Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 390
Filtrar
1.
Mol Biotechnol ; 66(1): 34-43, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36997697

RESUMO

An increasing number of studies reveal the deleterious effects of isoflurane (Iso) exposure during pregnancy on offspring cognition. However, no effective therapeutic strategy for Iso-induced deleterious effects has been well developed. Angelicin exerts an anti-inflammatory effect on neurons and glial cells. This study investigated the roles and mechanism of action of angelicin in Iso-induced neurotoxicity in vitro and in vivo. After exposing C57BL/6 J mice on embryonic day 15 (E15) to Iso for 3 and 6 h, respectively, neonatal mice on embryonic day 18 (E18) displayed obvious anesthetic neurotoxicity, which was revealed by the elevation of cerebral inflammatory factors and blood-brain barrier (BBB) permeability and cognitive dysfunction in mice. Angelicin treatment could not only significantly reduce the Iso-induced embryonic inflammation and BBB disruption but also improve the cognitive dysfunction of offspring mice. Iso exposure resulted in an increase of carbonic anhydrase (CA) 4 and aquaporin-4 (AQP4) expression at both mRNA and protein levels in vascular endothelial cells and mouse brain tissue collected from neonatal mice on E18. Remarkably, the Iso-induced upregulation of CA4 and AQP4 expression could be partially reversed by angelicin treatment. Moreover, GSK1016790A, an AQP4 agonist, was used to confirm the role of AQP4 in the protective effect of angelicin. Results showed that GSK1016790A abolished the therapeutic effect of angelicin on Iso-induced inflammation and BBB disruption in the embryonic brain and on the cognitive function of offspring mice. In conclusion, angelicin may serve as a potential therapeutic for Iso-induced neurotoxicity in neonatal mice by regulating the CA4/AQP4 pathway.


Assuntos
Disfunção Cognitiva , Furocumarinas , Isoflurano , Leucina/análogos & derivados , Sulfonamidas , Gravidez , Feminino , Camundongos , Animais , Isoflurano/toxicidade , Anidrase Carbônica IV/metabolismo , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL , Aquaporina 4/genética , Aquaporina 4/metabolismo , Furocumarinas/efeitos adversos , Inflamação , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/psicologia , Cognição
2.
Toxicol Mech Methods ; 34(3): 319-333, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38054380

RESUMO

PURPOSE: This study sought to identify drug target genes and their associated molecular mechanisms during isoflurane-induced anesthesia in clinical applications. METHODS: Microarray data (ID: GSE64617; isoflurane-treated vs. normal samples) were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened and hub genes were investigated using weighted correlation network analysis (WGCNA). Protein-protein interactions (PPIs) were constructed among the co-DEGs (common genes between DEGs and hub genes), followed by functional enrichment analyses. Then, three machine learning methods were used to reveal drug targets, followed by validation, nomogram analysis, and gene set enrichment analysis. Finally, an miRNA-target network was constructed. RESULTS: A total of 686 DEGs were identified between the two groups-of which, 183 DEGs integrated with genes revealed by WCGNA were identified as co-genes. These genes, including contactin-associated protein 1 (CNTNAP1), are mainly involved in functions such as action potentials. PPI network analysis revealed three models, with the machine learning analysis exploring four drug target genes: A2H, FAM155B, SCARF2, and SDR16C5. ROC and nomogram analyses demonstrated the ideal diagnostic value of these target genes. Finally, miRNA-mRNA pairs were constructed based on the four mRNAs and associated 174 miRNAs. CONCLUSION: FA2H, FAM155B, SCARF2, and SDR16C5 may be novel drug target genes for isoflurane-induced anesthesia. CNTNAP1 may participate in the progression of isoflurane-induced anesthesia via its action potential function.


Assuntos
Anestesia , Isoflurano , MicroRNAs , Isoflurano/toxicidade , Bases de Dados Factuais , Aprendizado de Máquina , MicroRNAs/genética , RNA Mensageiro
3.
Neurochem Res ; 49(3): 706-717, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38055149

RESUMO

Isoflurane, a widely used inhalation anesthetic in clinical practice, is associated with an increased risk of neuronal injury. Heat shock protein 90 (HSP90) plays a crucial role in maintaining neuronal homeostasis under stress conditions; however, its role during isoflurane exposure remains poorly understood. In this study, we aimed to investigate the protective effects of HSP90 inhibition and explore the regulatory mechanisms underlying these effects during isoflurane exposure. We found that the HSP90 inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17 AAG) has great protective effects in mitigating isoflurane-induced ferroptosis of mouse hippocampus and cultured neuronal cells. We focused on the activity of the crucial protein GPX4 in ferroptosis and found that 17 AAG exerted protective effects, preserving the physiological GPX4 activity under isoflurane exposure; further, 17 AAG restored the protein level of GPX4. Further, we observed that the chaperone-mediated autophagy (CMA) pathway was activated; 17 AAG also mediated GPX4 degradation under isoflurane exposure. Additionally, it interfered with the formation of complexes between HSP90 and Lamp-2a, inhibiting CMA activity, followed by the blockade of GPX4 degradation, further affecting the isoflurane-induced ferroptosis. Based on these findings, we proposed HSP90 inhibition as a protective mechanism against isoflurane-induced ferroptosis in neurons.


Assuntos
Antineoplásicos , Isoflurano , Lactamas Macrocíclicas , Humanos , Animais , Camundongos , Isoflurano/toxicidade , Proteínas de Choque Térmico HSP90/metabolismo , Benzoquinonas/farmacologia , Benzoquinonas/uso terapêutico , Antineoplásicos/farmacologia
4.
Anesthesiology ; 140(3): 463-482, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38118175

RESUMO

BACKGROUND: Carriers of mutations in the mitochondrial electron transport chain are at increased risk of anesthetic-induced neurotoxicity. To investigate the neurotoxicity mechanism and to test preconditioning as a protective strategy, this study used a Drosophila melanogaster model of Leigh syndrome. Model flies carried a mutation in ND23 (ND2360114) that encodes a mitochondrial electron transport chain complex I subunit. This study investigated why ND2360114 mutants become susceptible to lethal, oxygen-modulated neurotoxicity within 24 h of exposure to isoflurane but not sevoflurane. METHODS: This study used transcriptomics and quantitative real-time reverse transcription polymerase chain reaction to identify genes that are differentially expressed in ND2360114 but not wild-type fly heads at 30 min after exposure to high- versus low-toxicity conditions. This study also subjected ND2360114 flies to diverse stressors before isoflurane exposure to test whether isoflurane toxicity could be diminished by preconditioning. RESULTS: The ND2360114 mutation had a greater effect on isoflurane- than sevoflurane-mediated changes in gene expression. Isoflurane and sevoflurane did not affect expression of heat shock protein (Hsp) genes (Hsp22, Hsp27, and Hsp68) in wild-type flies, but isoflurane substantially increased expression of these genes in ND2360114 mutant flies. Furthermore, isoflurane and sevoflurane induced expression of oxidative (GstD1 and GstD2) and xenobiotic (Cyp6a8 and Cyp6a14) stress genes to a similar extent in wild-type flies, but the effect of isoflurane was largely reduced in ND2360114 flies. In addition, activating stress response pathways by pre-exposure to anesthetics, heat shock, hyperoxia, hypoxia, or oxidative stress did not suppress isoflurane-induced toxicity in ND2360114 mutant flies. CONCLUSIONS: Mutation of a mitochondrial electron transport chain complex I subunit generates differential effects of isoflurane and sevoflurane on gene expression that may underlie their differential effects on neurotoxicity. Additionally, the mutation produces resistance to preconditioning by stresses that protect the brain in other contexts. Therefore, complex I activity modifies molecular and physiologic effects of anesthetics in an anesthetic-specific manner.


Assuntos
Anestésicos Inalatórios , Isoflurano , Éteres Metílicos , Animais , Isoflurano/toxicidade , Sevoflurano/farmacologia , Anestésicos Inalatórios/toxicidade , Drosophila melanogaster/genética , Estresse Oxidativo , Complexo I de Transporte de Elétrons/genética , Éteres Metílicos/farmacologia
5.
J Toxicol Sci ; 48(12): 645-654, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38044126

RESUMO

Antiparasitic ivermectin has been reported to induce cardiovascular adverse events, including orthostatic hypotension, tachycardia and cardiopulmonary arrest, of which the underlying pathophysiology remains unknown. Since its drug repurposing as an antiviral agent is underway at higher doses than those for antiparasitic, we evaluated the cardiovascular safety pharmacology of ivermectin using isoflurane-anesthetized beagle dogs (n=4). Ivermectin in doses of 0.1 followed by 1 mg/kg was intravenously infused over 10 min with an interval of 20 min, attaining peak plasma concentrations of 0.94 ± 0.04 and 8.82 ± 1.25 µg/mL, which were 29-31 and 276-288 times higher than those observed after its antiparasitic oral dose of 12 mg/body, respectively. The latter peak concentration was > 2 times greater than those inhibiting proliferation of dengue virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and hepatitis B virus in vitro. Ivermectin decreased heart rate without altering mean blood pressure, suggesting that ivermectin does not cause hypotension or tachycardia directly. Ivermectin hardly altered atrioventricular nodal or intraventricular conduction, indicating a lack of inhibitory action on Ca2+ or Na+ channel in vivo. Ivermectin prolonged QT interval/QTcV in a dose-related manner and tended to slow the repolarization speed in a reverse frequency-dependent manner, supporting previously described its IKr inhibition, which would explain Tpeak-Tend prolongation and heart-rate reduction in this study. Meanwhile, ivermectin did not significantly prolong J-Tpeakc or terminal repolarization period, indicating torsadogenic potential of ivermectin leading to the onset of cardiopulmonary arrest would be small. Thus, ivermectin has a broad range of cardiovascular safety profiles, which will help facilitate its drug repurposing.


Assuntos
Parada Cardíaca , Isoflurano , Animais , Cães , Isoflurano/toxicidade , Ivermectina/toxicidade , Seguimentos , Taquicardia/induzido quimicamente , Antiparasitários/toxicidade , Frequência Cardíaca
6.
Brain Res Bull ; 204: 110790, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37852420

RESUMO

BACKGROUND: Postoperative cognitive dysfunction (POCD) is characterized by impaired learning and memory. 6 h duration isoflurane anesthesia is an important factor to induce POCD, and the dysfunction of ryanodine receptor (RyR) in the hippocampus may be involved in this process. We investigated the expression of RyR3 in the hippocampus of mice after 6-h duration isoflurane anesthesia, as well as the improvement of RyR receptor agonist caffeine on POCD mice, while attempting to identify the underlying molecular mechanism. MATERIALS: We constructed a POCD model using 8-week-old male C57BL/6J mice that were exposed to 6-h duration isoflurane. Prior to the three-day cognitive behavioral experiment, RyR agonist caffeine were injected. Fear conditioning and location memory tests were used in behavioral studies. We also exposed the mouse neuroblastoma cell line Neuro-2a (N2A) to 6-h duration isoflurane exposure to simulate the conditions of in vivo cognitive dysfunction. We administered ryanodine receptor agonist (caffeine) and inhibitor (ryanodine) to N2a cells. Following that, we performed a series of bioinformatics analysis to discover proteins that are involved in the development of cognitive dysfunction. Rt-PCR and Western blot were used to assess mRNA level and protein expression. RESULTS: 6-h duration isoflurane anesthesia induced cognitive dysfunction and increased RyR3 mRNA levels in hippocampus. The mRNA levels of RyR3 in cultured N2a cells after anesthesia were comparable to those in vivo, and the RyR agonist caffeine corrected the expression of some cognitive-related phenotypic proteins that were disturbed after anesthesia. Intraperitoneal injection of RyR agonist caffeine can improve cognitive function after isoflurane anesthesia in mice, and bioinformatics analyses suggest that CaMKⅣ may be involved in the molecular mechanism. CONCLUSION: Ryanodine receptor agonist caffeine may improve cognitive dysfunction in mice after isoflurane anesthesia.


Assuntos
Anestésicos Inalatórios , Disfunção Cognitiva , Isoflurano , Complicações Cognitivas Pós-Operatórias , Masculino , Camundongos , Animais , Isoflurano/toxicidade , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos adversos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Anestésicos Inalatórios/toxicidade , Cafeína/farmacologia , Camundongos Endogâmicos C57BL , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Complicações Cognitivas Pós-Operatórias/metabolismo , RNA Mensageiro/metabolismo , Hipocampo/metabolismo
7.
Br J Anaesth ; 131(5): 832-846, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37770252

RESUMO

BACKGROUND: Volatile anaesthetics are widely used in human medicine. Although generally safe, hypersensitivity and toxicity can occur in rare cases, such as in certain genetic disorders. Anaesthesia hypersensitivity is well-documented in a subset of mitochondrial diseases, but whether volatile anaesthetics are toxic in this setting has not been explored. METHODS: We exposed Ndufs4(-/-) mice, a model of Leigh syndrome, to isoflurane (0.2-0.6%), oxygen 100%, or air. Cardiorespiratory function, weight, blood metabolites, and survival were assessed. We exposed post-symptom onset and pre-symptom onset animals and animals treated with the macrophage depleting drug PLX3397/pexidartinib to define the role of overt neuroinflammation in volatile anaesthetic toxicities. RESULTS: Isoflurane induced hyperlactataemia, weight loss, and mortality in a concentration- and duration-dependent manner from 0.2% to 0.6% compared with carrier gas (O2 100%) or mock (air) exposures (lifespan after 30-min exposures ∗P<0.05 for isoflurane 0.4% vs air or vs O2, ∗∗P<0.005 for isoflurane 0.6% vs air or O2; 60-min exposures ∗∗P<0.005 for isoflurane 0.2% vs air, ∗P<0.05 for isoflurane 0.2% vs O2). Isoflurane toxicity was significantly reduced in Ndufs4(-/-) exposed before CNS disease onset, and the macrophage depleting drug pexidartinib attenuated sequelae of isoflurane toxicity (survival ∗∗∗P=0.0008 isoflurane 0.4% vs pexidartinib plus isoflurane 0.4%). Finally, the laboratory animal standard of care of 100% O2 as a carrier gas contributed significantly to weight loss and reduced survival, but not to metabolic changes, and increased acute mortality. CONCLUSIONS: Isoflurane is toxic in the Ndufs4(-/-) model of Leigh syndrome. Toxic effects are dependent on the status of underlying neurologic disease, largely prevented by the CSF1R inhibitor pexidartinib, and influenced by oxygen concentration in the carrier gas.


Assuntos
Anestésicos Inalatórios , Isoflurano , Doença de Leigh , Humanos , Animais , Camundongos , Isoflurano/toxicidade , Anestésicos Inalatórios/toxicidade , Doença de Leigh/genética , Oxigênio , Redução de Peso , Complexo I de Transporte de Elétrons
8.
Acta Physiol (Oxf) ; 239(1): e14009, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37330999

RESUMO

AIM: General anesthesia can induce cognitive deficits in both humans and rodents, correlating with pathological alterations in the hippocampus. However, whether general anesthesia affects olfactory behaviors remains controversial as clinical studies have produced inconsistent results. Therefore, we aimed to investigate how olfactory behaviors and neuronal activity are affected by isoflurane exposure in adult mice. METHODS: The olfactory detection test, olfactory sensitivity test, and olfactory preference/avoidance test were used to examine olfactory function. In vivo electrophysiology was performed in awake, head-fixed mice to record single-unit spiking and local field potentials in the olfactory bulb (OB). We also performed patch-clamp recordings of mitral cell activity. For morphological studies, immunofluorescence and Golgi-Cox staining were applied. RESULTS: Repeated exposure to isoflurane impaired olfactory detection in adult mice. The main olfactory epithelium, the first region exposed to anesthetics, displayed increased proliferation of basal stem cells. In the OB, a crucial hub for olfactory processing, repeated isoflurane exposure increased the odor responses of mitral/tufted cells. Furthermore, the odor-evoked high gamma response was decreased after isoflurane exposure. Whole-cell recordings further indicated that repeated isoflurane exposure increased the excitability of mitral cells, which may be due to weakened inhibitory input in isoflurane-exposed mice. In addition, elevated astrocyte activation and glutamate transporter-1 expression in the OB were observed in isoflurane-exposed mice. CONCLUSIONS: Our findings indicate that repeated isoflurane exposure impairs olfactory detection by increasing neuronal activity in the OB in adult mice.


Assuntos
Isoflurano , Olfato , Humanos , Camundongos , Animais , Olfato/fisiologia , Bulbo Olfatório/fisiologia , Isoflurano/toxicidade , Neurônios/fisiologia , Odorantes
9.
Folia Neuropathol ; 61(1): 68-76, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37114962

RESUMO

INTRODUCTION: As one of the most commonly used anesthetics, isoflurane has been demonstrated to possess a variety of protective effects. However, its' neurological impaired effect should be considered during clinical application. Roles of lncRNA BDNF-AS (BDNF-AS) and miR-214-3p in isoflurane-injured microglia and rats were investigated in this study, aiming to disclose the mechanism of isoflurane damage and to provide candidate therapeutic targets. MATERIAL AND METHODS: Isoflurane-induced microglia cells and rat models were established with 1.5% isoflurane. Inflammation and oxidative stress of microglia cells were evaluated with a level of pro-inflammation cytokines, malondialdehyde (MDA), superoxide dismutase (SOD), and nitrite. Cognitive and learning function of rats were assessed with Morris water maze task. Expressions of BDNF-AS and miR-214-3p and their function in the isoflurane-induced microglia cells and rats were estimated with PCR and corresponding transfection. RESULTS: Isoflurane induced significant neuro-inflammation and oxidative stress in the microglia cells. The increased BDNF-AS and the decreased miR-214-3p were noted, and BDNF-AS was found to negatively regulate miR-214-3p in the isoflurane-induced microglia cells. Isoflurane caused cognitive dysfunction in rats, and resulted in a significant inflammatory response. The knockdown of BDNF-AS significantly alleviated the neurological impairment induced by isoflurane, which was reversed by silencing miR-214-3p. CONCLUSIONS: In isoflurane-induced neuro-inflammation and cognitive dysfunction, BDNF-AS showed a significant protective effect on the neurological impairment induced by isoflurane through modulating miR-214-3p.


Assuntos
Disfunção Cognitiva , Isoflurano , MicroRNAs , RNA Longo não Codificante , Ratos , Animais , Isoflurano/toxicidade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Disfunção Cognitiva/induzido quimicamente , Inflamação/induzido quimicamente , MicroRNAs/genética , MicroRNAs/metabolismo
10.
Fundam Clin Pharmacol ; 37(5): 937-946, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37085979

RESUMO

Volatile anesthetics may cause vascular dysfunction; however, underlying effects are unclear. The aim of the present study was to investigate whether sevoflurane and isoflurane affect vascular function, nitric oxide (NO) bioavailability, and biomarkers of oxidative stress and inflammation. Wistar rats were divided into three experimental groups: Not anesthetized (control group) or submitted to anesthesia with isoflurane (Iso group) or sevoflurane (Sevo group). Hemodynamic parameters were monitored during anesthesia, and blood gas values and biochemical determinants were analyzed. Isometric contractions were recorded in aortic rings. Vasoconstriction induced by potassium chloride (KCl) and phenylephrine (Phe) were measured. No differences in hemodynamic parameters and blood gasses variables were observed. Impaired KCl and Phe-induced contractions were observed in endothelium-intact aorta of Sevo compared to Iso and Control groups. Redox imbalance was found in Sevo and Iso groups. Reduced NO bioavailability and increased activity of matrix metalloproteinase 2 (MMP-2) were observed in Sevo, but not in the Iso group. While reduced IL-10 and IL-1ß were observed in Sevo, increases in IL-1ß in the Iso group were found. Sevoflurane, but not isoflurane, anesthesia impairs vasocontraction, and reduced NO and cytokines and increased MMP-2 activity may be involved in vascular dysfunction after sevoflurane anesthesia.


Assuntos
Anestesia , Anestésicos Inalatórios , Isoflurano , Éteres Metílicos , Ratos , Animais , Isoflurano/toxicidade , Sevoflurano , Metaloproteinase 2 da Matriz , Éteres Metílicos/toxicidade , Anestésicos Inalatórios/toxicidade , Ratos Wistar
11.
Toxicol Mech Methods ; 33(4): 279-292, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36127839

RESUMO

Damage to hippocampus, cerebellum, and cortex associated with cognitive functions due to anesthetic-induced toxicity early in life may cause cognitive decline later. Aquaporin 4 (AQP4), a key protein in waste clearance pathway of brain, is involved in synaptic plasticity and neurocognition. We investigated the effects of single and repeated isoflurane (Iso) anesthesia on AQP4 levels and brain damage. Postnatal-day (P)7 Wistar albino rats were randomly assigned to Iso or Control (C) groups. For single-exposure, pups were exposed to 1.5% Iso in 30% oxygenated-air for 3-h at P7 (Iso1). For repeated-exposure, pups were exposed to Iso for 3 days, 3-h each day, at 1-day intervals (P7 + 9 + 11) starting at P7 (Iso3). C1 and C3 groups received only 30% oxygenated-air. Based on HE-staining and immunoblotting (Bax/Bcl-2, cleaved-caspase3 and PARP1) analyses, Iso exposures caused a higher degree of apoptosis in hippocampus. Anesthesia increased 4-hydroxynonenal (4HNE), oxidative stress marker; the highest ROS accumulation was determined in cerebellum. Increased inflammation (TNF-α, NF-κB) was detected. Multiple Iso-exposures caused more significant damage than single exposure. Moreover, 4HNE and TNF-α contributed synergistically to Iso-induced neurotoxicity. After anesthesia, higher expression of AQP4 was detected in cortex than hippocampus and cerebellum. There was an inverse correlation between increased AQP4 levels and apoptosis/ROS/inflammation. Correlation analysis indicated that AQP4 had a more substantial protective profile against oxidative stress than apoptosis. Remarkably, acutely increased AQP4 against Iso exhibited a more potent neuroprotective effect in cortex, especially frontal cortex. These findings promote further research to understand better the mechanisms underlying anesthesia-induced toxicity in the developing brain.


Assuntos
Isoflurano , Animais , Ratos , Isoflurano/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Aquaporina 4/metabolismo , Aquaporina 4/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Ratos Wistar , Hipocampo , Apoptose , Encéfalo/metabolismo , Inflamação/metabolismo , Animais Recém-Nascidos
12.
Cell Biol Toxicol ; 39(5): 2133-2148, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-35249202

RESUMO

BACKGROUND: Repeated neonatal exposure to anesthetics may disturb neurodevelopment and cause neuropsychological disorders. The m6A modification participates in the gene regulation of neurodevelopment in mouse fetuses exposed to anesthetics. This study aims to explore the underlying molecular mechanisms of neurotoxicity after early-life anesthesia exposure. METHODS: Mice were exposed to isoflurane (1.5%) or sevoflurane (2.3%) for 2 h daily during postnatal days (PND) 7-9. Sociability, spatial working memory, and anxiety-like behavior were assessed on PND 30-35. Synaptogenesis, epitranscriptome m6A, and the proteome of brain regions were evaluated on PND 21. RESULTS: Both isoflurane and sevoflurane produced abnormal social behaviors at the juvenile age, with different sociality patterns in each group. Synaptogenesis in the hippocampal area CA3 was increased in the sevoflurane-exposed mice. Both anesthetics led to numerous persistent m6A-induced alterations in the brain, associated with critical metabolic, developmental, and immune functions. The proteins altered by isoflurane exposure were mainly associated with epilepsy, ataxia, and brain development. As for sevoflurane, the altered proteins were involved in social behavior. CONCLUSIONS: Social interaction, the modulation patterns of the m6A modification, and protein expression were altered in an isoflurane or sevoflurane-specific way. Possible molecular pathways involved in brain impairment were revealed, as well as the mechanism underlying behavioral deficits following repeated exposure to anesthetics in newborns.


Assuntos
Anestésicos Inalatórios , Isoflurano , Éteres Metílicos , Animais , Camundongos , Isoflurano/toxicidade , Sevoflurano , Animais Recém-Nascidos , Proteoma , Anestésicos Inalatórios/toxicidade , Éteres Metílicos/toxicidade , Encéfalo
13.
Basic Clin Pharmacol Toxicol ; 132(2): 180-196, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36321664

RESUMO

Perioperative neurocognitive disorders (PND) is a common complication that occurs among elderly patients in the perioperative course. Current clinical evidence has shown that isoflurane exposure could cause cognitive decline, but the exact molecular mechanisms remain unclear. As both NMDARs-dependent synaptic plasticity and histone acetylation play vital roles in processing learning and memory, we postulated that these alternations might occur in the isoflurane-associated PND. Here, we found that isoflurane impaired fear memory in aged mice, decreased GluN2B-containing NMDA receptors phosphorylation and trafficking, as well as the expression of EphB2, a key regulator of synaptic localization of NMDA receptors. We also identified that isoflurane could increase the expression of HDAC2, which was significantly enriched at the ephb2 gene promoter and regulated the transcription of ephb2. Furthermore, we showed that suberoylanilide hydroxamic acid (SAHA), a nonselective HDAC inhibitor or knocking-down HDAC2 rescued the cognitive dysfunction in isoflurane-treated aged mice via increasing acetylation of H3Ac, expression of EphB2 and promoting NMDA receptor trafficking. Collectively, our study highlighted the crucial role of histone posttranslational modifications for EphB2-GluN2B signals in isoflurane-associated PND, and modulating HDAC2 might be a new therapeutic strategy for isoflurane-associated PND.


Assuntos
Isoflurano , Camundongos , Animais , Isoflurano/toxicidade , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Histonas/metabolismo , Acetilação , Hipocampo/metabolismo , Processamento de Proteína Pós-Traducional , Neurônios/metabolismo , Cognição
14.
Neurotox Res ; 40(6): 1924-1936, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36441450

RESUMO

Neonatal exposure to general anesthetics has been associated with neurotoxicity and morphologic changes in the developing brain. Isoflurane is a volatile anesthetic widely used in pediatric patients to induce general anesthesia, analgesia, and perioperative sedation. In the present study, we investigated the effects of a single neonatal isoflurane (3% in oxygen, 2 h) exposure in rats at postnatal day (PND) 7, in short-term (24 h - PND8) and long-term (adulthood) protocols. In PND8, ex vivo analysis of hippocampal and frontal cortex slices evaluated cell viability and susceptibility to in vitro glutamate challenge. In adult rats, behavioral parameters related to anxiety-like behavior, short-term memory, and locomotor activity (PND60-62) and ex vivo analysis of cell viability, membrane permeability, glutamate uptake, and susceptibility to in vitro glutamate challenge in hippocampal and cortical slices from PND65. A single isoflurane (3%, 2 h) exposure at PND7 did not acutely alter cell viability in cortical and hippocampal slices of infant rats (PND8) per se and did not alter slice susceptibility to in vitro glutamate challenge. In rat's adulthood, behavioral analysis revealed that the neonatal isoflurane exposure did not alter anxiety-like behavior and locomotor activity (open field and rotarod tests). However, isoflurane exposure impaired short-term memory evaluated in the novel object recognition task. Ex vivo analysis of brain slices showed isoflurane neonatal exposure selectively decreased cell viability and glutamate uptake in cortical slices, but it did not alter hippocampal slice viability or glutamate uptake (PND65). Isoflurane exposure did not alter in vitro glutamate-induced neurotoxicity to slices, and isoflurane exposure caused no significant long-term damage to cell membranes in hippocampal or cortical slices. These findings indicate that a single neonatal isoflurane exposure did not promote acute damage; however, it reduced cortical, but not hippocampal, slice viability and glutamate uptake in the adulthood. Additionally, behavioral analysis showed neonatal isoflurane exposure induces short-term recognition memory impairment, consolidating that neonatal exposure to volatile anesthetics may lead to behavioral impairment in the adulthood, although it may damage brain regions differentially.


Assuntos
Anestésicos Inalatórios , Anestésicos , Isoflurano , Ratos , Animais , Isoflurano/toxicidade , Ácido Glutâmico/metabolismo , Memória de Curto Prazo , Sobrevivência Celular , Hipocampo , Lobo Frontal/metabolismo , Córtex Cerebral/metabolismo , Anestésicos Inalatórios/toxicidade
15.
Hum Exp Toxicol ; 41: 9603271221132152, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263453

RESUMO

BACKGROUND: This observation aimed to investigate the effect of colorectal neoplasia differentially expressed (CRNDE) targeted miR-212-5p on cognitive impairment induced by isoflurane (ISO) anesthesia in rats. METHODS: The cognitive function of rats was measured by Morris water maze test. QRT-PCR detection of CRNDE and miR-212-5p expression levels in rats in each group. Double luciferase was used to verify the targeting relationship between miR-212-5p and CRNDE, and commercial kits were used to detect the level of inflammatory cytokines in hippocampus. RESULTS: The concentration of CRNDE was enhanced in rats treated by ISO anesthetic. The neurological severity score was elevated, the escape latency of rats was prolonged, the stay time in the quadrant of the platform, and the number of times crossing the platform decreased in the ISO group. The above indexes of rats in ISO + si-CRNDE were improved. MiR-212-5p is a mediator in the management of CRNDE on cognition and inflammation. CONCLUSION: CRNDE led to the deterioration of impairment on cognition induced by ISO through suppressing miR-212-5p expression and promoting neuroinflammation.


Assuntos
Anestésicos , Neoplasias Colorretais , Isoflurano , MicroRNAs , RNA Longo não Codificante , Ratos , Animais , RNA Longo não Codificante/genética , Isoflurano/toxicidade , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Colorretais/genética , Citocinas
16.
Life Sci ; 309: 121004, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36170891

RESUMO

In this study, the effects of exposure to isoflurane, sevoflurane and desflurane on the oxidative response and inflammation at different times was analyzed in the lungs of adult C57BL/6 mice. 120 animals were divided into 3 groups (n = 40): Isoflurane (ISO), Sevoflurane (SEV) and Desflurane (DES) and exposed to these anesthetics for 1 h (n = 10), 2 h (n = 10) and 3 h (n = 10), at a minimum alveolar concentration (MAC) equal to 1. The control group (CG) (n = 10) was exposed to ambient air. 24 h after the experimental protocol, the animals were euthanized and the bronchoalveolar lavage fluid (BALF), blood and lung tissue samples were collected. In the BALF, animals exposed to isoflurane for 2 h and 3 h showed a greater influx of leukocytes, especially macrophages compared to the CG. The ISO3h had lower leukocyte counts in the peripheral blood compared to CG, ISO1h and ISO2h. There was an increase in CCL-2 levels in the ISO3h compared to the CG. Superoxide dismutase activity was higher in ISO1h compared to CG. The activity of catalase was higher in the ISO1h and ISO2h compared to the CG. The lipid peroxidation, as well as carbonylated protein were higher in the ISO3h compared to the CG (p < 0.05). Similar results were observed in the exposure of SEV and DES compared to inflammation and redox imbalance in different periods. This study demonstrated that time is a determinant to promote a local and systemic inflammatory response to different inhalational anesthetics in a healthy murine model.


Assuntos
Anestésicos Inalatórios , Isoflurano , Éteres Metílicos , Camundongos , Animais , Isoflurano/toxicidade , Sevoflurano/efeitos adversos , Desflurano , Catalase/metabolismo , Camundongos Endogâmicos C57BL , Anestésicos Inalatórios/toxicidade , Superóxido Dismutase/metabolismo , Inflamação/induzido quimicamente , Éteres Metílicos/farmacologia
17.
Hum Exp Toxicol ; 41: 9603271221116276, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36000339

RESUMO

BACKGROUND: Post-operative recognition dysfunction (POCD) is a kind of central nervous system complication that appears after operative anesthesia. Recent studies on the mechanism of long non-coding RNA (lncRNA) in neurodegenerative diseases are abundant. AIMS: The study aimed to explore the expression pattern and role of lncRNA OIP5-AS1 in POCD and to investigate its underlying mechanism in old rats. METHODS: The old rats were exposed to isoflurane to mimic the POCD in the elderly, and their cognitive function was tested via Morris water maze (MWM) test. Enzyme linked immunosorbent assay was applied for the concentration detection of inflammation and oxidative stress-related factors. Luciferase reporter assay was done for the target gene analysis. RESULTS: Downregulation of OIP5-AS1 was accompanied by isoflurane treatment in rats, overexpression of OIP5-AS1 induced the rats to spend more time in the target quadrant, and shorten escape latency time. OIP5-AS1 inhibited the release of TNF-α, IL-6 and IL-1ß, GSH and superoxide dismutase, decreased the activation of caspase-3, but promoted malondialdehyde release. miR-186-5p was a target miRNA of OIP5-AS1, and exhibited high expression in rats after isoflurane exposure. miR-186-5p can abolish the beneficial role of OIP5-AS1 against cognitive impairment, inflammatory response, oxidative stress and neuron apoptosis. CONCLUSION: OIP5-AS1 plays a neuroprotective role in elderly POCD rats through sponging miR-186-5p, and it is related to OIP5-AS1/miR-186-5p mediated inflammatory response, oxidative stress and neuron apoptosis.


Assuntos
Disfunção Cognitiva , Isoflurano , MicroRNAs , RNA Longo não Codificante , Animais , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/genética , Regulação para Baixo , Isoflurano/toxicidade , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ratos
18.
Chem Biol Interact ; 367: 110114, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36027947

RESUMO

Dexmedetomidine (DEX) displays a neuroprotective role in aged rats with isoflurane (ISO)-induced cognitive impairment through antioxidant, and anti-inflammatory, and anti-apoptotic effects. Therefore, the present study was performed to define the molecular mechanism of DEX on ISO-induced neurological impairment in aged rats in relation to the MEK1/ERK1/Nrf2/HO-1 axis. The study enrolled elderly patients undergoing ISO anesthesia. Patient cognitive function following treatment with DEX was evaluated using mini-mental state examination (MMSE). The results revealed that DEX supplementation of anesthesia contributed to higher MMSE scores in patients one week post treatment. Rat model of neurological impairment was also induced in 18-month-age Wistar rats by ISO, followed by DEX treatment. Based on the results of Morris water maze experiment, ELISA, and TUNEL and hematoxylin-eosin staining, in vivo experiments confirmed that DEX could reduce the oxidative stress and neurological damage induced by ISO in rats. DEX activated the nuclear factor erythroid 2-related factor (Nrf2)/Heme Oxygenase 1 (HO-1) pathway. DEX upregulated the expression of Nrf2 and HO-1 by activating the MEK1/ERK1 pathway, whereby attenuating the ISO-caused oxidative stress and neurological damage in rats. Collectively, DEX suppresses the ISO-induced neurological impairment in the aged rats by promoting HO-1 through activation of the MEK1/ERK1/Nrf2 axis.


Assuntos
Dexmedetomidina , Isoflurano , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/metabolismo , Dexmedetomidina/farmacologia , Amarelo de Eosina-(YS)/farmacologia , Hematoxilina/farmacologia , Heme Oxigenase-1/metabolismo , Isoflurano/toxicidade , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 1/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Ratos Wistar
19.
Comput Math Methods Med ; 2022: 2517463, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35872948

RESUMO

Isoflurane (ISO) is a type of anesthetic that might cause neurotoxicity in children. Although miR-424-5p is considerably downregulated in ISO-treated rat brain samples, its physiological role in ISO-induced neuronal injury in human embryonic stem cell-derived neurons remains unknown (hESC-derived neurons). miR-424-5p expression and fatty acid synthase (FASN) in ISO-treated hESC-derived neurons were tested via qRT-PCR. The amount of protein for Bax, Cleaved-caspase-8, Bcl-2, and FASN was investigated through western blot analysis. The viability and apoptosis of hESC-derived neurons were estimated through cell counting kit-8 assessment and TUNEL assay, accordingly. Superoxide dismutase, glutathione, and malondialdehyde levels were discovered via corresponding kits. The contents of inflammatory factors including interleukin-6 and tumor necrosis factor-α were examined by enzyme-linked immunosorbent assays. The combination between FASN and miR-424-5p was resolute via dual-luciferase reporter assessment. After exposure to ISO, induced neurotoxicity and a decreased miR-424-5p production were identified in hESC-derived neurons. Upregulation of miR-424-5p repressed ISO-induced apoptosis and mitigated ISO-induced inflammatory response and oxidative stress in vitro. FASN expression levels were reduced by elevation of miR-424-5p and upregulated after ISO treatment. Mechanically, FASN was directly targeted by miR-424-5p in hESC-derived neurons. Of note, the miR-424-5p elevation-suppressed neuronal apoptosis, inflammatory response, and oxidative stress were countered by upregulation of FASN.


Assuntos
Anestesia , Ácido Graxo Sintase Tipo I , Isoflurano , MicroRNAs , Neurônios , Apoptose/genética , Ácido Graxo Sintase Tipo I/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Isoflurano/toxicidade , MicroRNAs/genética , MicroRNAs/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia
20.
Anesthesiology ; 137(2): 212-231, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35504002

RESUMO

BACKGROUND: Inhalational anesthetics are known to disrupt PDZ2 domain-mediated protein-protein interactions of the postsynaptic density (PSD)-95 protein. The aim of this study is to investigate the underlying mechanisms in response to early isoflurane exposure on synaptic PSD-95 PDZ2 domain disruption that altered spine densities and cognitive function. The authors hypothesized that activation of protein kinase-G by the components of nitric oxide (NO) signaling pathway constitutes a mechanism that prevents loss of early dendritic spines and synapse in neurons and cognitive impairment in mice in response to disruption of PDZ2 domain of the PSD-95 protein. METHODS: Postnatal day 7 mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active PSD-95 wild-type PDZ2 peptide or soluble guanylyl cyclase activator YC-1 along with their respective controls. Primary neurons at 7 days in vitro were exposed to isoflurane or PSD-95 wild-type PDZ2 peptide for 4 h. Coimmunoprecipitation, spine density, synapses, cyclic guanosine monophosphate-dependent protein kinase activity, and novel object recognition memory were assessed. RESULTS: Exposure of isoflurane or PSD-95 wild-type PDZ2 peptide relative to controls causes the following. First, there is a decrease in PSD-95 coimmunoprecipitate relative to N-methyl-d-aspartate receptor subunits NR2A and NR2B precipitate (mean ± SD [in percentage of control]: isoflurane, 54.73 ± 16.52, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 51.32 ± 12.93, P = 0.001). Second, there is a loss in spine density (mean ± SD [spine density per 10 µm]: control, 5.28 ± 0.56 vs. isoflurane, 2.23 ± 0.67, P < 0.0001; and PSD-95 mutant PDZ2 peptide, 4.74 ± 0.94 vs. PSD-95 wild-type PDZ2 peptide, 1.47 ± 0.87, P < 0.001) and a decrease in synaptic puncta (mean ± SD [in percentage of control]: isoflurane, 41.1 ± 14.38, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 50.49 ± 14.31, P < 0.001). NO donor or cyclic guanosine monophosphate analog prevents the spines and synapse loss and decline in the cyclic guanosine monophosphate-dependent protein kinase activity, but this prevention was blocked by soluble guanylyl cyclase or protein kinase-G inhibitors in primary neurons. Third, there were deficits in object recognition at 5 weeks (mean ± SD [recognition index]: male, control, 64.08 ± 10.57 vs. isoflurane, 48.49 ± 13.41, P = 0.001, n = 60; and female, control, 67.13 ± 11.17 vs. isoflurane, 53.76 ± 6.64, P = 0.003, n = 58). Isoflurane-induced impairment in recognition memory was preventable by the introduction of YC-1. CONCLUSIONS: Activation of soluble guanylyl cyclase or protein kinase-G prevents isoflurane or PSD-95 wild-type PDZ2 peptide-induced loss of dendritic spines and synapse. Prevention of recognition memory with YC-1, a NO-independent activator of guanylyl cyclase, supports a role for the soluble guanylyl cyclase mediated protein kinase-G signaling in countering the effects of isoflurane-induced cognitive impairment.


Assuntos
Disfunção Cognitiva , Proteínas Quinases Dependentes de GMP Cíclico , Proteína 4 Homóloga a Disks-Large , Isoflurano , Animais , Disfunção Cognitiva/induzido quimicamente , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Feminino , Guanosina Monofosfato , Isoflurano/toxicidade , Masculino , Camundongos , Óxido Nítrico/metabolismo , Peptídeos , Densidade Pós-Sináptica , Transdução de Sinais , Guanilil Ciclase Solúvel , Sinapses
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...