highlight that functional suppression of CXCL8 promotes neuroglial activation and inhibits neuroinflammation by regulating the PI3K/Akt/NF-κB-signaling pathway in mice with ischemic stroke, which might provide new insight for ischemic stroke treatment.


Assuntos
Isquemia Encefálica/enzimologia , Encéfalo/enzimologia , Inflamação/enzimologia , Interleucina-8/genética , NF-kappa B/metabolismo , Neuroglia/enzimologia , Fosfatidilinositol 3-Quinase/metabolismo , Interferência de RNA , Acidente Vascular Cerebral/enzimologia , Animais , Apoptose , Comportamento Animal , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Citocinas/metabolismo , Bases de Dados Genéticas , Modelos Animais de Doenças , Humanos , Inflamação/genética , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Interleucina-8/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Neuroglia/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
5.
Cell Mol Neurobiol ; 29(3): 347-53, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18987968

RESUMO

The objective of the paper is to evaluate the effect of acellular nerve allografts (ANA) seeded with Schwann cells to promote nerve regeneration after bridging the sciatic nerve defects of rats and to discuss its acting mechanisms. Schwann cells were isolated from neonatal Wistar rats. In vitro Schwann cells were microinjected into acellular nerve allografts and co-cultured. Twenty-four Wistar rats weighing 180-220 g were randomly divided into three groups with eight rats in each group: ANA seeded with Schwann cells (ANA + SCs), ANA group and autografts group. All the grafts were, respectively, served for bridging a 10-mm long surgically created sciatic nerve gap. Examinations of regeneration nerve were performed after 12 weeks by transmission electron microscope (TEM), scanning electron microscope (SEM), and electrophysiological methods, and then analyzed statistically. The results obtained indicated that in vitro Schwann cells displayed the feature of bipolar morphology with oval nuclei. Compared with ANA group, the conduction velocity of ANA + SCs group and autograft group was faster after 12 weeks, latent period was shorter, and wave amplitude was higher (P < 0.05). The difference between ANA + SCs group and autograft group is not significant (P > 0.05). Regeneration nerve myelinated fiber number, myelin sheath thickness, and myelinated fibers/total nerves (%) in both ANA + SCs group and autograft group are higher than that in ANA group; the difference is significant (P < 0.05). The difference between the former two is not significant (P > 0.05). In conclusion, ANA seeded with SCs could improve nerve regeneration and functional recovery after bridging the sciatic nerve gap of rats, which offers a novel approach for the repair peripheral nerve defect.


Assuntos
Regeneração Nervosa , Células de Schwann/transplante , Nervo Isquiático/patologia , Nervo Isquiático/transplante , Animais , Fenômenos Eletrofisiológicos , Bainha de Mielina/ultraestrutura , Ratos , Ratos Wistar , Células de Schwann/ultraestrutura , Nervo Isquiático/fisiopatologia , Nervo Isquiático/ultraestrutura , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA