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.
Mol Med Rep ; 22(2): 1391-1399, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32626997

RESUMO

Postoperative cognitive dysfunction (POCD) is a prevalent neurocognitive disorder following surgery and anesthesia, particularly in elderly patients. Isoflurane is a widely used anesthetic agent, which is associated with the development of POCD; however, the precise mechanisms remain unclear. In the present study, aged rats were exposed to 2% isoflurane to establish a POCD model. The expression of PYRIN­containing Apaf1­like protein 1 (PYPAF1) was knocked down using a lentivirus containing specific short hairpin RNA. Subsequently, the spatial learning ability of rats was assessed using the Morris water maze. In addition, mRNA and protein expression levels were detected using reverse transcription­quantitative PCR and western blot analysis, respectively. Immunofluorescence double staining was also used to determine the expression of PYPAF1 and Iba­1 in the hippocampus. Neural apoptosis was observed using TUNEL­NeuN double staining. The results revealed that isoflurane exposure impaired the spatial learning ability of rats, while PYPAF1 knockdown alleviated cognitive impairment. In addition, isoflurane exposure induced activation of the PYPAF1 inflammasome, as evidenced by elevated expression of PYPAF1 and apoptosis­associated speck­like protein containing a caspase recruitment domain, while silencing of PYPAF1 partially reversed this effect. Furthermore, isoflurane exposure promoted the activation of microglia and caspase­1, and the secretion of interleukin (IL)­1ß and IL­18, all of which were alleviated following PYPAF1 silencing. Moreover, isoflurane exposure induced neuronal apoptosis, elevated the levels of Bax and cleaved caspase­3, and inhibited the expression of Bcl­2; all of these effects were partially abrogated following PYPAF1 silencing. In conclusion, the results of the present study indicated that PYPAF1 silencing partially abolished isoflurane­induced cognitive impairment, neuroinflammation and neuronal apoptosis. Therefore, PYPAF1 may be a potential therapeutic target for treatment of POCD.


Assuntos
Hipocampo/metabolismo , Inflamação/metabolismo , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Complicações Cognitivas Pós-Operatórias/metabolismo , Envelhecimento , Anestésicos Inalatórios/efeitos adversos , Animais , Modelos Animais de Doenças , Inativação Gênica , Hipocampo/patologia , Isoflurano/efeitos adversos , Masculino , Aprendizagem em Labirinto , Microglia/patologia , Complicações Cognitivas Pós-Operatórias/induzido quimicamente , Ratos , Ratos Sprague-Dawley
3.
Anal Chem ; 91(9): 5762-5767, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30957481

RESUMO

Glutathione (GSH) can help the body maintain the function of the normal immune system and its level change is associated with a variety of diseases. To achieve the ultrasensitive assay of GSH, a "switch on" nanosensor is designed on the basis of GSH regulating the inner filter effect (IFE) of MnO2 nanosheets (MnO2 NS) on boron nitride quantum dots (BNQDs). Here, the fluorescence of BNQDs is quenched efficiently in the presence of redoxable MnO2 NS because of the superior light absorption capability; however, the introduction of GSH can trigger the decomposition of MnO2 to Mn2+ and weaken the IFE, causing the partial fluorescence recovery. The recovered fluorescence is dependent on the concentration of GSH. Under the optimal conditions, this sensing platform shows the response to GSH in the range of 0.5-250 µM with the detection limit of 160 nM. On the basis of the GSH activated reduction of MnO2 NS, the MnO2 NS/BNQDs nanoprobes exhibit good selectivity to GSH. The practical application of the proposed system is demonstrated by detecting the GSH in human plasma samples with satisfying results.


Assuntos
Técnicas Biossensoriais/métodos , Compostos de Boro/química , Fluorescência , Glutationa/sangue , Compostos de Manganês/química , Pontos Quânticos , Bioensaio , Corantes Fluorescentes , Glutationa/química , Humanos , Limite de Detecção
4.
Biochem Biophys Res Commun ; 503(1): 215-221, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-29879429

RESUMO

Ischemia/reperfusion (IR) injury causes damage in aerobically metabolizing organs or tissues, which is an essential injury mechanism in various clinical settings. SRY-related high mobility group-Box gene 9 (SOX9) is a transcription factor of the SRY family, modulating various cellular processes, including fibrosis formation and tumor growth. However, the effects of SOX9 on hepatic IR injury have not been explored. In the present study, a hepatic IR injury model was established, supported by a significant histological alteration with high Suzuki scores, and a remarkable up-regulation of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Importantly, we found that SOX9 was over-expressed in liver of mice after IR operation. Suppressing SOX9 markedly reduced inflammatory response, as evidenced by the reduced mRNA expressions of tumor necrosis factor α (TNF-α), interleukin (IL)-6 and IL-1ß and inactivation of inhibitor of κBα (IκBα)/nuclear factor (NF)-κB pathway. In addition, SOX9 suppression alleviated apoptosis in liver of mice after IR injury, as supported by the reduced number of terminal deoxyribonucleotidyl transferse (TdT)-mediated biotin-16-dUTP nick-end labelling (TUNEL)-staining cells and decreased expression of Caspase-3 in liver tissue sections. The role of SOX9 in accelerating hepatic IR injury was further confirmed in primary hepatocytes under hypoxiaand reoxygenation (HR) treatment by enhancing inflammatory response and apoptosis. Of note, we found that transforming growth factor (TGF)-ß1 was highly induced in liver of mice after IR injury. HR treatment also stimulated TGF-ß1 expressions in vitro. Significantly, SOX9 over-expression-induced inflammation and apoptosis were obviously reduced by pirfenidone (Pirf), TGF-ß1 inhibitor. In contrast, TGF-ß1 exposure to cells further enhanced inflammation and apoptosis in HR-operated cells either with SOX9 knockdown or over-expression. Therefore, we identified a novel SOX9-dependent pathway that contributed to hepatic IR injury through enhancing inflammation and apoptosis by activating TGF-ß1.


Assuntos
Fígado/lesões , Traumatismo por Reperfusão/etiologia , Fatores de Transcrição SOX9/genética , Fator de Crescimento Transformador beta1/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Células Cultivadas , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Fatores de Transcrição SOX9/antagonistas & inibidores , Fatores de Transcrição SOX9/metabolismo , Fator de Crescimento Transformador beta1/genética , Regulação para Cima
5.
Cell Biol Int ; 39(8): 933-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25808510

RESUMO

Local anesthetic of bupivacaine may inhibit neurite outgrowth and induce apoptosis in mouse dorsal root ganglia (DRG) neurons. In this work, we intended to investigate the functional role of microRNA 26a (miR-26a) in regulating bupivacaine-induced nerve injury in DRG neurons. DRG neurons were extracted from C57BL/6 mice and cultured in vitro. Bupivacaine was applied in vitro and it induced apoptosis, inhibited neurite growth, and significantly down-regulated miR-26a gene in DRG neurons. MiR-26a mimic was then used to up-regulate miR-26a expression in DRG neurons. We found that miR-26a up-regulation promoted neurite outgrowth and reduced apoptosis in bupivacaine-injured DRG neurons. Luciferase assay and Western blot confirmed that Phosphatase and tensin homolog (PTEN) was down-stream target of miR-26a in DRG neurons. Ectopic PTEN up-regulation was then able to reverse the protective effect of miR-26a overexpression on bupivacaine-induced nerve injury in DRG neurons. Overall, this work demonstrated that miR-26a had a functional role in regulating bupivacaine-induced nerve injury in DRG neurons. Up-regulating miR-26a to suppress PTEN signaling pathway may be an effective method to protect local anesthetic-induced nerve injury in spinal cord.


Assuntos
Bupivacaína/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , PTEN Fosfo-Hidrolase/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA