Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Cell Physiol Biochem ; 46(2): 781-792, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29621765

RESUMO

BACKGROUND/AIMS: Nuclear erythroid 2-related factor-2 (Nrf2) is a major stress-response transcription factor that has been implicated in regulating ischemic angiogenesis. We investigated the effects of Nrf2 in regulating revascularization and modulating acute lung injury. METHODS: The expression of Nrf2 and sirtuin1 (Sirt1) was assessed in lung tissue by western blotting and immunofluorescence staining after intestinal ischemia/reperfusion (IIR) in Nrf2-/- and wild-type (WT) mice. The involvement of Nrf2 in angiogenesis, cell viability, and migration was investigated in human pulmonary microvascular endothelial cells (PMVECs). Additionally, the influence of Nrf2 expression on NOX pathway activation was measured in PMVECs after oxygen-glucose deprivation/reoxygenation. RESULTS: We found activation and nuclear accumulation of Nrf2 in lung tissue after IIR. Compared to IIR in WT mice, IIR in Nrf2-/- mice significantly enhanced leukocyte infiltration and collagen deposit, and inhibited endothelial cell marker CD31 expression. Nrf2 upregulation and translocation into the nucleus stimulated by Sirt1 overexpression exhibited remission of histopathologic changes and enhanced CD31 expression. Nrf2 knockdown repressed non-phagocytic cell oxidase 4 (NOX4), hypoxia-inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF) expression after IIR. Nrf2 upregulation by Sirt1 enhances NOX4, HIF-1α and VEGF expression after IIR in WT mice. Furthermore, Nrf2 knockdown suppressed cell viability, capillary tube formation and cell migration in PMVECs after oxygen-glucose deprivation/reoxygenation and also inhibited NOX4, HIF-1 and VEGF expression. Moreover, NOX4 knockdown in PMVECs decreased the levels of VEGF, HIF-1α and angiogenesis. CONCLUSION: Nrf2 stimulation by Sirt1 plays an important role in sustaining angiogenic potential through NOX4-mediated gene regulation.


Assuntos
Lesão Pulmonar Aguda/patologia , NADPH Oxidase 4/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/patologia , Sirtuína 1/metabolismo , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Colágeno/metabolismo , Regulação para Baixo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Leucócitos/citologia , Leucócitos/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 4/antagonistas & inibidores , NADPH Oxidase 4/genética , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Neovascularização Fisiológica , Traumatismo por Reperfusão/complicações , Sirtuína 1/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Inhal Toxicol ; 28(14): 731-738, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27973945

RESUMO

Exposure to sevoflurane in neonatal rats could induce learning deficits and abnormal social behaviors, but the specific molecular mechanism is unknown. Postnatal day-7 SD rats were treated with 3% sevoflurane plus 30% oxygen/air or 30% oxygen/air. As the rats grew, the Morris water maze (MWM) and fear conditioning tests were performed to evaluate cognitive function, while the expression of LIMK1 was analyzed by western blot. Luciferase reporter assay was performed to investigate the interaction between LIMK1 and miR-27b. The expression of miR-27b was measured by real-time polymerase chain reaction (PCR) after exposure to sevoflurane. Once the miR-27b inhibitor was transfected into the neurons, the expression of LIMK1 was analyzed by real-time PCR and western blot. Exposure to sevoflurane in neonatal rats induced memory and learning impairments according to the MWM and fear conditioning tests. Sevoflurane increased the expression of miR-27b and reduced the expression of LIMK1 in the brain tissues of rats compared to the control group. The results of the luciferase reporter assay showed that LIMK1 was a direct target of miR-27b. In the primary neurons, the inhibition of miR-27b could reverse the down-regulating effects of sevoflurane on LIMK1 expression. We suggest that sevoflurane-induced learning and memory impairments in rats might be mediated via the miR-27b-LIMK1-signaling pathway.


Assuntos
Anestésicos Inalatórios/toxicidade , Disfunção Cognitiva/induzido quimicamente , Quinases Lim/genética , Éteres Metílicos/toxicidade , MicroRNAs/genética , Animais , Comportamento Animal , Células Cultivadas , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Medo , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Quinases Lim/metabolismo , Masculino , Aprendizagem em Labirinto , Gravidez , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos Sprague-Dawley , Sevoflurano , Transdução de Sinais
3.
Mol Med Rep ; 21(6): 2443-2451, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32236601

RESUMO

Ketamine is a widely used general anesthetic and has been reported to demonstrate neurotoxicity and neuroprotection. Investigation into the regulatory mechanism of ketamine on influencing neural development is of importance for a better and safer way of relieving pain. Reverse transcription­quantitative polymerase chain reaction and western blotting were used to detect the critical neural associated gene expression, and flow cytometry to detect the neural differentiation effect. Hence, in the present study the underlying mechanism of ketamine (50 nM) on neural differentiation of the mouse embryonic stem cell (mESC) line 46C was investigated. The results demonstrated that a low dose of ketamine (50 nM) promoted the differentiation of mESCs to neural stem cells (NSCs) and activated mammalian target of rapamycin (mTOR) by upregulating the expression levels of phosphorylated (p)­mTOR. Furthermore, inhibition of the mTOR signaling pathway by rapamycin or knockdown of mTOR suppressed neural differentiation. A rescue experiment further confirmed that downregulation of mTOR inhibited the promotion of neural differentiation induced by ketamine. Taken together, the present study indicated that a low level of ketamine upregulated p­mTOR expression levels, promoting neural differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Ketamina/farmacologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Fosforilação , Fatores de Transcrição SOXB1 , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Regulação para Cima/efeitos dos fármacos
4.
EBioMedicine ; 43: 473-486, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31060905

RESUMO

BACKGROUND: Exposure to anesthetics during early life may impair cognitive functions. However, the underlying mechanisms remain largely unknown. We set out to determine effects of sevoflurane anesthesia on folate metabolism and myelination in young non-human primates, mice and children. METHODS: Young rhesus macaque and mice received 2.5 to 3% sevoflurane daily for three days. DNA and RNA sequencing and immunohistochemistry among others were used in the studies. We performed unbiased transcriptome profiling in prefrontal cortex of rhesus macaques and mice after the sevoflurane anesthesia. We constructed a brain blood barrier-crossing AAV-PHP.EB vector to harbor ERMN expression in rescue studies. We measured blood folate levels in children after anesthesia and surgery. FINDINGS: We found that thymidylate synthase (TYMS) gene was downregulated after the sevoflurane anesthesia in both rhesus macaque and mice. There was a reduction in blood folate levels in children after the anesthesia and surgery. Combined with transcriptome and genome-wide DNA methylation analysis, we identified that ERMN was the primary target of the disrupted folate metabolism. Myelination was compromised by the anesthesia in the young mice, which was rescued by systematic administration of folic acid or expression of ERMN in the brain through brain-specific delivery of the adeno-associated virus. Moreover, folic acid and expression of ERMN alleviated the cognitive impairment caused by the sevoflurane anesthesia in the mice. INTERPRETATION: General anesthesia leads to disrupted folate metabolism and subsequently defects in myelination in the developmental brain, and ERMN is the important target affected by the anesthesia via epigenetic mechanisms.


Assuntos
Anestesia/efeitos adversos , Biomarcadores , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Epigênese Genética , Ácido Fólico/metabolismo , Regulação da Expressão Gênica , Animais , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Biologia Computacional/métodos , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Ontologia Genética , Genoma , Genômica/métodos , Macaca mulatta , Masculino , Aprendizagem em Labirinto , Camundongos , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Transdução de Sinais , Timidilato Sintase/genética , Timidilato Sintase/metabolismo
5.
Cell Prolif ; 50(5)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28737263

RESUMO

OBJECTIVES: Lidocaine is the most commonly used local anaesthetic in clinical and can inhibit proliferation, suppress invasion and migration and induce apoptosis in human lung adenocarcinoma (LAD) cells. However, its specific downstream molecular mechanism is unclear. MATERIALS AND METHODS: LAD cell lines, A549 and H1299 cells, were treated with lidocaine. The proliferation was evaluated by the methylthiazolyldiphenyl-tetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assay. The expression level of related proteins was detected by real-time quantitative PCR (qPCR) and Western blot assay. RESULTS: The results indicated that lidocaine dose-dependently suppressed the proliferation of A549 and H1299 cells. In the LAD patients' samples, GOLT1A was upregulated and involved in the poor prognosis and higher grade malignancy. Additionally, GOLT1A mediates the function of lidocaine on repressing proliferation by regulating the cell cycle in A549 cells. CONCLUSIONS: Our findings suggest that lidocaine downregulates the GOLT1A expression to repress the proliferation of lung cancer cells.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anestésicos Locais/farmacologia , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lidocaína/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Transporte Vesicular/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA