Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Med ; 30(1): 39, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38493090

RESUMO

OBJECTIVE: Anesthetics have been linked to cognitive alterations, particularly in the elderly. The current research delineates how Fibroblast Growth Factor 2 (Fgf2) modulates tau protein phosphorylation, contributing to cognitive impairments in aged rats upon sevoflurane administration. METHODS: Rats aged 3, 12, and 18 months were subjected to a 2.5% sevoflurane exposure to form a neurotoxicity model. Cognitive performance was gauged, and the GEO database was employed to identify differentially expressed genes (DEGs) in the 18-month-old cohort post sevoflurane exposure. Bioinformatics tools, inclusive of STRING and GeneCards, facilitated detailed analysis. Experimental validations, both in vivo and in vitro, examined Fgf2's effect on tau phosphorylation. RESULTS: Sevoflurane notably altered cognitive behavior in older rats. Out of 128 DEGs discerned, Fgf2 stood out as instrumental in regulating tau protein phosphorylation. Sevoflurane exposure spiked Fgf2 expression in cortical neurons, intensifying tau phosphorylation via the PI3K/AKT/Gsk3b trajectory. Diminishing Fgf2 expression correspondingly curtailed tau phosphorylation, neurofibrillary tangles, and enhanced cognitive capacities in aged rats. CONCLUSION: Sevoflurane elicits a surge in Fgf2 expression in aging rats, directing tau protein phosphorylation through the PI3K/AKT/Gsk3b route, instigating cognitive aberrations.


Assuntos
Anestésicos Inalatórios , Disfunção Cognitiva , Éteres Metílicos , Idoso , Animais , Humanos , Lactente , Ratos , Anestésicos Inalatórios/efeitos adversos , Anestésicos Inalatórios/metabolismo , Cognição , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Éteres Metílicos/farmacologia , Éteres Metílicos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Proteínas tau/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo
2.
Adv Sci (Weinh) ; 11(10): e2305989, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38145349

RESUMO

Lipid metabolism has been considered as a potential therapeutic target in sevoflurane-induced neurotoxicity that can potentially affect the learning and memory function in the developmental brain. Recently, triggering receptor expressed on myeloid cells 2 (TREM2) is identified as a crucial step in regulating lipid metabolism and associated with the pathogenesis of neurodegenerative diseases. Herein, it is reported that quercetin modified Cu2- x Se (abbreviated as CSPQ) nanoparticles can ameliorate sevoflurane-induced neurotoxicity by tuning the microglial lipid metabolism and promoting microglial M2-like polarization via TREM2 signaling pathway, in which the apolipoprotein E (ApoE), and adenosine triphosphate-binding cassette transporters (ABCA1 and ABCG1) levels are upregulated. Furthermore, the protective effects of CSPQ nanoparticles against sevoflurane-induced neurotoxicity via TREM2 are further demonstrated by the small interfering RNA (siRNA)-TREM2 transfected BV2 cells, which are obviously not influenced by CSPQ nanoparticles. The cell membrane coated CSPQ (referred as CSPQ@CM) nanoparticles can significantly reduce sevoflurane-induced learning and memory deficits, improve lipid metabolism dysfunction, and promote the remyelination in the hippocampus of mice. The study shows great potential of targeting microglial lipid metabolism in promoting remyelination of neurons for treatment of neurotoxicity and neurodegenerative diseases.


Assuntos
Microglia , Doenças Neurodegenerativas , Camundongos , Animais , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Microglia/metabolismo , Metabolismo dos Lipídeos , Biomimética , Transdução de Sinais , Doenças Neurodegenerativas/metabolismo
3.
BMC Gastroenterol ; 23(1): 279, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37568083

RESUMO

BACKGROUND: Cholangiocarcinoma (CCA) is a refractory malignancy derived from bile duct epithelial cells. This study aimed to explore the role and molecular mechanisms of action of sevoflurane in CCA. METHODS: CCK-8 assay was used to assess the proliferation of cholangiocarcinoma cells, and flow cytometry was used to detect cholangiocarcinoma cell apoptosis. The effects of sevoflurane on TFK1 and QBC939 cell migration and invasion were investigated using a Transwell assay. Western blotting and RT-qPCR were used to assess the expression of apoptosis-related proteins and genes, and gene expression of the Wnt/ß-catenin signaling pathway. RESULTS: Our study found that sevoflurane inhibited cholangiocarcinoma cell proliferation in a dose-dependent manner. In addition, sevoflurane induced cholangiocarcinoma cell apoptosis, inhibited cholangiocarcinoma cell migration and invasion, as well as the Wnt/ß-catenin signaling pathway evidenced by decreased Wnt3a, ß-catenin, c-Myc, and Cyclin D1 protein and mRNA expression, reduced p-GSK3ß protein expression and p-GSK3ß/GSK3ß ratio. Further mechanistic studies revealed that Wnt/ß-catenin pathway inducer SKL2001 reversed the inhibitory effect of sevoflurane on cholangiocarcinoma cells. CONCLUSIONS: Sevoflurane induces apoptosis and inhibits the growth, migration, and invasion of cholangiocarcinoma cells by inhibiting the Wnt/ß-catenin signaling pathway. This study not only revealed the role of sevoflurane in the development of CCA but also elucidated new therapeutic agents for CCA.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Humanos , Via de Sinalização Wnt/genética , Sevoflurano/farmacologia , Sevoflurano/metabolismo , beta Catenina/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicogênio Sintase Quinase 3 beta/farmacologia , Linhagem Celular Tumoral , Colangiocarcinoma/patologia , Proliferação de Células/genética , Ductos Biliares Intra-Hepáticos , Neoplasias dos Ductos Biliares/patologia , Movimento Celular , Apoptose/genética
4.
Neuropathology ; 43(6): 431-440, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37037475

RESUMO

Sevoflurane (SEV) is a commonly used anesthetic in pediatric surgery. Recent studies reported that repeated use of SEV contributes to cognitive impairment. Engeletin has been discovered to exert anti-inflammatory effects in various diseases. However, the detailed roles and mechanisms of engeletin in SEV-induced cognitive dysfunction of neonatal mice remain unclear. In this study, C57BL/6 neonatal mice were randomly divided into Ctrl, SEV, SEV + Engeletin (10 mg /kg), SEV + Engeletin (20 mg/kg), and SEV + Engeletin (40 mg/kg) groups. The Morris water maze (MWM) test suggested that engeletin treatment significantly improved SEV-induced cognitive impairment in neonatal mice. Employing ELISA and Nissl staining analysis, engeletin reduced neuroinflammation and loss of nerve cells caused by SEV, respectively. The treatment of engeletin dramatically suppressed the activation of microglia and apoptosis induced by SEV in the hippocampus of neonatal mice. Furthermore, the inhibition of PPAR-γ obviously reversed the abovementioned effects of engeletin in the hippocampus of newborn mice. In conclusion, this study verified that engeletin notably ameliorated SEV-induced cognitive deficiencies in neonatal mice at least partially by mediating the expression of PPAR-γ.


Assuntos
Disfunção Cognitiva , Éteres Metílicos , Animais , Camundongos , Animais Recém-Nascidos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Hipocampo , Éteres Metílicos/efeitos adversos , Éteres Metílicos/metabolismo , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo , PPAR gama/farmacologia , Sevoflurano/efeitos adversos , Sevoflurano/metabolismo
5.
Cardiovasc Toxicol ; 23(2): 86-92, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36800141

RESUMO

To investigate the mechanism whereby sevoflurane (Sev) protects cardiomyocytes from hypoxia/reoxygenation (H/R) injury. The rat cardiomyocyte line H9C2 was exposed to hypoxia (1% oxygen) for 24 h, followed by reoxygenation for 2 h to construct a model of H/R injury. H9C2 was exposed to 2.4% Sev for 45 min before creating a hypoxic environment to observe the effect of Sev. MTT was taken to assess the viability of each group of cells, flow cytometry to detect cell apoptosis, and qRT-PCR or western blot to detect the expression of iron metabolism-related proteins and apoptosis-related proteins in the cells. And the kit determined the levels of total Fe and Fe2+ as well as factors related to oxidative stress in the cells. Administration of Sev significantly increased the cell viability of the H/R group while decreasing the expression of apoptosis-related proteins (Bax, cleaved caspase-3). Ferroportin 1 and mitochondrial ferritin, which are associated with iron metabolism, were considerably up-regulated by Sev, while iron regulatory protein 1, divalent metal transporter 1, and transferrin receptor 1 were significantly down-regulated in H/R cells. Additionally, Sev substantially reduced the levels of total Fe and Fe2+, reactive oxygen species, malondialdehyde, and 4-hydroxynonenal in H/R cells. In conclusion, Sev relieves H/R-induced cardiomyocyte injury by regulating iron homeostasis and ferroptosis.


Assuntos
Ferroptose , Miócitos Cardíacos , Animais , Ratos , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Hipóxia , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Homeostase
6.
J Clin Pharmacol ; 63(1): 91-104, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35943164

RESUMO

The susceptibility of different individuals to anesthetics varies widely, and sevoflurane is no exception. We hypothesized that polymorphisms in genes involved in pharmacokinetics and pharmacodynamics may explain this variation. A total of 151 individuals undergoing otorhinolaryngology surgery were included. The influence of genetic polymorphisms on sevoflurane sensitivity were investigated through SNaPshot technology. Individuals carrying KCNK2 rs6686529 G > C, MTRR rs3733784 TT, rs2307116 GG, or rs1801394 AA polymorphisms had a higher sensitivity to the sedative effect of sevoflurane than those without those polymorphisms. The univariate linear regression analysis indicated that MTRR rs3733784 TT, rs2307116 GG, and rs1801394 AA were potentially significant predictors of higher sensitivity to the sedative effect of sevoflurane. Moreover, CYP2E1 rs3813867 G > C and rs2031920 C > T, GABRG1 rs279858 T > C, KCNK3 rs1275988 CC, GRIN2B rs1806201 GG, MTRR rs2307116 G > A, and rs1801394 A > G were associated with a higher sensitivity to the cardiovascular effect of sevoflurane. Our results suggested that 9 single nucleotide polymorphisms in genes involved in metabolizing enzymes, transport proteins, target proteins of sevoflurane and folate metabolism may help to explain individual differences in the susceptibility to the sedative or cardiovascular effect of sevoflurane.


Assuntos
Citocromo P-450 CYP2E1 , Hipnóticos e Sedativos , Polimorfismo de Nucleotídeo Único , Sevoflurano , Humanos , Estudos de Casos e Controles , Citocromo P-450 CYP2E1/metabolismo , Genótipo , Hipnóticos e Sedativos/metabolismo , Hipnóticos e Sedativos/farmacocinética , Sevoflurano/metabolismo , Sevoflurano/farmacocinética
7.
Int Immunopharmacol ; 113(Pt A): 109380, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36330914

RESUMO

Sevoflurane (Sev) is commonly used for cancer surgeries, but its effect on cancer cell biology remains unclear. This study aims to delineate the mechanistic actions underlying the oncogenic and metastatic potential of Sev on macrophage M2 polarization and lung metastasis in prostate cancer (PCa). We performed bioinformatics analysis to predict Sev- and PCa-related targets HO-1 and IL-6, and validated their expression in response to Sev exposure. The culture supernatant of macrophages differentiated from mouse bone marrow was co-cultured with mouse PCa cell line RM-1. HO-1 and IL-6 was found to be upregulated in macrophages exposed to Sev. Ectopic expression or knockdown of HO-1 and IL-6 was introduced to macrophages to probe their effect on macrophage polarization, migratory and invasive capacities of PCa cells and lung metastasis in tumor-bearing mice. It was demonstrated that Sev augmented macrophage M2 polarization and migratory and invasive potential of PCa cells as well as lung metastasis, which was associated with HO-1 upregulation. Mechanistic investigation indicated that Sev elevated IL-6 expression to enhance HO-1 expression. In vivo experiments verified that Sev facilitated macrophage M2 polarization and lung metastasis of RM-1 cells via IL-6/HO-1 in mice. Taken together, our work reveals a novel IL-6/HO-1-mediated regulatory network underlying Sev function that may be a viable target for preventing lung metastasis of PCa.


Assuntos
Neoplasias Pulmonares , Neoplasias da Próstata , Masculino , Camundongos , Animais , Humanos , Sevoflurano/farmacologia , Sevoflurano/metabolismo , Interleucina-6/metabolismo , Neoplasias Pulmonares/patologia , Macrófagos/metabolismo , Neoplasias da Próstata/patologia
8.
Pharm Biol ; 60(1): 1915-1924, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36205592

RESUMO

CONTEXT: Sevoflurane (Sev) is a commonly used surgical anaesthetic; it has neurotoxic effects on the brain. Echinatin (Ech) is reported to have anti-inflammatory and antioxidant activity. OBJECTIVE: This research confirms the effect of Ech on Sev-induced neurotoxicity and cognitive deficits. MATERIALS AND METHODS: Primary rat hippocampal neurons were treated with 4.1% Sev for 6 h in the presence of Ech (5, 10, and 20 µM) or vehicle, followed by a further 42 h of culture. Male Sprague-Dawley aged rats were divided into 6 groups (n = 6): control, Sev, Sev + Ech (20 mg/kg;), Sev + Ech (40 mg/kg), and Sev + Ech (80 mg/kg). Rats were intraperitoneally injected with Ech or vehicle 1 h before Sev exposure (2% Sev for 5 h). RESULTS: We found that Ech (5, 10, and 20 µM) elevated cell viability (1.29-, 1.51-, 1.68-fold) but mitigated apoptosis (23.87% vs. 16.48%, 12.72%, 9.02%), oxidative stress, and ferroptosis in hippocampal neurons with Sev treatment. Ech activated the Nrf2 expression in Sev-induced in vitro and in vivo models of anaesthetic neurotoxicity. Ech also weakened neurotoxicity in hippocampal neurons with Sev treatment by increasing Nrf2 expression level. Moreover, Ech alleviated hippocampus neurons apoptosis (19.38% vs. 16.05%, 11.71%, 8.88%), oxidative stress, and ferroptosis in rats with Sev treatment. Ech improved Sev-induced cognitive deficits in rats. CONCLUSIONS: Ech alleviates Sev-induced neurotoxicity and cognitive deficits by mitigation of ferroptosis and oxidative stress. Ech may be developed as a new promising therapeutic drug for treatment of cerebral nerve injury caused by surgical anaesthesia.


Assuntos
Sobrecarga de Ferro , Síndromes Neurotóxicas , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Apoptose , Chalconas , Cognição , Hipocampo , Sobrecarga de Ferro/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/prevenção & controle , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Sevoflurano/metabolismo , Sevoflurano/toxicidade
9.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(8): 1108-1119, 2022 Aug 28.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-36097779

RESUMO

OBJECTIVES: Myocardial ischemia reperfusion injury (IRI) occurs occasionally in the process of ischemic heart disease. Sevoflurane preconditioning has an effect on attenuating IRI. Preserving the structural and functional integrity of mitochondria is the key to reduce myocardial IRI. Silent information regulator 3 (SIRT3), a class of nicotinamide adenine dinucleotide (NAD+) dependent deacetylases, is an important signal-regulating molecule in mitochondria. This study aims to explore the role of mitochondrial NAD+-SIRT3 pathway in attenuating myocardial IRI in rats by sevoflurane preconditioning. METHODS: A total of 60 male Sprague Dawley (SD) rats were randomly divided into 5 groups (n=12): A sham group (Sham group), an ischemia reperfusion group (IR group), a sevoflurane preconditioning group (Sev group, inhaled 2.5% sevoflurane for 30 min), a sevoflurane preconditioning+SIRT3 inhibitor 3-TYP group (Sev+3-TYP group, inhaled 2.5% sevoflurane for 30 min and received 5 mg/kg 3-TYP), and a 3-TYP group (5 mg/kg 3-TYP). Except for the Sham group, the IR model in the other 4 groups was established by ligating the left anterior descending coronary artery. The size of myocardial infarction was determined by double staining. Serum cardiac troponin I (cTnI) level was measured. The contents of NAD+ and ATP, the activities of mitochondrial complexes I, II, and IV, the content of MDA, the activity of SOD, and the changes of mitochondrial permeability were measured. The protein expression levels of SIRT3, SOD2, catalase (CAT), and voltage dependent anion channel 1 (VDAC1) were detected by Western blotting. The ultrastructure of myocardium was observed under transmission electron microscope. MAP and HR were recorded immediately before ischemia (T0), 30 min after ischemia (T1), 30 min after reperfusion (T2), 60 min after reperfusion (T3), and 120 min after reperfusion (T4). RESULTS: After ischemia reperfusion, the content of NAD+ in cardiac tissues and the expression level of SIRT3 protein were decreased (both P<0.01), and an obvious myocardial injury occurred, including the increase of myocardial infarction size and serum cTnI level (both P<0.01). Correspondingly, the mitochondria also showed obvious damage on energy metabolism, antioxidant function, and structural integrity, which was manifested as: the activities of mitochondrial complexes I, II, and IV, ATP content, protein expression levels of SOD2 and CAT were decreased, while MDA content, VDAC1 protein expression level and mitochondrial permeability were increased (all P<0.01). Compared with the IR group, the content of NAD+ in cardiac tissues and the expression level of SIRT3 protein were increased in the Sev group (both P<0.01); the size of myocardial infarction and the level of serum cTnI were decreased in the Sev group (both P<0.01); the activities of mitochondrial complexes I, II, and IV, ATP content, protein expression levels of SOD2 and CAT were increased, while MDA content, VDAC1 protein expression level, and mitochondrial permeability were decreased in the Sev group (all P<0.01). Compared with the Sev group, the content of NAD+ in cardiac tissues and the expression level of SIRT3 protein were decreased in the Sev+3-TYP group (both P<0.01); the size of myocardial infarction and the level of serum cTnI were increased in the Sev+3-TYP group (both P<0.01); the activities of mitochondrial complexes I, II, and IV, ATP content, protein expression levels of SOD2 and CAT were decreased, while MDA content, VDAC1 protein expression level, and mitochondrial permeability were increased in the Sev+3-TYP group (all P<0.01). CONCLUSIONS: Sevoflurane preconditioning attenuates myocardial IRI through activating the mitochondrial NAD+-SIRT3 pathway to preserve the mitochondrial function.


Assuntos
Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Sirtuína 3 , Trifosfato de Adenosina/metabolismo , Animais , Masculino , Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , NAD/metabolismo , Ratos , Ratos Sprague-Dawley , Sevoflurano/metabolismo , Sirtuína 3/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo
10.
Metab Brain Dis ; 37(7): 2457-2466, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35838869

RESUMO

Sevoflurane, a commonly used anesthetic, has been found to cause neural stem cell (NSC) injury, thereby contributing to neurocognitive impairment following general anesthesia. Tetramethylpyrazine (TMP), one of the most widely used medicinal compounds isolated from a traditional Chinese herb, possess neuroprotective activity. However, its effect on sevoflurane-induced NSC injury remains unclear. NSCs were pretreated with indicated concentrations of TMP for 2 h and then exposed to sevoflurane for 6 h. Cell injury was measured using lactate dehydrogenase (LDH) release assay. Cell viability and proliferation were detected by cell counting kit-8 (CCK-8) assay and 5-bromo-2'-deoxyuridine (BrdU) labeling, respectively. Apoptotic cells were detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The levels of cleaved caspase-3, phosphorylated protein kinase B (Akt) and phosphorylated glycogen synthase kinase-3ß (GSK-3ß) were detected by western blotting. Our results showed exposure to sevoflurane decreased the viability and proliferation of NSCs, while TMP preserved NSC viability and proliferation after sevoflurane exposure. In addition, the expression of cleaved caspase-3 and TUNEL positive cells were markedly decreased in TMP-treated NSCs compared with the control. Furthermore, pretreatment with TMP significantly increased the levels of phosphorylated Akt and GSK-3ß in sevoflurane-injured NSCs. However, an upstream inhibitor of Akt, LY294002 abolished the protective of TMP on the cell viability of NSCs. In conclusion, these findings indicate that TMP protects NSCs from sevoflurane-induced toxicity through Akt/GSK-3ß pathway.


Assuntos
Células-Tronco Neurais , Proteínas Proto-Oncogênicas c-akt , Ratos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Caspase 3/metabolismo , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/farmacologia , DNA Nucleotidilexotransferase/metabolismo , DNA Nucleotidilexotransferase/farmacologia , Transdução de Sinais , Ratos Sprague-Dawley , Células-Tronco Neurais/metabolismo , Lactato Desidrogenases/metabolismo , Apoptose
11.
Aging Cell ; 21(4): e13592, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35299279

RESUMO

Delirium is the most common postoperative complication in older patients after prolonged anesthesia and surgery and is associated with accelerated cognitive decline and dementia. The neuronal pathogenesis of postoperative delirium is largely unknown. The unfolded protein response (UPR) is an adaptive reaction of cells to perturbations in endoplasmic reticulum function. Dysregulation of UPR has been implicated in a variety of diseases including Alzheimer's disease and related dementias. However, whether UPR plays a role in anesthesia-induced cognitive impairment remains unexplored. By performing in vivo calcium imaging in the mouse frontal cortex, we showed that exposure of aged mice to the inhalational anesthetic sevoflurane for 2 hours resulted in a marked elevation of neuronal activity during recovery, which lasted for at least 24 hours after the end of exposure. Concomitantly, sevoflurane anesthesia caused a prolonged increase in phosphorylation of PERK and eIF2α, the markers of UPR activation. Genetic deletion or pharmacological inhibition of PERK prevented neuronal hyperactivity and memory impairment induced by sevoflurane. Moreover, we showed that PERK suppression also reversed various molecular and synaptic changes induced by sevoflurane anesthesia, including alterations of synaptic NMDA receptors, tau protein phosphorylation, and dendritic spine loss. Together, these findings suggest that sevoflurane anesthesia causes abnormal UPR in the aged brain, which contributes to neuronal hyperactivity, synapse loss and cognitive decline in aged mice.


Assuntos
Anestesia , Delírio , Idoso , Animais , Delírio/induzido quimicamente , Delírio/metabolismo , Humanos , Camundongos , Sevoflurano/efeitos adversos , Sevoflurano/metabolismo , Sinapses/metabolismo , Resposta a Proteínas não Dobradas , eIF-2 Quinase/metabolismo
12.
Cell Biol Toxicol ; 38(5): 825-845, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34018092

RESUMO

BACKGROUND: Circular RNA of vimentin (circ-VIM) is a predictor for poor prognosis of acute myeloid leukemia, but we had little information on its function in esophageal cancer (EC). Here we examined the effects of circ-VIM together with sevoflurane on immune escape and multiple oncogenic activities of EC. METHODS: Bioinformatic tools, luciferase assay, and RNA immunoprecipitation were used to examine regulations between circ-VIM, miR-124-3p (miR-124), and PD-L1. CCK-8, wound healing, and Transwell assays were used to measure cell proliferation, migration, and invasion, respectively. The impacts of EC cells on cytotoxicity, proliferation, and apoptosis of CD8+ T cells were examined using LDH assay, CFSE staining, and Annexin V/PI staining, respectively. The in vivo tumorigenesis and lung metastases were assessed using xenograft model and tail vein injection of EC cells. RESULTS: Significant upregulation of circ-VIM and PD-L1 and downregulation of miR-124 were detected in EC tissues or cells. Circ-VIM sponged miR-124 and released its suppression on the downstream target PD-L1. Sevoflurane, independent of circ-VIM, also upregulated miR-124 to lower PD-L1 expression. By modulating miR-124/PD-L1 axis, silencing circ-VIM and applying sevoflurane both inhibited immune escape and multiple oncogenic activities of EC in vitro, and suppressed xenograft growth and lung metastases in vivo. The inactivation of Ras/ERK signaling pathway was involved in suppression of malignant phenotypes by silencing circ-VIM and sevoflurane treatment. CONCLUSIONS: Silencing circ-VIM and applying sevoflurane, by separately regulating miR-124/PD-L1 axis, presented synergistic effects in inhibiting immune escape and multiple malignant phenotypes of EC cells.


Assuntos
Neoplasias Esofágicas , Neoplasias Pulmonares , MicroRNAs , Anexina A5/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Esofágicas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , RNA Circular/genética , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Sincalida/metabolismo , Vimentina/metabolismo
13.
Stem Cell Res Ther ; 12(1): 584, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34809715

RESUMO

BACKGROUND: Ischemic heart diseases is one of the leading causes of death worldwide. Although revascularization timely is an effective therapeutic intervention to salvage the ischemic myocardium, reperfusion itself causes additional myocardial injury called ischemia/reperfusion (I/R) injury. Bone marrow-derived mesenchymal stem cells (MSCs) is one of the promising cells to alleviate ischemic myocardial injury. However, this cell therapy is limited by poor MSCs survival after transplantation. Here, we investigated whether sevoflurane preconditioning could promote MSCs to attenuate myocardial I/R injury via transient receptor potential canonical channel 6 (TRPC6)-induced angiogenesis. METHODS: The anti-apoptotic effect of sevoflurane preconditioning on MSCs was determined by Annexin V-FITC/propidium iodide staining. TRPC6, hypoxia-inducible factor-1α (HIF-1α), Chemokine receptor 4 (CXCR4) and vascular endothelial growth factor (VEGF) protein expressions and VEGF release from MSCs were determined after hypoxia and reoxygenation (H/R). Small interfering RNA (siRNA) was used to knock down TRPC6 gene expression in MSCs. The angiogenesis of human umbilical vein endothelial cells (HUVECs) co-cultured with MSCs was determined by Matrigel tube formation. Myocardial I/R mouse model was induced by occluding left anterior descending coronary artery for 30 min and then reperfusion. MSCs or sevoflurane preconditioned MSCs were injected around the ligature border zone 5 min before reperfusion. Left ventricle systolic function, infarction size, serum LDH, cTnI and inflammatory cytokines were determined after reperfusion. RESULTS: Sevoflurane preconditioning up-regulated TRPC6, HIF-1α, CXCR4 and VEGF expressions in MSCs and VEGF release from MSCs under H/R, which were reversed by knockdown of TRPC6 gene using siRNA in MSCs. Furthermore, sevoflurane preconditioning promoted the angiogenic and anti-inflammatory effect of HUVECs co-cultured with MSCs. Sevoflurane preconditioned MSCs improved left ventricle systolic function and alleviated myocardial infarction and inflammation in mice subjected to I/R insult. CONCLUSION: The current findings reveal that sevoflurane preconditioned MSCs boost angiogenesis in HUVECs subjected to H/R insult and attenuate myocardial I/R injury, which may be mediated by TRPC6 up-regulated HIF-1α, CXCR4 and VEGF.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Traumatismo por Reperfusão Miocárdica , Canais de Potencial de Receptor Transitório , Animais , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/terapia , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Canal de Cátion TRPC6/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Potencial de Receptor Transitório/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Brain Res ; 1767: 147543, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34089702

RESUMO

BACKGROUND: Sevoflurane is a common inhalational anesthetic, which has been revealed to have anticancer effect in glioma. However, the mechanisms of sevoflurane in glioma progression remain largely unclear. METHODS: Cell proliferation, cell cycle, apoptosis and metastasis were monitored by cell counting kit-8 (CCK-8), flow cytometry, Transwell and Western blot assays. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays were used to examine the expression levels of circ_0079593, microRNA (miR)-633 and ROCK1 (Rho Associated Coiled-Coil Containing Protein Kinase 1). The dual-luciferase reporter assay was employed to confirm the targeting relationship between miR-633 and circ_0079593 or ROCK1. Animal experiment was conducted to explore the effect of sevoflurane in vivo. RESULTS: Sevoflurane inhibited glioma cell proliferation, metastasis and induced apoptosis in vitro as well as impeded tumor growth in vivo. The expression of circ_0079593 was higher in glioma tissues and cells, and was decreased by sevoflurane treatment in glioma cells. Functional experiments showed that circ_0079593 overexpression in glioma cells reversed the inhibitory effects of sevoflurane on cell growth and metastasis. In a mechanism analysis, circ_0079593 acted as a sponge for miR-633 to elevate ROCK1 expression in glioma cells, and sevoflurane could regulate ROCK1 expression via circ_0079593/miR-633 axis. Besides that, circ_0079593/miR-633/ROCK1 axis mediated the protective effects of sevoflurane on glioma cell tumorigenesis. CONCLUSION: Sevoflurane repressed glioma tumorigenesis via regulating circ_0079593/miR-633/ROCK1 axis, suggesting a new insight into the application of sevoflurane in glioma therapy.


Assuntos
Glioma/genética , Glioma/metabolismo , Sevoflurano/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Nucleicos Livres/genética , Ácidos Nucleicos Livres/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/tratamento farmacológico , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular , Sevoflurano/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
15.
Commun Biol ; 4(1): 560, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980987

RESUMO

Accumulation and spread of tau in Alzheimer's disease and other tauopathies occur in a prion-like manner. However, the mechanisms and downstream consequences of tau trafficking remain largely unknown. We hypothesized that tau traffics from neurons to microglia via extracellular vesicles (EVs), leading to IL-6 generation and cognitive impairment. We assessed mice and neurons treated with anesthetics sevoflurane and desflurane, and applied nanobeam-sensor technology, an ultrasensitive method, to measure tau/p-tau amounts. Sevoflurane, but not desflurane, increased tau or p-tau amounts in blood, neuron culture medium, or EVs. Sevoflurane increased p-tau amounts in brain interstitial fluid. Microglia from tau knockout mice took up tau and p-tau when treated with sevoflurane-conditioned neuron culture medium, leading to IL-6 generation. Tau phosphorylation inhibitor lithium and EVs generation inhibitor GW4869 attenuated tau trafficking. GW4869 mitigated sevoflurane-induced cognitive impairment in mice. Thus, tau trafficking could occur from neurons to microglia to generate IL-6, leading to cognitive impairment.


Assuntos
Transporte Proteico/efeitos dos fármacos , Sevoflurano/farmacologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Encéfalo/metabolismo , Disfunção Cognitiva/induzido quimicamente , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiologia , Feminino , Hipocampo/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Transporte Proteico/fisiologia , Sevoflurano/metabolismo , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/fisiologia
16.
Life Sci ; 253: 117675, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32360621

RESUMO

AIMS: Gliomas are responsible for the majority of deaths from primary brain tumours. Sevoflurane showed inhibition effects on the tumor progression in vitro. However, whether sevoflurane could affect the stemness of glioma stem cells (GSCs) and the potential molecular mechanism have not been well elucidated. MAIN METHODS: Effects of sevoflurane on cell viability, proliferation and invasion ability of glioma cells as well as tumor growth in vivo were assessed. Sphere formation assay was performed to evaluate the effect of sevoflurane on the stemness of GSCs. Effects of sevoflurane on mitochondrial function was evaluated by intracellular/mitochondrial reactive oxygen species (ROS) level and mitochondrial membrane potential. Expression levels of proliferation-related proteins, stemness markers and proteins in CaMKII/JNK cascade were measured by Western blot. KEY FINDINGS: Sevoflurane inhibited the viability, proliferation and invasion ability of glioma cells (U87MG and U373MG). Western blot showed that sevoflurane decreased the expression levels of proliferation and invasion-related proteins. Sphere formation ability of GSCs, expression levels of stemness markers and mitochondrial function were significantly suppressed by sevoflurane. Moreover, sevoflurane treatment significantly increased the Ca2+ concentration and stimulated phosphorylation of CaMKII, JNK and IRS1. Ca2+ chelator BAPTA-AM combined with sevoflurane synergistically inhibited colony forming ability and the expression levels of proliferation-related proteins and stemness markers. In addition, the in vivo study further confirmed that sevoflurane inhibited tumor growth via Ca2+-dependent CaMKII/JNK cascade. SIGNIFICANCE: The present study demonstrated that sevoflurane inhibited glioma tumorigenesis and modulated the cancer stem cell-like properties and mitochondrial membrane potential via activation of Ca2+-dependent CaMKII/JNK cascade.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , MAP Quinase Quinase 4/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Quelantes/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/metabolismo , Glioma , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
17.
Life Sci ; 239: 116916, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31626792

RESUMO

AIMS: To investigate the effects of sevoflurane on proliferation, cell cycle, apoptosis, autophagy, invasion and epithelial-mesenchymal transition of colon cancer cell line SW480, and to explore its possible mechanism. MATERIALS AND METHODS: SW480 and SW620 cells were treated with a mixture of 95% O2+5% CO2 containing different concentrations of sevoflurane (1.7% SAV, 3.4% SAV and 5.1% SAV) for 6 h. Meanwhile, we performed a rescue experiment by treating cells with the ERK pathway activator LM22B-10 prior to treatment of cells with 5.1% sevoflurane。 KEY FINDINGS: High concentration (5.1%) of sevoflurane significantly inhibited the proliferation and invasion of cells, causing G0/G1 phase arrest and promoted apoptosis and autophagy. 5.1% sevoflurane can participate in the regulation of EMT by regulating the expression of E-cadherin, Vimentin and N-cadherin proteins. LM22B-10 promoted proliferation and invasion of cancer cells and inhibited apoptosis and autophagy, while 5.1% sevoflurane could reverse the effect of LM22B-10 on the biological characteristics of cells. Sevoflurane can significantly inhibit tumor growth in SW480 cells transplanted nude mice. Moreover, 5.1% sevoflurane significantly increased the expression of p-Raf, p-MEK1/2, and p-ERK1/2 in SW480 cells and tumor tissues without affecting p-JNK and p-p38 proteins, meanwhile, 5.1% sevoflurane can inhibit the activation of ERK signaling pathway by LM22B-10 in vitro and in vivo. SIGNIFICANCE: Sevoflurane can inhibit the proliferation and invasion of colon cancer cells, induce apoptosis and autophagy, and participate in the regulation of epithelial-mesenchymal transition, which may be related to its inhibition of the ERK signaling pathway.


Assuntos
Neoplasias do Colo/metabolismo , Sevoflurano/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Caderinas/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , China , Neoplasias do Colo/fisiopatologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sevoflurano/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos
18.
Life Sci ; 221: 143-151, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30763576

RESUMO

The neurotoxicity of anesthetics on developing brain has been a focused issue for years. However, controversy exists between human and animal studies and surgery may be a potential reason for this. The discovery of glymphatic system, a pathway eliminating soluble substance from central nervous system (CNS), together with recent evidence that surgery-induced Aß increase contributes to cognition dysfunction made us rethink about the influence of anesthetics on cognitive function. The function of glymphatic system was proved to be enhanced by sleep and sedation, so we assumed that under clinical situation Aß1-40 whose accumulation played important role in cognitive dysfunction was increased by surgery and eliminated from CNS by glymphatic system. The function of glymphatic system is facilitated by aquaporin-4 (AQP-4), a water channel expressed in highly polarized manor in astrocytic endfeet, whose transcription is regulated by nuclear factor of activated T cells 5 (NFAT5). Our results suggest that under brief operation and sevoflurane exposure, surgery may be the main cause of Aß increase and sevoflurane increase the elimination of Aß by up-regulating AQP-4 which is the key component in glymphatic system.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Aquaporina 4/metabolismo , Fragmentos de Peptídeos/metabolismo , Sevoflurano/metabolismo , Peptídeos beta-Amiloides/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/metabolismo , Sistema Nervoso Central , Disfunção Cognitiva/metabolismo , Sistema Glinfático , Hipocampo/metabolismo , Masculino , Fatores de Transcrição NFATC/metabolismo , Fragmentos de Peptídeos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sevoflurano/farmacologia , Ativação Transcricional , Regulação para Cima
19.
Sci Rep ; 7(1): 5734, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28720769

RESUMO

Membrane proteins are primary targets for most therapeutic indications in cancer and neurological diseases, binding over 50% of all known small molecule drugs. Understanding how such ligands impact membrane proteins requires knowledge on the molecular structure of ligand binding, a reasoning that has driven relentless efforts in drug discovery and translational research. Binding of small ligands appears however highly complex involving interaction to multiple transmembrane protein sites featuring single or multiple occupancy states. Within this scenario, looking for new developments in the field, we investigate the concentration-dependent binding of ligands to multiple saturable sites in membrane proteins. The study relying on docking and free-energy perturbation provides us with an extensive description of the probability density of protein-ligand states that allows for computation of thermodynamic properties of interest. It also provides one- and three-dimensional spatial descriptions for the ligand density across the protein-membrane system which can be of interest for structural purposes. Illustration and discussion of the results are shown for binding of the general anesthetic sevoflurane against Kv1.2, a mammalian ion channel for which experimental data are available.


Assuntos
Anestésicos Inalatórios/química , Anestésicos Inalatórios/metabolismo , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/metabolismo , Sevoflurano/química , Sevoflurano/metabolismo , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA