Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 292
Filtrar
1.
J Gene Med ; 26(5): e3692, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38745073

RESUMO

BACKGROUND: Sevoflurane (Sevo) preconditioning and postconditioning play a protective role against injury induced by hepatic ischemia/reperfusion (I/R). At the same time, the involvement of macrophage infiltration in this process and the precise mechanisms are unclear. Here, we designed this research to elucidate the protective effects of Sevo against hepatic I/R injury and the molecules involved. METHODS: The alleviating effect of Sevo on the liver injury was analyzed by liver function analysis, hematoxylin and eosin staining, Masson trichrome staining, terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling, western blot analysis and an enzyme-linked immunosorbent assay. An in vitro cell model was developed using alpha mouse liver 12 (AML12) cells, and the cell model was treated with oxygen-glucose deprivation and reoxygenation and Sevo. Multiple bioinformatics databases were used to screen transcriptional regulators related to hepatic I/R injury and the targets of Krueppel-like factor 5 (KLF5). KLF5 expression was artificially upregulated alone or with integrin beta-2 (ITGB2) knockdown to substantiate their involvement in Sevo-mediated hepatoprotection. RESULTS: Sevo protected the liver against I/R injury by reducing cell apoptosis and inflammatory response. KLF5 was upregulated in liver tissues following I/R injury, whereas KLF5 overexpression aggravated macrophage infiltration and liver injury induced by I/R injury. KLF5 bound to the promoter of ITGB2 to enhance ITGB2 transcription. Knockdown of ITGB2 reversed the aggravation of injury caused by KLF5 overexpression in mice and AML12 cells. CONCLUSIONS: Sevo blocked KLF5-mediated transcriptional activation of ITGB2, thereby inhibiting macrophage infiltration in hepatic I/R injury.


Assuntos
Fatores de Transcrição Kruppel-Like , Fígado , Macrófagos , Traumatismo por Reperfusão , Sevoflurano , Animais , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/genética , Camundongos , Macrófagos/metabolismo , Sevoflurano/farmacologia , Fígado/metabolismo , Fígado/patologia , Ativação Transcricional , Masculino , Modelos Animais de Doenças , Apoptose , Antígenos CD18/metabolismo , Antígenos CD18/genética , Linhagem Celular , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica
2.
BMC Anesthesiol ; 24(1): 167, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702608

RESUMO

The exact mechanisms and the neural circuits involved in anesthesia induced unconsciousness are still not fully understood. To elucidate them valid animal models are necessary. Since the most commonly used species in neuroscience are mice, we established a murine model for commonly used anesthetics/sedatives and evaluated the epidural electroencephalographic (EEG) patterns during slow anesthesia induction and emergence. Forty-four mice underwent surgery in which we inserted a central venous catheter and implanted nine intracranial electrodes above the prefrontal, motor, sensory, and visual cortex. After at least one week of recovery, mice were anesthetized either by inhalational sevoflurane or intravenous propofol, ketamine, or dexmedetomidine. We evaluated the loss and return of righting reflex (LORR/RORR) and recorded the electrocorticogram. For spectral analysis we focused on the prefrontal and visual cortex. In addition to analyzing the power spectral density at specific time points we evaluated the changes in the spectral power distribution longitudinally. The median time to LORR after start anesthesia ranged from 1080 [1st quartile: 960; 3rd quartile: 1080]s under sevoflurane anesthesia to 1541 [1455; 1890]s with ketamine. Around LORR sevoflurane as well as propofol induced a decrease in the theta/alpha band and an increase in the beta/gamma band. Dexmedetomidine infusion resulted in a shift towards lower frequencies with an increase in the delta range. Ketamine induced stronger activity in the higher frequencies. Our results showed substance-specific changes in EEG patterns during slow anesthesia induction. These patterns were partially identical to previous observations in humans, but also included significant differences, especially in the low frequencies. Our study emphasizes strengths and limitations of murine models in neuroscience and provides an important basis for future studies investigating complex neurophysiological mechanisms.


Assuntos
Anestésicos Inalatórios , Dexmedetomidina , Eletroencefalografia , Ketamina , Propofol , Sevoflurano , Animais , Camundongos , Ketamina/farmacologia , Ketamina/administração & dosagem , Sevoflurano/farmacologia , Sevoflurano/administração & dosagem , Dexmedetomidina/farmacologia , Eletroencefalografia/efeitos dos fármacos , Eletroencefalografia/métodos , Propofol/farmacologia , Propofol/administração & dosagem , Masculino , Anestésicos Inalatórios/farmacologia , Anestésicos Inalatórios/administração & dosagem , Reflexo de Endireitamento/efeitos dos fármacos , Reflexo de Endireitamento/fisiologia , Camundongos Endogâmicos C57BL , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/administração & dosagem , Anestésicos Intravenosos/farmacologia , Anestésicos Intravenosos/administração & dosagem , Anestesia/métodos
3.
Medicine (Baltimore) ; 103(16): e37552, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38640331

RESUMO

Motor-evoked potential (MEP) monitoring is commonly used in children. MEP monitoring in infants is difficult due to smaller signals requiring higher stimulation voltages. There is limited information on the effect of different anesthetics on MEP monitoring in this age group. This case series describes the effect of different anesthetic regimens on MEP monitoring in infants. Patients <1 year of age who underwent spinal surgery with MEP monitoring between February 2022 and July 2023 at a single tertiary care children hospital were reviewed. The motor-evoked potential amplitudes were classified into 4 levels based on the voltage in the upper and lower limbs (none, responded, acceptable, sufficient). "Acceptable" or "sufficient" levels were defined as successful monitoring. A total of 19 infants were identified, involving 3 anesthesia regimens: 4/19 (21.1%) cases were anesthetized with propofol/remifentanil total intravenous anesthesia (TIVA), 3/19 (15.8%) with propofol/remifentanil/low-dose sevoflurane and another 12/19 (63.2%) cases who initially received propofol/remifentanil/sevoflurane and were converted to propofol/remifentanil anesthesia intraoperatively. The 4 cases with propofol/remifentanil showed 20/32 (62.5%) successful monitoring points. In contrast, 6/24 (25%) successful points were achieved with propofol/remifentanil intravenous anesthesia/0.5 age-adjusted minimum alveolar concentration sevoflurane. In 12 cases converted from propofol/remifentanil/low-dose inhalational anesthetics to TIVA alone, successful MEP monitoring points increased from 46/96 (47.9%) to 81/96 (84.4%). Adding low-dose inhalation anesthetic to propofol-based TIVA suppresses MEP amplitudes in infants. The optimal anesthetic regimen for infants requires further investigation.


Assuntos
Anestésicos Inalatórios , Propofol , Criança , Lactente , Humanos , Sevoflurano/farmacologia , Remifentanil , Anestésicos Inalatórios/farmacologia , Potencial Evocado Motor/fisiologia , Anestesia Geral , Anestésicos Intravenosos/farmacologia
4.
Cell Signal ; 119: 111195, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38688381

RESUMO

OBJECTIVE: The specific mechanisms of sevoflurane-induced neurotoxicity are still undetermined. The aim of the current study was to investigate the role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway in sevoflurane-induced neuronal necroptosis. METHODS: BV2 microglial cells were divided into a control group and a 4% sevoflurane exposure group. Western blotting was used to detect expression of the M1 polarization marker inducible nitric oxide synthase (iNOS). RNA was collected for RNA sequencing analysis. After STING knockdown in microglia, western blotting was performed to examine expression of the pro-inflammatory markers CD16 and CD32. The tumor necrosis factor-α (TNF-α) level in media was detected using an enzyme-linked immunosorbent assay. BV2 microglia conditioned media was collected to incubate HT22 neuronal cells, and their cell activity was measured using a CCK8 assay. Calcium was observed by fluorescence. Western blotting was performed to evaluate receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) expression. Neuronal necroptosis rate were detected using flow cytometry. RESULTS: Sevoflurane exposure promoted microglial M1 polarization. The cGAS/STING pathway was screened and identified by RNA sequencing analysis of sevoflurane-exposed microglia and the control group. Compared with the control group, STING knockdown in microglia rescued the amoeboid morphology, inhibited TNF-α release, and significantly decreased iNOS, CD16, and CD32 expression. Moreover, calcium ions and necroptosis within neurons were decreased, and RIPK1, RIPK3, and p-MLKL expression was markedly decreased in microglia media culture with STING knockdown. CONCLUSION: These results suggest that sevoflurane can regulate microglial M1 polarization by activating the cGAS/STING signaling pathway and increasing immune factor release, thus accelerating the neuronal necroptosis induced by calcium overload.


Assuntos
Proteínas de Membrana , Microglia , Necroptose , Neurônios , Nucleotidiltransferases , Sevoflurano , Transdução de Sinais , Microglia/metabolismo , Microglia/efeitos dos fármacos , Animais , Transdução de Sinais/efeitos dos fármacos , Sevoflurano/farmacologia , Camundongos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Necroptose/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Nucleotidiltransferases/metabolismo , Linhagem Celular , Fator de Necrose Tumoral alfa/metabolismo
5.
Exp Cell Res ; 438(1): 114030, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38583855

RESUMO

Acute respiratory distress syndrome (ARDS) is a serious lung condition that often leads to hospitalization in intensive care units and a high mortality rate. Sevoflurane is a volatile anesthetic with growing interest for sedation in ventilated patients with ARDS. It has been shown to have potential lung-protective effects, such as reduced inflammation and lung edema, or improved arterial oxygenation. In this study, we investigated the effects of sevoflurane on lung injury in cultured human carcinoma-derived lung alveolar epithelial (A549) cells. We found that sevoflurane was associated with improved wound healing after exposure to inflammatory cytokines, with preserved cell proliferation but no effect on cell migration properties. Sevoflurane exposure was also associated with enhanced cell viability and active autophagy in A549 cells exposed to cytokines. These findings suggest that sevoflurane may have beneficial effects on lung epithelial injury by promoting alveolar epithelial wound healing and by influencing the survival and proliferation of A549 epithelial cells in vitro. Further research is needed to confirm these findings and to investigate the key cellular mechanisms explaining sevoflurane's potential effects on lung epithelial injury.


Assuntos
Proliferação de Células , Sobrevivência Celular , Síndrome do Desconforto Respiratório , Sevoflurano , Cicatrização , Sevoflurano/farmacologia , Humanos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/patologia , Cicatrização/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células A549 , Proliferação de Células/efeitos dos fármacos , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Movimento Celular/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Citocinas/metabolismo , Autofagia/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia
6.
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
7.
Cell Mol Biol (Noisy-le-grand) ; 70(2): 62-66, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38430040

RESUMO

One of the most prevalent co-operative disorders is postoperative cognitive dysfunction (POCD), however, its pathogenesis remains unclear. Thus, the aim of this work was to evaluate SIRT3's impact on cognitive decline in aged mice under anesthesia. Adeno-associated virus SIRT3 vector (AAV-SIRT3) or empty vector (AAV-VEH) was injected into the hippocampal region of aged mice after sevoflurane induction in order to upregulate the expression of SIRT3. The expression levels of SIRT3, pro-inflammatory cytokines, and apoptotic factors in hippocampus tissues were identified by PCR, Western blotting, TUNEL staining, and enzyme-linked immunosorbent assay (ELISA), and the cognitive function of mice was assessed. The SIRT3 expression was down-regulated in the hippocampal tissue of anesthetized mice. SIRT3 overexpression can improve the learning and memory ability, reduce the escape latency, and increase the residence time in the platform and platform crossing ability of mice. The overexpression of SIRT3 in hippocampus can reduce the oxidative stress response and inflammatory response induced by anesthesia in mice, increase the superoxide dismutase (SOD) expression level, and decrease the expression level of MDA and inflammatory factors in hippocampus. In addition, SIRT3 overexpression can also reduce anesthetic-induced hippocampal cell apoptosis. By reducing the hippocampus mitochondrial oxidative stress response, SIRT3 plays a significant role in the pathophysiology of POCD in mice and is a potential target for POCD treatment and diagnosis.


Assuntos
Disfunção Cognitiva , Complicações Cognitivas Pós-Operatórias , Sirtuína 3 , Animais , Camundongos , Disfunção Cognitiva/genética , Estresse Oxidativo , Complicações Cognitivas Pós-Operatórias/genética , Sevoflurano/efeitos adversos , Sevoflurano/farmacologia , Sirtuína 3/genética , Sirtuína 3/metabolismo
8.
Neuroscience ; 545: 1-15, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38447691

RESUMO

Sevoflurane (Sev) anesthesia is associated with cognitive deficits and neurotoxicity. This study explores the epigenetic mechanism of SET domain containing 1B (SETD1B) in Sev-induced cognitive impairment in neonatal mice. Neonatal mice (C57BL/6, n = 72) were exposed to 3% Sev for 2 h per day at P6, 7, and 8, and the control neonatal mice were only separated from the mother for 2 h. The mice were divided into groups of 12 individuals, with an equal number of male and female mice in each group. Mice were intraperitoneally injected with adenovirus-packaged SETD1B overexpression vector. Behavioral tests (Morris water maze, open field test, T-maze, novel object recognition, etc.) were performed at P30. Mouse hippocampal neuronal cells were cultured in vitro. SETD1B, C-X-C motif chemokine receptor 4 (CXCR4), NLR family pyrin domain containing 1 (NLRP1), Cleaved Caspase1, and GSDMD-N expressions in hippocampal tissues or cells were determined by quantitative real-time polymerase chain reaction and Western blot. SETD1B and histone H3 lysine 4 methylation (H3K4me1, H3K4me2, and H3K4me3) enrichment on the CXCR4 promoter was analyzed by ChIP. Sev insulted cognitive impairment and diminished SETD1B expression in mouse hippocampal tissues. SETD1B overexpression mitigated cognitive impairment, enhanced H3K4me3 levels in hippocampal tissues, and restrained hippocampal neuronal pyroptosis. SETD1B increased CXCR4 expression by elevating the H3K4me3 level on the CXCR4 promoter, thereby curbing NLRP1/Caspase1-mediated hippocampal neuronal pyroptosis. To conclude, SETD1B enhances CXCR4 expression by elevating the H3K4me3 level on the CXCR4 promoter, thereby suppressing NLRP1/Caspase1-triggered hippocampal neuronal pyroptosis and alleviating Sev-induced cognitive impairment in neonatal mice.


Assuntos
Anestésicos Inalatórios , Animais Recém-Nascidos , Disfunção Cognitiva , Epigênese Genética , Hipocampo , Histona-Lisina N-Metiltransferase , Histonas , Camundongos Endogâmicos C57BL , Sevoflurano , Animais , Sevoflurano/farmacologia , Sevoflurano/toxicidade , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Epigênese Genética/efeitos dos fármacos , Camundongos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Anestésicos Inalatórios/toxicidade , Histonas/metabolismo , Feminino , Masculino , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Metilação/efeitos dos fármacos
9.
Mol Med Rep ; 29(3)2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38334145

RESUMO

The aim of the present study was to examine the effect of fullerenol C60 on lung and kidney tissue in sevoflurane­treated rats with lower extremity ischemia­reperfusion (IR) injury. A total of 30 Wistar albino rats weighing 225­275 g were used and were equally divided into five groups (n=6/group): i) Sham; ii) IR; iii) IR­fullerenol C60 (IR­FUL); iv) IR­sevoflurane; and v) IR­fullerenol C60­sevoflurane (IR­FUL­SEVO). Fullerenol C60 was administered intraperitoneally prior to lower extremity IR induction and sevoflurane was administered during the IR injury. Subsequently, lung and kidney histopathological examinations, and serum biochemical analyses were performed. Lung tissue showed markedly increased congestion and neutrophil infiltration in the IR group compared with in the sham group, and notable decreases in congestion and neutrophil infiltration were observed in the treatment groups compared with in the IR group. In the histopathological evaluation of the kidney samples, vacuolization, loss of brush border in tubular epithelial cells, tubular epithelial loss and varying degrees of tubular damage were observed in all groups that underwent IR. There was a significant increase in the mean renal tubule injury score in all IR groups compared with that in the sham group. In addition, the mean kidney injury score was significantly lower in the IR­FUL and IR­FUL­SEVO groups than that in the IR group. It was observed that the expression levels of tumor necrosis factor­α, interleukin 1ß and intercellular adhesion molecule 1 in the lung and kidney tissues were increased following IR, and were decreased in the groups treated with fullerenol C60 and sevoflurane. Notably, it was determined that the reduction in cytokine expression was greatest in the IR­FUL group. When the oxidant status parameters in the lungs and kidneys were examined, thiobarbituric acid reactive substances levels, and catalase and glutathione S­transferase enzyme activities were significantly different in the groups receiving sevoflurane or fullerenol C60 treatment compared with those in the IR group. The present study demonstrated the protective effects of fullerenol C60 on the lung and kidney tissues of rats under sevoflurane anesthesia after establishment of lower extremity IR. The results of the present study showed that fullerenol C60 can reduce oxidative and histopathological damage in the lungs and kidneys following IR of the lower extremities.


Assuntos
Fulerenos , Pulmão , Traumatismo por Reperfusão , Ratos , Animais , Ratos Wistar , Sevoflurano/farmacologia , Pulmão/patologia , Rim/patologia , Traumatismo por Reperfusão/metabolismo , Isquemia/metabolismo , Extremidade Inferior
10.
Tissue Cell ; 86: 102289, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38194851

RESUMO

Hypoxic-ischemic brain damage (HIBD) frequently induces cognitive impairments. Investigating the role of sevoflurane postconditioning (SPC) in HIBD, we conducted experiments involving HIBD modeling, SPC treatment, and interventions with the PERK inhibitor GSK2656157 or the PERK activator CCT020312, administered 30 min before modeling, followed by SPC treatment. Behavioral testing using the Morris water maze test and Neurological Deficiency Scale (NDS) was conducted. Additionally, Nissl staining assessed hippocampal CA1 area neuronal density, TUNEL staining evaluated hippocampal CA1 area neuronal apoptosis, and Western blot determined hippocampal CA1 area protein levels, including Bax, Bcl-2, p-PERK/PERK, p-eIF2/eIF2, ATF4, CHOP, GRP78, Bax, and Bcl-2 protein levels. Following SPC treatment, HIBD rats exhibited improved spatial learning and memory abilities, reduced neuronal apoptosis, increased neuronal density in the hippocampal CA1 area, elevated Bcl-2 protein level, decreased Bax protein levels, and decreased levels of endoplasmic reticulum stress pathway related proteins (p-PERK/PERK, p-eIF2/eIF2, ATF4, CHOP and GRP78). Pre-modeling treatment with the PERK inhibitor treatment improved outcomes in HIBD rats. However, pre-modeling treatment with the PERK activator CCT020312 counteracted the protective effects of SPC against HIBD in rats. In conclusion, SPC alleviates neuronal apoptosis in the hippocampus CA1 area of HIBD rats by inhibiting the endoplasmic reticulum stress pathway PERK/ATF4/CHOP, thereby mitigating HIBD in rats.


Assuntos
Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Hipóxia-Isquemia Encefálica , Sevoflurano , Animais , Ratos , Apoptose , Proteína X Associada a bcl-2/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 2 em Eucariotos/farmacologia , Hipocampo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Sevoflurano/farmacologia
11.
Chem Biol Drug Des ; 103(1): e14362, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37770418

RESUMO

Oral squamous cell carcinoma (OSCC) is a kind of oral malignant tumor with the highest incidence. This study investigated whether sevoflurane (SEV) inhibited OSCC cell progression by regulating circular RNA_0000857 (circ_0000857). OSCC cells were anesthetized with SEV at different concentrations. The expression of circ_0000857 and microRNA-145-5p (miR-145-5p) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability was assayed by the Cell Counting Kit-8 (CCK-8), and cell migration and invasion were examined by the wound-healing assay and transwell. Tube formation assay detected angiogenesis. Western blot was used to detect the expression of related proteins. Compared with the control group, SEV inhibited OSCC cell migration, invasion, and angiogenesis. SEV treatment significantly decreased circ_0000857 expression level, but increased miR-145-5p expression level in SCC4 and HSC3 cells. MiR-145-5p was a target of circ_0000857, and miR-145-5p inhibitor reversed the suppressing effects mediated by circ_0000857 silencing on OSCC cell migration, invasion, and angiogenesis. SEV inhibited the level of matrix metalloproteinases 2 (MMP2), MMP9, and vascular endothelial growth factor A (VEGFA) protein by regulating the circ_0000857/miR-145-5p axis. In all, SEV regulated the migration, invasion, and angiogenesis of OSCC cells through the circ_0000857/miR-145-5p axis, which provided a basis for the potential role of SEV in the treatment of OSCC.


Assuntos
Carcinoma de Células Escamosas , MicroRNAs , Neoplasias Bucais , Humanos , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , RNA Circular/genética , Sevoflurano/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , MicroRNAs/genética , Proliferação de Células , Linhagem Celular Tumoral
12.
J Neurosurg Anesthesiol ; 36(2): 150-158, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-36805419

RESUMO

BACKGROUND: There is debate on the impact of inhalational esthetic agents on transcranial motor evoked potentials (TcMEPs) during intraoperative neuromonitoring. Current guidelines advise their avoidance, which contrasts with common clinical practice. METHODS: This retrospective cohort study of 150 consecutive cervical spine surgeries at a single institution compared stimulation voltages and TcMEP amplitudes in patients who did and did not receive sevoflurane as part of a balanced anesthetic technique. Patients were divided into 3 groups stratified by the presence or absence of increased signal intensity within the cervical spinal cord on T2-weighted magnetic resonance imaging (indicative or myelopathy/spinal cord injury [SCI]) and sevoflurane use. RESULTS: Patients with no magnetic resonance imaging evidence of myelopathy/SCI that received sevoflurane (n=80) had the lowest stimulation voltages and largest TcMEP amplitude responses in the lower extremities compared with those with no magnetic resonance imaging evidence of myelopathy/SCI (n=30). In patients with evidence of myelopathy/SCI who did not receive sevoflurane (n=19), lower extremity TcMEP amplitudes were similar to patients with a myelopathy/SCI that received sevoflurane. Six of these 19 patients had initial low-dose sevoflurane discontinued because of concerns of low/absent baseline TcMEP amplitudes. CONCLUSIONS: Balanced anesthesia with 0.5 MAC sevoflurane in patients with and without radiological evidence of myelopathy/SCI allows reliable TcMEP monitoring. However, in communication with surgical and neuromonitoring teams, it may be advisable in a subset of patients to avoid or discontinue sevoflurane in favor of a propofol/opioid-based anesthetic to ensure adequate and reproducible TcMEPs.


Assuntos
Anestésicos , Doenças da Medula Espinal , Traumatismos da Medula Espinal , Humanos , Sevoflurano/farmacologia , Potencial Evocado Motor/fisiologia , Estudos Retrospectivos , Monitorização Intraoperatória/métodos , Doenças da Medula Espinal/diagnóstico por imagem , Doenças da Medula Espinal/cirurgia , Doenças da Medula Espinal/complicações , Traumatismos da Medula Espinal/etiologia , Vértebras Cervicais/diagnóstico por imagem , Vértebras Cervicais/cirurgia , Anestésicos/farmacologia , Imageamento por Ressonância Magnética
13.
CNS Neurosci Ther ; 30(2): e14406, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37577850

RESUMO

BACKGROUND: Patients undergoing surgical anesthesia increasingly suffer from preoperative depression. Clinical studies have shown that depression is a risk factor for perioperative neurocognitive disorders (PNDs) in elder patients. However, the underlying mechanism, especially at the neural circuit level, remains poorly understood. METHODS: Right carotid artery separation under sevoflurane and chronic social defeat stress (CSDS) in adult mice were used to establish surgical anesthesia and chronic depression models. Cognitive function was assessed by the Y maze and novel object recognition tests. A chemogenetic approach was used to modulate the locus coeruleus-dorsal hippocampal CA1 (LC-dCA1) circuit. Hippocampal synaptic alterations were evaluated by Golgi staining and whole-cell patch clamp recording. RESULTS: We found that CSDS induced synaptic impairments in dorsal hippocampal CA1 pyramidal neurons and cognitive deficits in adult mice after surgery under sevoflurane. Chemogenetic activation of the LC-dCA1 pathway significantly alleviated the CSDS-induced synaptic impairments and cognitive dysfunction. On the contrary, inhibition of this pathway could mimic CSDS-induced deficits. Furthermore, we showed that dopamine played an important role in CSDS-induced PNDs in adult mice after surgery/sevoflurane. CONCLUSION: Overall, our results have demonstrated a vital role for the LC-dCA1 pathway in CSDS-induced PNDs in adult mice undergoing surgery with sevoflurane anesthesia.


Assuntos
Depressão , Locus Cerúleo , Camundongos , Humanos , Animais , Idoso , Sevoflurano/farmacologia , Hipocampo , Transtornos Neurocognitivos , Camundongos Endogâmicos C57BL
14.
CNS Neurosci Ther ; 30(3): e14436, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37736695

RESUMO

BACKGROUND: Postoperative sleep disorder (PSD) and delirium, which may be associated with surgery and inhalational anesthetics, induce adverse effects in old adults. Emerging evidence indicates that circadian rhythm contributes to various neuropathological diseases, including Alzheimer's disease. Thus, we analyzed the potential role of circadian rhythm in PSD and delirium-like behavior in aged mice and determined whether exogenous melatonin could facilitate entrainment of the circadian rhythm after laparotomy under sevoflurane anesthesia. METHODS: We selected old C57BL/6J mice which receiving laparotomy/sevoflurane anesthesia as model animals. We employed buried food, open field, and Y maze test to assess delirium-like behavior, and electroencephalography/electromyography (EEG/EMG) were used to investigate sleep changes. We analyzed the transcription rhythm of clock genes in superchiasmatic nucleus (SCN) to explore the effects of surgery and melatonin pretreatment on the circadian rhythm. Then, we measured melatonin receptor levels in SCN and ERK/CREB pathway-related proteins in hippocampus and prefrontal cortex to assess their role in PSDs and delirium-like behavior. RESULTS: Laparotomy under sevoflurane anesthesia had a greater influence than sevoflurane alone, leading to sleep disorder, a shift in sleep-wake rhythm, and delirium-like behavior. Bmal1, Clock, and Cry1 mRNA expression showed a peak shift, MT1 melatonin receptor expression level was increased in the SCN, and p-ERK/ERK and p-CREB/CREB were decreased in hippocampus and prefrontal cortex of aged mice 1 day after laparotomy. Melatonin showed significant efficacy in ameliorating PSD and delirium-like behavior and restoring the circadian rhythm, reversing melatonin receptor and ERK/CREB pathway expression abnormalities. In addition, most of the beneficial effect of melatonin was antagonized by luzindole, a melatonin receptor antagonist. CONCLUSIONS: Melatonin receptors in SCN, circadian rhythm, and ERK/CREB signaling pathway participate in the pathophysiological processes of PSD and delirium-like behavior. Melatonin intervention could be a potential preventative approach for PSD and delirium.


Assuntos
Delírio , Melatonina , Transtornos do Sono-Vigília , Animais , Camundongos , Melatonina/farmacologia , Melatonina/uso terapêutico , Receptores de Melatonina , Sevoflurano/farmacologia , Camundongos Endogâmicos C57BL , Ritmo Circadiano/fisiologia , Transtornos do Sono-Vigília/tratamento farmacológico , Transtornos do Sono-Vigília/etiologia
15.
CNS Neurosci Ther ; 30(2): e14379, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37545014

RESUMO

BACKGROUND: Cerebral ischemia-hypoxia leads to excitotoxicity-mediated neuronal damage and cognitive dysfunction, especially in the elderly. Excessive intracellular [Cl- ]i accumulation weakens γ-aminobutyric acid (GABA) compensatory effects. Sub-anesthetic dose of propofol protected the brain against ischemia-hypoxia, which was abolished by blocking Cl- efflux transporter K+ /Cl- cotransporter 2 (KCC2). We aimed to determine whether low-dose anesthetic combined with [Cl- ]i regulators could restore the compensatory GABAergic system and improve cognitive function. METHODS: Chronic cerebral hypoxia (CCH) model was established by bilateral carotid artery ligation in aged rats. Sub-dose of anesthetics (propofol and sevoflurane) with or without KCC2 agonist N-ethylmaleimide (NEM) or Na+ /K+ /Cl- cotransporter 1 (NKCC1) antagonist bumetanide (BTN) was administered systemically 30 days post-surgery. Primary rat hippocampal neuronal cultures were subjected to hypoxic injury with or without drug treatment. Memory function, hippocampal neuronal survival, GABAergic system functioning, and brain-derived neurotrophic factor (BDNF) expressions were evaluated. RESULTS: Sub-anesthetic dose of combined propofol (1.2 µg mL-1 ) and sevoflurane [0.7 MAC (minimum alveolar concentration)] did not aggravate the hypoxic brain injury in rats or cell damage in neuronal cultures. Adding either BTN or NEM protected against hypoxic injury, associated with improved cognitive function in vivo, less intracellular accumulation of [Cl- ]i , reduced cell death, restored GABAergic compensation, and increased BDNF expression both in vivo and in vitro. CONCLUSION: Sub-anesthetic dose of propofol and sevoflurane is a recommended anesthesia regimen in at-risk patients. Restoration of [Cl- ]i homeostasis and GABAergic could further reduce the brain damage caused by ischemia-hypoxia.


Assuntos
Anestésicos , Hipóxia-Isquemia Encefálica , Propofol , Humanos , Ratos , Animais , Idoso , Propofol/farmacologia , Propofol/uso terapêutico , Cloretos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sevoflurano/farmacologia , Encéfalo/metabolismo , Bumetanida , Hipóxia/tratamento farmacológico , Cotransportadores de K e Cl- , Isquemia
16.
Cell Signal ; 114: 110987, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38029946

RESUMO

BACKGROUND: Sevoflurane (SEV), a commonly used inhalational anesthetic, reportedly inhibits colorectal cancer (CRC) malignancy, but whether SEV can inhibit the malignancy of CRC by regulating circular RNAs (circRNAs) remains unclear. Therefore, we aimed to identify specific circRNAs that may be affected by SEV and to investigate their functional roles in CRC. METHODS: RT-qPCR was employed to detect the expression of circRNAs and mRNAs in CRC cells and tissues. Fluorescence in situ hybridization (FISH) was used to determine the location of circSKA3. Protein expression was assessed by western blot analysis. Function-based in vitro and in vivo experiments, including CCK-8, colony formation, transwell, and apoptosis assays and mouse xenograft tumor models, were conducted using circSKA3-knockdown and circSKA3-overexpression cell lines. RNA immunoprecipitation, RNA pull-down and mass spectrometry analyses were performed to explore the related mechanism. RESULTS: Our findings revealed that SEV could inhibit CRC cell activity, proliferation and migration and promote apoptosis in CRC cells. We found that circSKA3 was upregulated in CRC and associated with poorer survival and that its expression could be reduced by SEV. The overexpression of circSKA3 reversed the effects of SEV on inhibiting cell activity, proliferation and migration and promoting apoptosis. The mechanistic analysis revealed that circSKA3 could bind to the ARM structural domain of ß-catenin and thereby disrupt its interaction with the CK1/GSK3ß/ß-TrCP1 destruction complex, resulting in the ubiquitinated degradation of ß-catenin and the activation of Wnt/ß-catenin signaling. In addition, SEV downregulated circSKA3 in vivo to inhibit tumor growth. CONCLUSIONS: All the results showed that SEV could inhibit CRC progression via circSKA3 by increasing ß-catenin ubiquitination degradation.


Assuntos
Neoplasias Colorretais , beta Catenina , Humanos , Animais , Camundongos , beta Catenina/metabolismo , Sevoflurano/farmacologia , RNA Circular/genética , Hibridização in Situ Fluorescente , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Ubiquitinação , Proliferação de Células/genética , Linhagem Celular Tumoral , Via de Sinalização Wnt/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética
17.
Eur J Neurosci ; 59(1): 36-53, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37985440

RESUMO

Dexmedetomidine (Dex) may exert neuroprotective effects by attenuating inflammatory responses. However, whether Dex specifically improves postoperative cognitive dysfunction (POCD) by inhibiting microglial inflammation through what pathway remains unclear. In this study, the POCD model was constructed by performing open surgery after 3 h of continuous inhalation of 3% sevoflurane to rats, which were intraperitoneally injected with 25 µg/kg Dex .5 h before anaesthesia. The results displayed that Dex intervention decreased rat escape latency, maintained swimming speed and increased the number of times rats crossed the platform and the time spent in the target quadrant. Furthermore, the rat neuronal injury was restored, alleviated POCD modelling-induced rat hippocampal microglial activation and inhibited microglial M1 type polarization. Besides, we administered Dex injection and/or CCAAT/enhancer-binding protein beta (CEBPB) knockdown on the basis of sevoflurane exposure and open surgery and found that CEBPB was knocked down, resulting in the inability of Dex to function, which confirmed CEBPB as a target for Dex treatment. To sum up, Dex improved POCD by considering CEBPB as a drug target to activate the c-Jun N-terminal kinase (JNK)/p-38 signaling pathway, inhibiting microglial M1 polarization-mediated inflammation in the central nervous system.


Assuntos
Anestesia , Disfunção Cognitiva , Dexmedetomidina , Ratos , Animais , Sevoflurano/farmacologia , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Inflamação/metabolismo
18.
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
19.
Neuroreport ; 35(2): 98-106, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38109363

RESUMO

Diabetes and inhaled anesthesia are associated with an increased likelihood of developing postoperative cognitive dysfunction in humans and animal models, but the mechanisms are unclear. This study aimed to investigate the effect and mechanism of sevoflurane anesthesia on cognitive function in diabetic (DM) mice. Spontaneously diabetic db/db and control db/m mice were subject to sevoflurane anesthesia or allowed to breathe air, respectively. The Morris water maze test as spatial learning and novel object recognition test as recognition memory were performed. The expression of inflammatory cytokines and neurotoxicity-related genes in the hippocampus of four groups was measured using real-time PCR. The expression level of neurotoxicity and neuroprotection-related proteins in DM mice hippocampus were estimated using Western blot assay. It is found that DM mice developed cognitive impairment; however, the cognitive impairment was not exacerbated in sevoflurane-exposed mice. Sevoflurane anesthesia led to a decrease in mRNA levels of inflammatory cytokines in DM mice hippocampi, including interleukin 17 (IL-17), C-C motif chemokine (CCL20), CCL7 as well as high mobility group box 1 and beta-site amyloid-ß cleaving enzyme 1; and no effect was observed on the expression of neurotoxicity genes, including amyloid precursor protein, choline O-acetyltransferase, tumor necrosis factor, alpha-induced protein 1, B-cell lymphoma 2 and estrogen receptor 2. In addition, we observed elevated phosphorylation of cAMP response element-binding protein in DM mice exposed to sevoflurane anesthesia. In conclusion, sevoflurane did not exacerbate DM-associated cognitive impairment.


Assuntos
Anestesia , Diabetes Mellitus Experimental , Síndromes Neurotóxicas , Humanos , Camundongos , Animais , Sevoflurano/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Diabetes Mellitus Experimental/metabolismo , Síndromes Neurotóxicas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Hipocampo/metabolismo
20.
Chin J Physiol ; 66(6): 456-465, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38149558

RESUMO

Recently, evidence has shown that microRNA-100-3p (miR-100-3p) has been revealed as a tumor suppressor in diverse human diseases, while its capability in lung cancer warrants further validation. In this work, we aimed to discuss the impact of sevoflurane on biological functions of lung cancer cells by modulating the miR-100-3p/sterol O-acyltransferase 1 (SOAT1) axis. Lung cancer cell lines (A549 and H460) were treated with various concentrations of sevoflurane. Cell viability, proliferation, migration, and invasion were evaluated using MTT, colony formation, wound healing, and transwell assays. Moreover, miR-100-3p and SOAT1 expressions were evaluated by reverse transcription-quantitative polymerase chain reaction in lung cancer cells. The target interaction between miR-100-3p and SOAT1 was predicted by bioinformatics analysis and verified by the dual-luciferase reporter gene assay. The findings of our work demonstrated that sevoflurane impeded the abilities on viability, proliferation, migration, and invasion of A549 and H460 cells. The expression of miR-100-3p was reduced, and SOAT1 expression was elevated in lung cancer cells. miR-100-3p targeted SOAT1. Besides, sevoflurane could lead to expressed improvement of miR-100-3p or limitation of SOAT1. Downregulation of miR-100-3p or upregulation of SOAT1 restored the suppression of sevoflurane on abilities of viability, proliferation, migration, and invasion in A549 and H460 cells. In the rescue experiment, downregulation of SOAT1 reversed the impacts of downregulation of miR-100-3p on sevoflurane on lung cancer cells. Collectively, our study provides evidence that sevoflurane restrained the proliferation and invasion in lung cancer cells by modulating the miR-100-3p/SOAT1 axis. This article provides a new idea for further study of the pathogenesis of lung cancer.


Assuntos
Antineoplásicos , Neoplasias Pulmonares , Sevoflurano , Sevoflurano/farmacologia , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , MicroRNAs/metabolismo , Esterol O-Aciltransferase/metabolismo , Linhagem Celular Tumoral , Células A549 , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Antineoplásicos/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA