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
País de afiliação
Intervalo de ano de publicação
1.
CNS Neurosci Ther ; 30(2): e14612, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38334030

RESUMO

AIMS: Numerous studies on animals have shown that exposure to general anesthetics in infant stage may cause neurocognitive impairment. However, the exact mechanism is not clear. The dysfunction of iron metabolism can cause neurodevelopmental disorders. Therefore, we investigated the effect of iron metabolism disorder induced by sevoflurane (Sev) on cognitive function and the proliferation of neural precursor cells (NPCs) and neural stem cells (NSCs) in infant mice. METHODS: C57BL/6 mice of postnatal day 14 and neural stem cells NE4C were treated with 2% Sev for 6 h. We used the Morris water maze (MWM) to test the cognitive function of infant mice. The proliferation of NPCs was measured using bromodeoxyuridine (BrdU) label and their markers Ki67 and Pax6 in infant brain tissues 12 h after anesthesia. Meanwhile, we used immunohistochemical stain, immunofluorescence assay, western blot, and flow cytometer to evaluate the myelinogenesis, iron levels, and cell proliferation in cortex and hippocampus or in NE4C cells. RESULTS: The results showed that Sev significantly caused cognitive deficiency in infant mice. Further, we found that Sev inhibited oligodendrocytes proliferation and myelinogenesis by decreasing MBP and CC-1 expression and iron levels. Meanwhile, Sev also induced the iron deficiency in neurons and NSCs by downregulating FtH and FtL expression and upregulating the TfR1 expression in the cortex and hippocampus, which dramatically suppressed the proliferation of NSCs and NPCs as indicated by decreasing the colocalization of Pax6+ and BrdU+ cells, and caused the decrease in the number of neurons. Interestingly, iron supplementation before anesthesia significantly improved iron deficiency in cortex and hippocampus and cognitive deficiency induced by Sev in infant mice. Iron therapy inhibited the decrease of MBP expression, iron levels in neurons and oligodendrocytes, and DNA synthesis of Pax6+ cells in hippocampus induced by Sev. Meanwhile, the number of neurons was partially recovered in hippocampus. CONCLUSION: The results from the present study demonstrated that Sev-induced iron deficiency might be a new mechanism of cognitive impairment caused by inhaled anesthetics in infant mice. Iron supplementation before anesthesia is an effective strategy to prevent cognitive impairment caused by Sev in infants.


Assuntos
Disfunção Cognitiva , Deficiências de Ferro , Células-Tronco Neurais , Humanos , Camundongos , Animais , Sevoflurano/toxicidade , Células-Tronco Neurais/metabolismo , Bromodesoxiuridina/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Proliferação de Células , Ferro/metabolismo , Hipocampo/metabolismo
2.
J Med Food ; 25(5): 557-563, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35420459

RESUMO

Inhalation anesthetics have been shown to cause neurodevelopmental disorders and neurotoxic effects. In this study, we aimed to investigate the effect of resveratrol on the possible neurotoxic effect of sevoflurane and the brain-derived neurotrophic factor (BDNF) pathway in newborn rats. The animals were divided into four groups: control, sevoflurane, sevoflurane+resveratrol 25 mg/kg, and sevoflurane+resveratrol 50 mg/kg. The groups that received anesthesia were given 3% sevoflurane for 2 h on the postnatal seventh, eighth, and ninth days. Control gas was applied to the control group. The Morris water maze (MWM) test was performed on postnatal 35th day. After performing the open field test on the postnatal 41st day, the animals were dissected, and the hippocampal BDNF levels were determined by Western blot method. In the MWM test, there was a significant decrease in the time spent in the target quadrant in the sevoflurane anesthesia group compared with control group. This reduction was reversed with the resveratrol pretreatment. Sevoflurane exposure significantly decreased hippocampal BDNF levels compared with the control group. The resveratrol 25 mg/kg pretreatment did not reverse this reduction, whereas resveratrol 50 mg/kg ameliorated this impairment. Sevoflurane did not cause any significant difference in the rats' performance in the open field test. However, 50 mg/kg resveratrol pretreatment caused a statistically significant increase in this performance. Our results showed that sevoflurane impaired learning and memory functions in newborn rats and resveratrol reversed this deterioration. Also BDNF might play a role in this beneficial effect of resveratrol.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Hipocampo , Resveratrol , Sevoflurano , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/efeitos dos fármacos , Aprendizagem em Labirinto , Ratos , Ratos Sprague-Dawley , Resveratrol/farmacologia , Sevoflurano/toxicidade
3.
ACS Chem Neurosci ; 12(18): 3314-3322, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34445868

RESUMO

Luteolin is a flavone compound occurring in a variety of medicinal plants, which is reported to have neuroprotective properties. In this study, we aimed to explore the effects of luteolin in alleviating sevoflurane-induced neurotoxicity. GeneCards and Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform were employed to screen luteolin, sevoflurane, and neurotoxicity-related genes. Subsequently, we isolated primary neurons from the hippocampus of 1-day-old C57BL/6J mice and tested for cytotoxicity after treatment of different concentrations of luteolin. Next, we measured the expression of apoptosis by flow cytometry and assessed inflammation-related factors, including heme oxygenase-1 expression detected by immunohistochemical staining and neuronal apoptosis. Finally, water maze, open field, and fear conditioning tests were conducted to observe the interaction between luteolin and sevoflurane in cognitive impairment of mice. Luteolin had the lowest cytotoxicity at concentrations of 30 or 60 µg/mL; we selected 30 µg/mL for drug administration experiments in vitro. Luteolin inhibited sevoflurane-induced neuronal apoptosis and inflammatory responses through the autophagic pathway and thus ameliorated sevoflurane-induced cognitive impairment in mice. Mechanistically, luteolin up-regulated heme oxygenase-1 expression, which activated the autophagy pathway in vitro. This was confirmed by subsequent histological experiments in mice and behavioral results showing rescue cognitive impairment. Our findings uncovered an inhibitory role of luteolin in sevoflurane-induced neuronal apoptosis and inflammatory response through activation of autophagy arising from up-regulation of heme oxygenase-1, thereby alleviating sevoflurane-induced cognitive impairment in mice.


Assuntos
Heme Oxigenase-1 , Luteolina , Animais , Apoptose , Autofagia , Hipocampo , Luteolina/farmacologia , Aprendizagem em Labirinto , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Sevoflurano/toxicidade
4.
Am J Chin Med ; 48(4): 945-966, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32476431

RESUMO

Tetramethylpyrazine has shown neuroprotective and axonal outgrowth-promoting effects and can improve cognitive deficit in a rat model of chronic hypoperfusion. However, the role of tetramethylpyrazine in sevoflurane-induced neurotoxicity is still vague. Therefore, this study was designed to investigate the effects and mechanisms of tetramethylpyrazine on sevoflurane-induced autophagy, apoptosis, and the expression of BACE1 and A[Formula: see text] in SH-SY5Y cells. We measured the expression levels of the apoptosis protein markers Bax and Bcl-2, autophagy protein markers Atg5 and LC3-II, BACE1, and A[Formula: see text] in SH-SY5Y cells after sevoflurane treatment and determined the effects of tetramethylpyrazine on sevoflurane-induced expression of these proteins after silencing GPR50 or Atg5 with siRNA in vitro. We found that exposure to 3.4% sevoflurane for 6 h decreased the expression of autophagy protein markers and increased the expression of the apoptosis protein markers, BACE1, and A[Formula: see text] in SH-SY5Y cells. The number of red puncta (autolysosomes) and yellow puncta (autophagosomes) in each SH-SY5Y cell decreased after transient transfection with the mRFP-GFP-LC3 expression plasmid. Silencing of GPR50 decreased the expression of pCREB, Atg5, and LC3-II, while silencing of Atg5 increased the expression of BACE1 and A[Formula: see text] in SH-SY5Y cells. Our results demonstrate that tetramethylpyrazine attenuated sevoflurane-induced neurotoxicity by enhancing autophagy through the GPR50/CREB pathway in SH-SY5Y cells.


Assuntos
Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína de Ligação a CREB/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroprostanos , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Receptores Acoplados a Proteínas G/metabolismo , Sevoflurano/toxicidade , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Células Tumorais Cultivadas
5.
Neurochem Int ; 135: 104693, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32035889

RESUMO

Maternal anesthetic exposure during pregnancy is associated with an increased risk of cognitive impairment in offspring. The balance of cerebral iron metabolism is essential for the development of brain tissue. Iron deficiency affects the myelinogenesis and nerve tissue development, especially in fetus or infant, which has a key role in cognitive function. We aimed to investigate whether maternal sevoflurane (Sev) exposure caused cognitive impairment in offspring through inducing iron deficiency and inhibiting myelinogenesis. Pregnant mice (gestation stage day 14) were treated with 2% Sev for 6 h. Cognitive function of offspring mice was determined by the Morris water maze and Context fear conditioning test. Iron levels were assayed by Perl's iron staining and synchrotron imaging. Hippocampus and cortex tissues or cerebral microvascular endothelial cells of offspring mice (postnatal day 35) were harvested and subjected to Western blot and/or immunhistochemistry to assess ferritin, transferrin receptor 1(TfR1), Ferroportin-1 (FpN1), myelin basic protein (MBP), tight junction protein ZO-1, occludin, and claudin-5 levels. Beginning with postnatal day 30, the offspring were treated with iron therapy for 30 days, and the indicators above were tested. Our results showed Sev dramatically decreased the iron levels of brain and impaired cognitive function in offspring mice. Sev decreased the expression of heavy chain ferritin (FtH), light chain ferritin (FtL), MBP, ZO-1, occludin, claudin-5, and FpN1, and increased TfR1 in hippocampus and cortex or cerebral microvascular endothelial cells of offspring mice, indicating that Sev caused the iron deficiency and impaired the myelinogenesis in the brain of offspring. Interestingly, iron therapy prompted the myelinogenesis and improved impaired cognitive function at postnatal day 60. Our research uncovered a new mechanism which showed that iron deficiency induced by Sev and myelin formation disorder due to decreased iron of brain may be an important risk factor for cognitive impairment in offspring. It was necessary for offspring to be supplied iron supplement whose mother suffered exposure to sevoflurane during pregnancy.


Assuntos
Anemia Ferropriva/induzido quimicamente , Anestésicos Inalatórios/toxicidade , Disfunção Cognitiva/induzido quimicamente , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Sevoflurano/toxicidade , Administração por Inalação , Anemia Ferropriva/metabolismo , Anemia Ferropriva/patologia , Anestésicos Inalatórios/administração & dosagem , Animais , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Sevoflurano/administração & dosagem
6.
J Cell Mol Med ; 24(2): 1345-1359, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31802591

RESUMO

This study aimed to investigate the protective effects and underlying mechanisms of cistanche on sevoflurane-induced aged cognitive dysfunction rat model. Aged (24 months) male SD rats were randomly assigned to four groups: control group, sevoflurane group, control + cistanche and sevoflurane + cistanche group. Subsequently, inflammatory cytokine levels were measured by ELISA, and the cognitive dysfunction of rats was evaluated by water maze test, open-field test and the fear conditioning test. Three days following anaesthesia, the rats were killed and hippocampus was harvested for the analysis of relative biomolecules. The oxidative stress level was indicated as nitrite and MDA concentration, along with the SOD and CAT activity. Finally, PPAR-γ antagonist was used to explore the mechanism of cistanche in vivo. The results showed that after inhaling the sevoflurane, 24- but not 3-month-old male SD rats developed obvious cognitive impairments in the behaviour test 3 days after anaesthesia. Intraperitoneal injection of cistanche at the dose of 50 mg/kg for 3 consecutive days before anaesthesia alleviated the sevoflurane-induced elevation of neuroinflammation levels and significantly attenuated the hippocampus-dependent memory impairments in 24-month-old rats. Cistanche also reduced the oxidative stress by decreasing nitrite and MDA while increasing the SOD and CAT activity. Moreover, such treatment also inhibited the activation of microglia. In addition, we demonstrated that PPAR-γ inhibition conversely alleviated cistanche-induced protective effect. Taken together, we demonstrated that cistanche can exert antioxidant, anti-inflammatory, anti-apoptosis and anti-activation of microglia effects on the development of sevoflurane-induced cognitive dysfunction by activating PPAR-γ signalling.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Cistanche/química , Disfunção Cognitiva/tratamento farmacológico , PPAR gama/metabolismo , Extratos Vegetais/farmacologia , Sevoflurano/toxicidade , Animais , Apoptose , Comportamento Animal , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Masculino , Estresse Oxidativo , PPAR gama/genética , Inibidores da Agregação Plaquetária/toxicidade , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA