Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.405
Filter
1.
Drug Des Devel Ther ; 18: 1727-1741, 2024.
Article in English | MEDLINE | ID: mdl-38803563

ABSTRACT

Purpose: To investigate and quantify the effect of continuous esketamine infusion at different doses on the bispectral index (BIS) during sevoflurane anesthesia. Methods: A total of 120 patients scheduled for elective laparoscopic renal surgery were randomly divided into three groups. Under steady anesthesia and surgical situations, the patient was started on continuous infusion of the study drug: 0.125 mg/kg/h esketamine (group E1), 0.25 mg/kg/h esketamine (group E2), and the same volume of saline (group C). The primary outcome was changes in BIS value after 15 min (T15), 30 min (T30), 45 min (T45), and 60 min (T60) of drug infusion. The secondary outcomes were 95% spectral edge frequency (SEF95), electromyogram (EMG), heart rate (HR), and mean arterial pressure (MAP) from T0 to T60. Furthermore, postoperative pain, postoperative recovery, and perioperative adverse events were evaluated. Results: Compared with group C, group E1 exhibited significant BIS elevation at T30-T60 and group E2 at T15-T60 (P < 0.001). Compared with group E1, group E2 showed a more significant BIS elevation at T15-T60 (P < 0.001). The area under the curve (AUC) of BIS and SEF95 were significantly higher in group E2 than in groups C and E1 (P < 0.05). BIS value for any of the three groups was significantly correlated with SEF95 (P < 0.001). No significant differences were observed in the AUC of EMG, HR, and MAP among the three groups. Intraoperative remifentanil consumption and postoperative NRS of pain on movement were significantly reduced in group E2 compared with groups C and E1 (P < 0.05). Conclusion: Continuous infusion of both 0.125 and 0.25 mg/kg/h of esketamine increased the BIS value during sevoflurane anesthesia, and the BIS value gradually stabilized with the prolongation of the infusion time.


Subject(s)
Ketamine , Sevoflurane , Humans , Sevoflurane/administration & dosage , Sevoflurane/pharmacology , Ketamine/administration & dosage , Ketamine/pharmacology , Male , Female , Middle Aged , Adult , Dose-Response Relationship, Drug , Infusions, Intravenous , Anesthetics, Inhalation/administration & dosage , Double-Blind Method , Pain, Postoperative/drug therapy , Pain, Postoperative/prevention & control
2.
J Gene Med ; 26(5): e3692, 2024 May.
Article in English | MEDLINE | ID: mdl-38745073

ABSTRACT

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.


Subject(s)
Kruppel-Like Transcription Factors , Liver , Macrophages , Reperfusion Injury , Sevoflurane , Animals , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Mice , Macrophages/metabolism , Sevoflurane/pharmacology , Liver/metabolism , Liver/pathology , Transcriptional Activation , Male , Disease Models, Animal , Apoptosis , CD18 Antigens/metabolism , CD18 Antigens/genetics , Cell Line , Mice, Inbred C57BL , Gene Expression Regulation
3.
Sci Rep ; 14(1): 11178, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38750181

ABSTRACT

Although sevoflurane is generally considered safe, reports suggest that sevoflurane may cause postoperative liver injury more frequently than previously believed. Therefore, we aimed to compare the incidence of clinically significant postoperative liver injury following non-cardiac surgery between patients who underwent sevoflurane anesthesia and propofol-based total intravenous anesthesia. We retrospectively reviewed adult surgical patients from January 2010 to September 2022 who underwent general anesthesia in our center using sevoflurane or propofol over 3 h. After 1:1 propensity score matching, the incidence of postoperative liver injury was compared between the two groups. Out of 58,300 patients reviewed, 44,345 patients were included in the analysis. After propensity score matching, 7767 patients were included in each group. The incidence of postoperative liver injury was 1.4% in the sevoflurane group, which was similar to that in the propofol group (1.6%; p = 0.432). Comparison of the severity of postoperative alanine aminotransferase elevation showed that the incidence of borderline and mild elevation was higher in the sevoflurane group, but there was no difference in the incidence of moderate and severe elevation. In conclusion, sevoflurane anesthesia over 3 h was not associated with a higher incidence of clinically significant postoperative liver injury compared to propofol anesthesia.


Subject(s)
Postoperative Complications , Propofol , Sevoflurane , Humans , Sevoflurane/adverse effects , Propofol/adverse effects , Propofol/administration & dosage , Male , Female , Retrospective Studies , Middle Aged , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Aged , Anesthetics, Intravenous/adverse effects , Anesthetics, Intravenous/administration & dosage , Incidence , Anesthetics, Inhalation/adverse effects , Adult , Propensity Score , Liver/drug effects , Anesthesia, General/adverse effects , Chemical and Drug Induced Liver Injury/epidemiology , Chemical and Drug Induced Liver Injury/etiology
4.
Ann Transplant ; 29: e943281, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38803088

ABSTRACT

BACKGROUND We aimed to assess the effect of dexmedetomidine (Dex) combined with remifentanil on emergence agitation (EA) during awakening from sevoflurane anesthesia for pediatric liver surgery. MATERIAL AND METHODS Sixty children who underwent liver surgery in our hospital were prospectively selected and randomly allocated into group A (placebo+remifentanil+sevoflurane) or group B (Dex+remifentanil+sevoflurane). Mean arterial pressure (MAP) and heart rate (HR) at different time points, agitation score during awakening, behavioral status, pain level, and the incidence of postoperative adverse effects were compared in both groups. RESULTS Children in group B had lower HR and MAP levels immediately after tracheal extubation and 5 min after tracheal extubation than those in group A. The Aono's scores, PAED agitation scores, and CHIPP scores at 15 min and 30 min of admission to the PACU were lower in group B than in group A. The incidence of agitation during postoperative anesthesia awakening was lower in group B in contrast to group A. There was no significant difference in postoperative adverse reactions between group A and group B. CONCLUSIONS In pediatric liver surgery, the use of Dex+remifentanil+sevoflurane anesthesia can reduce the incidence of EA during the awakening period, stabilize hemodynamic levels, and relieve postoperative pain, and has fewer postoperative adverse effects, which warrants clinical application.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Emergence Delirium , Remifentanil , Sevoflurane , Humans , Dexmedetomidine/administration & dosage , Dexmedetomidine/therapeutic use , Remifentanil/administration & dosage , Remifentanil/therapeutic use , Sevoflurane/administration & dosage , Female , Male , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/adverse effects , Child, Preschool , Emergence Delirium/prevention & control , Emergence Delirium/etiology , Emergence Delirium/epidemiology , Prospective Studies , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/therapeutic use , Infant , Child , Psychomotor Agitation/prevention & control , Psychomotor Agitation/etiology , Liver/surgery , Anesthesia Recovery Period , Piperidines/administration & dosage , Piperidines/therapeutic use , Piperidines/adverse effects , Double-Blind Method , Drug Therapy, Combination , Methyl Ethers/administration & dosage , Methyl Ethers/adverse effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use
5.
BMC Cardiovasc Disord ; 24(1): 280, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811893

ABSTRACT

BACKGROUND: Myocardial ischemia-reperfusion injury (I/RI) is a major cause of perioperative cardiac-related adverse events and death. Studies have shown that sevoflurane postconditioning (SpostC), which attenuates I/R injury and exerts cardioprotective effects, regulates mitochondrial dynamic balance via HIF-1α, but the exact mechanism is unknown. This study investigates whether the PI3K/AKT pathway in SpostC regulates mitochondrial dynamic balance by mediating HIF-1α, thereby exerting myocardial protective effects. METHODS: The H9C2 cardiomyocytes were cultured to establish the hypoxia-reoxygenation (H/R) model and randomly divided into 4 groups: Control group, H/R group, sevoflurane postconditioning (H/R + SpostC) group and PI3K/AKT blocker (H/R + SpostC + LY) group. Cell survival rate was determined by CCK-8; Apoptosis rate was determined by flow cytometry; mitochondrial membrane potential was evaluated by Mito Tracker™ Red; mRNA expression levels of AKT, HIF-1α, Opa1and Drp1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR); Western Blot assay was used to detect the protein expression levels of AKT, phosphorylated AKT (p-AKT), HIF-1α, Opa1 and Drp1. RESULTS: Compared with the H/R group, the survival rate of cardiomyocytes in the H/R + SpostC group increased, the apoptosis rate decreased and the mitochondrial membrane potential increased. qRT-PCR showed that the mRNA expression of HIF-1α and Opa1 were higher in the H/R + SpostC group compared with the H/R group, whereas the transcription level of Drp1 was lower in the H/R + SpostC group. In the H/R + SpostC + LY group, the mRNA expression of HIF-1α was lower than the H/R + SpostC group. There was no difference in the expression of Opa1 mRNA between the H/R group and the H/R + SpostC + LY group. WB assay results showed that compared with the H/R group, the protein expression levels of HIF-1α, Opa1, P-AKT were increased and Drp1 protein expression levels were decreased in the H/R + SpostC group. HIF-1α, P-AKT protein expression levels were decreased in the H/R + SpostC + LY group compared to the H/R + SpostC group. CONCLUSION: SpostC mediates HIF-1α-regulated mitochondrial fission and fusion-related protein expression to maintain mitochondrial dynamic balance by activating the PI3K/AKT pathway and increasing AKT phosphorylation, thereby attenuating myocardial I/R injury.


Subject(s)
Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Membrane Potential, Mitochondrial , Mitochondria, Heart , Mitochondrial Dynamics , Myocardial Reperfusion Injury , Myocytes, Cardiac , Phosphatidylinositol 3-Kinase , Proto-Oncogene Proteins c-akt , Sevoflurane , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/enzymology , Sevoflurane/pharmacology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/enzymology , Mitochondrial Dynamics/drug effects , Cell Line , Rats , Apoptosis/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Membrane Potential, Mitochondrial/drug effects , Cell Hypoxia , Dynamins/metabolism , Dynamins/genetics , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Cytoprotection , Ischemic Postconditioning , Phosphorylation
6.
Exp Neurol ; 377: 114804, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704083

ABSTRACT

BACKGROUND: Sevoflurane (SEV) has been found to induce neurotoxicity and cognitive impairment, leading to the development of degenerative diseases. Protein kinase C delta (PRKCD) is upregulated in the hippocampus of SEV-treated mice and may be related to SEV-related neurotoxicity. However, the underlying molecular mechanisms by which SEV mediates neurotoxicity via PRKCD remain unclear. METHODS: Normal mice and PRKCD knockout (KO) mice were exposed to SEV. Hippocampal neurons were isolated from mice hippocampal tissues. H&E staining was used for pathological morphology of hippocampal tissues, and NISSL staining was used to analyze the number of hippocampal neurons. The mRNA and protein levels were determined using quantitative real-time PCR, western blot, immunofluorescence staining and immunohistochemical staining. The mitochondrial microstructure was observed by transmission electron microscopy. Cell viability was detected by cell counting kit 8 assay, and ferroptosis was assessed by detecting related marker levels. The cognitive ability of mice was assessed by morris water maze test. And the protein levels of PRKCD, ferroptosis-related markers and Hippo pathway-related markers were examined by western bolt. RESULTS: SEV increased PRKCD expression and ferroptosis in hippocampal tissues of mice. Also, SEV promoted mouse hippocampal neuron injury by inducing ferroptosis via upregulating PRKCD expression. Knockout of PRKCD alleviated SEV-induced neurotoxicity and cognitive impairment in mice, and relieved SEV-induced ferroptosis in hippocampal neurons. PRKCD could inhibit the activity of Hippo pathway, and its knockdown also overturned SEV-mediated ferroptosis by activating Hippo pathway. CONCLUSION: SEV could induce neurotoxicity and cognitive impairment by promoting ferroptosis via inactivating Hippo pathway through increasing PRKCD expression.


Subject(s)
Cognitive Dysfunction , Ferroptosis , Hippo Signaling Pathway , Hippocampus , Mice, Knockout , Protein Kinase C-delta , Protein Serine-Threonine Kinases , Sevoflurane , Signal Transduction , Up-Regulation , Animals , Sevoflurane/toxicity , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/genetics , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Up-Regulation/drug effects , Protein Kinase C-delta/metabolism , Protein Kinase C-delta/genetics , Ferroptosis/drug effects , Ferroptosis/physiology , Mice, Inbred C57BL , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Anesthetics, Inhalation/toxicity , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/metabolism
7.
Exp Neurol ; 377: 114807, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704082

ABSTRACT

Repeated sevoflurane exposure in neonatal mice can leads to neuronal apoptosis and mitochondrial dysfunction. The mitochondria are responsible for energy production to maintain homeostasis in the central nervous system. The mitochondria-associated endoplasmic reticulum membrane (MAM) is located between the mitochondria and endoplasmic reticulum (ER), and it is critical for mitochondrial function and cell survival. MAM malfunction contributes to neurodegeneration, however, whether it is involved in sevoflurane-induced neurotoxicity remains unknown. Our study demonstrated that repeated sevoflurane exposure induced mitochondrial dysfunction and dampened the MAM structure. The upregulated ER-mitochondria tethering enhanced Ca2+ transition from the cytosol to the mitochondria. Overload of mitochondrial Ca2+ contributed to opening of the mitochondrial permeability transition pore (mPTP), which caused neuronal apoptosis. Mitofusin 2(Mfn2), a key regulator of ER-mitochondria contacts, was found to be suppressed after repeated sevoflurane exposure, while restoration of Mfn2 expression alleviated cognitive dysfunction due to repeated sevoflurane exposure in the adult mice. These evidences suggest that sevoflurane-induced MAM malfunction is vulnerable to Mfn2 suppression, and the enhanced ER-mitochondria contacts promotes mitochondrial Ca2+ overload, contributing to mPTP opening and neuronal apoptosis. This paper sheds light on a novel mechanism of sevoflurane-induced neurotoxicity. Furthermore, targeting Mfn2-mediated regulation of the MAM structure and mitochondrial function may provide a therapeutic advantage in sevoflurane-induced neurodegeneration.


Subject(s)
Endoplasmic Reticulum , GTP Phosphohydrolases , Mitochondria , Sevoflurane , Animals , Sevoflurane/toxicity , Sevoflurane/pharmacology , GTP Phosphohydrolases/metabolism , Mice , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mice, Inbred C57BL , Apoptosis/drug effects , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/pharmacology , Male , Calcium/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/drug effects
8.
CNS Neurosci Ther ; 30(5): e14716, 2024 05.
Article in English | MEDLINE | ID: mdl-38698533

ABSTRACT

BACKGROUND: Sevoflurane is a superior agent for maintaining anesthesia during surgical procedures. However, the neurotoxic mechanisms of clinical concentration remain poorly understood. Sevoflurane can interfere with the normal function of neurons and synapses and impair cognitive function by acting on α5-GABAAR. METHODS: Using MWM test, we evaluated cognitive abilities in mice following 1 h of anesthesia with 2.7%-3% sevoflurane. Based on hippocampal transcriptome analysis, we analyzed the differential genes and IL-6 24 h post-anesthesia. Western blot and RT-PCR were performed to measure the levels of α5-GABAAR, Radixin, P-ERM, P-Radixin, Gephyrin, IL-6, and ROCK. The spatial distribution and expression of α5-GABAAR on neuronal somata were analyzed using histological and three-dimensional imaging techniques. RESULTS: MWM test indicated that partial long-term learning and memory impairment. Combining molecular biology and histological analysis, our studies have demonstrated that sevoflurane induces immunosuppression, characterized by reduced IL-6 expression levels, and that enhanced Radixin dephosphorylation undermines the microstructural stability of α5-GABAAR, leading to its dissociation from synaptic exterior and resulting in a disordered distribution in α5-GABAAR expression within neuronal cell bodies. On the synaptic cleft, the expression level of α5-GABAAR remained unchanged, the spatial distribution became more compact, with an increased fluorescence intensity per voxel. On the extra-synaptic space, the expression level of α5-GABAAR decreased within unchanged spatial distribution, accompanied by an increased fluorescence intensity per voxel. CONCLUSION: Dysregulated α5-GABAAR expression and distribution contributes to sevoflurane-induced partial long-term learning and memory impairment, which lays the foundation for elucidating the underlying mechanisms in future studies.


Subject(s)
Anesthetics, Inhalation , Hippocampus , Memory Disorders , Receptors, GABA-A , Sevoflurane , Sevoflurane/toxicity , Animals , Mice , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Anesthetics, Inhalation/toxicity , Receptors, GABA-A/metabolism , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Inbred C57BL , Maze Learning/drug effects , Maze Learning/physiology
9.
Zool Res ; 45(3): 663-678, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38766748

ABSTRACT

A growing number of studies have demonstrated that repeated exposure to sevoflurane during development results in persistent social abnormalities and cognitive impairment. Davunetide, an active fragment of the activity-dependent neuroprotective protein (ADNP), has been implicated in social and cognitive protection. However, the potential of davunetide to attenuate social deficits following sevoflurane exposure and the underlying developmental mechanisms remain poorly understood. In this study, ribosome and proteome profiles were analyzed to investigate the molecular basis of sevoflurane-induced social deficits in neonatal mice. The neuropathological basis was also explored using Golgi staining, morphological analysis, western blotting, electrophysiological analysis, and behavioral analysis. Results indicated that ADNP was significantly down-regulated following developmental exposure to sevoflurane. In adulthood, anterior cingulate cortex (ACC) neurons exposed to sevoflurane exhibited a decrease in dendrite number, total dendrite length, and spine density. Furthermore, the expression levels of Homer, PSD95, synaptophysin, and vglut2 were significantly reduced in the sevoflurane group. Patch-clamp recordings indicated reductions in both the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs). Notably, davunetide significantly ameliorated the synaptic defects, social behavior deficits, and cognitive impairments induced by sevoflurane. Mechanistic analysis revealed that loss of ADNP led to dysregulation of Ca 2+ activity via the Wnt/ß-catenin signaling, resulting in decreased expression of synaptic proteins. Suppression of Wnt signaling was restored in the davunetide-treated group. Thus, ADNP was identified as a promising therapeutic target for the prevention and treatment of neurodevelopmental toxicity caused by general anesthetics. This study provides important insights into the mechanisms underlying social and cognitive disturbances caused by sevoflurane exposure in neonatal mice and elucidates the regulatory pathways involved.


Subject(s)
Animals, Newborn , Cognitive Dysfunction , Proteome , Sevoflurane , Social Behavior , Animals , Sevoflurane/adverse effects , Mice , Cognitive Dysfunction/chemically induced , Ribosomes/drug effects , Ribosomes/metabolism , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/pharmacology , Nerve Tissue Proteins/metabolism , Male , Behavior, Animal/drug effects
10.
Zool Res ; 45(3): 679-690, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38766749

ABSTRACT

General anesthesia is widely applied in clinical practice. However, the precise mechanism of loss of consciousness induced by general anesthetics remains unknown. Here, we measured the dynamics of five neurotransmitters, including γ-aminobutyric acid, glutamate, norepinephrine, acetylcholine, and dopamine, in the medial prefrontal cortex and primary visual cortex of C57BL/6 mice through in vivo fiber photometry and genetically encoded neurotransmitter sensors under anesthesia to reveal the mechanism of general anesthesia from a neurotransmitter perspective. Results revealed that the concentrations of γ-aminobutyric acid, glutamate, norepinephrine, and acetylcholine increased in the cortex during propofol-induced loss of consciousness. Dopamine levels did not change following the hypnotic dose of propofol but increased significantly following surgical doses of propofol anesthesia. Notably, the concentrations of the five neurotransmitters generally decreased during sevoflurane-induced loss of consciousness. Furthermore, the neurotransmitter dynamic networks were not synchronized in the non-anesthesia groups but were highly synchronized in the anesthetic groups. These findings suggest that neurotransmitter dynamic network synchronization may cause anesthetic-induced loss of consciousness.


Subject(s)
Anesthetics, Inhalation , Mice, Inbred C57BL , Neurotransmitter Agents , Propofol , Sevoflurane , Sevoflurane/pharmacology , Animals , Propofol/pharmacology , Neurotransmitter Agents/metabolism , Mice , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
11.
Braz J Med Biol Res ; 57: e13437, 2024.
Article in English | MEDLINE | ID: mdl-38808889

ABSTRACT

Clinical studies have found that neonatal sevoflurane exposure can increase the risk of cognitive dysfunction. However, recent studies have found that it can exhibit neuroprotective effects in some situations. In this study, we aimed to explore the effects of sevoflurane neonatal exposure in rats. A total of 144 rat pups (72 males and 72 females) were assigned to six groups and separately according to sevoflurane exposure of different times on the seventh day after birth. Blood gas analysis and western blot detection in the hippocampus were conducted after exposure. The Morris water maze test was conducted on the 32nd to 38th days after birth. The expression of PSD95 and synaptophysin in the hippocampus was detected after the Morris water maze test. We found that neonatal exposure to sevoflurane promoted apoptosis in the hippocampus, and Bax and caspase-3 were increased in a dose-dependent manner. The 2-h exposure had the greatest effects on cognitive dysfunction. However, with the extension of exposure time to 6 h, the effects on cognitive function were partly compensated. In addition, sevoflurane exposure decreased synaptogenesis in the hippocampus. However, as the exposure time was extended, the suppression of synaptogenesis was attenuated. In conclusion, neonatal sevoflurane exposure exhibited duration-dependent effects on cognitive function via Bax-caspase-3-dependent apoptosis and bidirectional effects on synaptogenesis in rats.


Subject(s)
Animals, Newborn , Cognition , Hippocampus , Sevoflurane , Sevoflurane/pharmacology , Animals , Female , Male , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Cognition/drug effects , Time Factors , Maze Learning/drug effects , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/adverse effects , Apoptosis/drug effects , Sex Factors , Rats, Sprague-Dawley , Methyl Ethers/pharmacology , Blotting, Western , Blood Gas Analysis , Cognitive Dysfunction/chemically induced
12.
Sci Rep ; 14(1): 11889, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38789558

ABSTRACT

Pediatricians use sevoflurane due to its fast action and short recovery time. However, studies have shown that repeated exposure to anesthesia can affect learning and memory. Melatonin, an indole-type neuroendocrine hormone, has significant anti-inflammatory, and neuroprotective properties. Melatonin's impact on cognitive behavior in sevoflurane-anesthetized males and females of the Wistar rats during preadolescence was examined in this research. The cognitive function was evaluated by shuttle box and morris water maze tests, while interleukin-10, Catalase (CAT), Malondialdehyde (MDA), and Tumor Necrosis Factor-α (TNF-α) were evaluated using ELISA kits. The expression levels of the apoptosis-linked proteins, Bax, Bcl-2, and caspase-3, were determined using the western blotting technique. The learning and memory latencies of the rats were more significant in the sevoflurane groups than in the control group; however, the latencies were significantly shorter in the sevoflurane and melatonin groups than in the control group. The levels of MDA, TNF-α, Bax, and caspase-3 were significantly higher in the sevoflurane groups than in the control group. We also found that the levels of CAT and Bcl-2 were significantly reduced in the sevoflurane groups compared to the control group. Increasing levels of CAT, Bcl-2, and decreasing levels of MDA, TNF-α, Bax, and caspase-3 in response to melatonin indicate a possible contribution to the recovery from the sevoflurane impairment. Melatonin shows neuroprotective effects in male and female rats with sevoflurane-induced cognitive impairment. This suggests melatonin could be a valuable treatment for learning and memory deficits resulting from repeated exposure to sevoflurane, possibly by controlling apoptosis, oxidative stress, and inflammation.


Subject(s)
Melatonin , Rats, Wistar , Sevoflurane , Animals , Sevoflurane/adverse effects , Sevoflurane/pharmacology , Melatonin/pharmacology , Male , Female , Rats , Apoptosis/drug effects , Anesthetics, Inhalation/adverse effects , Maze Learning/drug effects , Tumor Necrosis Factor-alpha/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Memory/drug effects , Malondialdehyde/metabolism
13.
BMC Anesthesiol ; 24(1): 167, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702608

ABSTRACT

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.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Electroencephalography , Ketamine , Propofol , Sevoflurane , Animals , Mice , Ketamine/pharmacology , Ketamine/administration & dosage , Sevoflurane/pharmacology , Sevoflurane/administration & dosage , Dexmedetomidine/pharmacology , Electroencephalography/drug effects , Electroencephalography/methods , Propofol/pharmacology , Propofol/administration & dosage , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Reflex, Righting/drug effects , Reflex, Righting/physiology , Mice, Inbred C57BL , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/administration & dosage , Anesthesia/methods
14.
Biomed Pharmacother ; 175: 116693, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701566

ABSTRACT

Sevoflurane postconditioning has been shown to provide neuroprotection against cerebral hypoxia-ischemia injury, but the mechanisms remain elusive. Microtubule-associated protein 2 (MAP2) is implicated in early neuronal hypoxia-ischemia injury. This study aimed to investigate whether the neuroprotective effects of sevoflurane postconditioning are related to the Akt/GSK-3ß pathway and its downstream target MAP2 in zebrafish hypoxia/reoxygenation (H/R) model. Sevoflurane postconditioning or GSK-3ß inhibitor TDZD-8 were used to treat H/R zebrafish. The cerebral infarction, neuronal apoptosis, and mitochondrial changes were evaluated using TTC staining, TUNEL staining, and transmission electron microscopy, respectively. The distribution of MAP2 in the brain was determined by immunofluorescence imaging. The levels of Akt, p-Akt, GSK-3ß, p-GSK-3ß, and MAP2 proteins were evaluated by Western blotting. The neurobehavioral recovery of zebrafish was assessed based on optokinetic response behavior. Our results indicated that sevoflurane postconditioning and TDZD-8 significantly reduced the cerebral infarction area, suppressed cell apoptosis, and improved mitochondrial integrity in zebrafish subjected to H/R. Furthermore, sevoflurane postconditioning and TDZD-8 elevated the ratios of p-Akt/Akt and p-GSK-3ß/GSK-3ß. However, the neuroprotective effect of sevoflurane postconditioning was effectively abolished upon suppression of MAP2 expression. In conclusion, sevoflurane postconditioning ameliorated cerebral H/R injury and facilitated the restoration of neurobehavioral function through the activation of Akt/GSK-3ß pathway and promotion of MAP2 expression.


Subject(s)
Glycogen Synthase Kinase 3 beta , Microtubule-Associated Proteins , Neuroprotective Agents , Proto-Oncogene Proteins c-akt , Sevoflurane , Signal Transduction , Zebrafish , Animals , Sevoflurane/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Microtubule-Associated Proteins/metabolism , Apoptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Ischemic Postconditioning/methods , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Zebrafish Proteins/metabolism , Disease Models, Animal , Mitochondria/drug effects , Mitochondria/metabolism , Male
15.
Neurol Res ; 46(7): 593-604, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38747300

ABSTRACT

BACKGROUND: Sevoflurane treatment increases the incidence of postoperative cognitive dysfunction (POCD), and patients with POCD show a decline in cognitive abilities compared to preoperative levels. OBJECTIVES: This study aimed to investigate whether the activation of α7 nicotinic acetylcholine receptor (α7nAChR) and the expression of M1 acetylcholine receptor (mAChR M1) in the hippocampus affects the cognitive function of aged rats. METHODS: Forty-eight Sprague-Dawley (SD) rats of 1-week- and 12-months-old were divided into eight groups: four groups for α7nAChR and four groups for mAChR M1, respectively. All SD rats received 1.0-02% sevoflurane for α7nAChR and 1.0-02% sevoflurane for mAChR M1 for 2-6 h, respectively. The Y-maze test was used to assess the ability to learn and memory after receiving sevoflurane for 7 days at the same moment portion. RT-PCR was used to determine the expression of α7nAChR and mAChR M1 in the hippocampus of rats. RESULTS: The α7nAChR mitigated the formation of sevoflurane-induced memory impairment by modulating the translocation of NR2B from the intracellular reservoir to the cell surface reservoir within the hippocampus. Next, sevoflurane-induced decline of cognitive function and significantly decreased mAChR M1 expression at mRNA levels. CONCLUSION: α7nAChR regulates the trafficking of NR2B in the hippocampus of rats via the Src-family tyrosine kinase (SFK) pathway. This regulation is associated with cognitive deficits induced by sevoflurane in hippocampal development. Sevoflurane affects the cognitive function of rats by suppressing the mAChR M1 expression at mRNA levels in the hippocampus.


α7nAChR attenuates sevoflurane-induced memory deficits by regulating NR2B.α7nAChR controls NR2B via the SFK in the hippocampus of rats that contribute to sevoflurane-induced cognitive deficits.Sevoflurane may affect cognitive function in rats by suppressing the mAChR M1 expression at the mRNA levels in the hippocampus.Dysregulation of the α7nAChR and mAChR M1 receptors may contribute to cognitive deficits and neurodegenerative disorders.


Subject(s)
Hippocampus , Rats, Sprague-Dawley , Receptor, Muscarinic M1 , Sevoflurane , alpha7 Nicotinic Acetylcholine Receptor , Animals , Sevoflurane/pharmacology , Sevoflurane/adverse effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , Hippocampus/metabolism , Hippocampus/drug effects , Male , Receptor, Muscarinic M1/metabolism , Aging/drug effects , Aging/metabolism , Rats , Maze Learning/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/adverse effects , Disease Models, Animal
16.
Med Sci Monit ; 30: e945269, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38808453

ABSTRACT

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Haijin Huang, Cuicui Hu, Lin Xu, Xiaoping Zhu, Lili Zhao, Jia Min. The Effects of Hesperidin on Neuronal Apoptosis and Cognitive Impairment in the Sevoflurane Anesthetized Rat are Mediated Through the PI3/Akt/PTEN and Nuclear Factor-kappaB (NF-kappaB) Signaling Pathways. Med Sci Monit, 2020; 26: e920522. DOI: 10.12659/MSM.920522.


Subject(s)
Apoptosis , Cognitive Dysfunction , Hesperidin , NF-kappa B , Neurons , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Sevoflurane , Signal Transduction , Animals , Sevoflurane/pharmacology , Apoptosis/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , PTEN Phosphohydrolase/metabolism , Neurons/drug effects , Neurons/metabolism , Cognitive Dysfunction/metabolism , Rats , Hesperidin/pharmacology , Male , Phosphatidylinositol 3-Kinases/metabolism
17.
Res Vet Sci ; 172: 105254, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582048

ABSTRACT

Two randomized crossover trials evaluated the effects of nicardipine constant rate infusion (CRI) on 1) the anesthetic potency of sevoflurane and 2) the ability to attenuate dexmedetomidine-induced cardiovascular depression in anesthetized dogs. First, six healthy Beagle dogs weighing 11.7 ± 0.9 kg were allocated to one of three treatments that administered a CRI of carrier (saline) or dexmedetomidine 0.5 or 3.0 µg/kg/h following a loading dose. The minimum alveolar concentration (MAC) of sevoflurane was determined utilizing electric stimuli before and after the loading dose of nicardipine (20 µg/kg intravenously for 10 min), followed by CRI at 40 µg/kg/h with 60 min of equilibration. Subsequently, cardiovascular and blood gas variables were evaluated in another trial under sevoflurane anesthesia at the individual 1.5 MAC. After baseline measurements, the dogs were assigned to two treatments (dexmedetomidine CRI at 0.5 or 3.0 µg/kg/h following a loading dose) with sevoflurane doses adjusted to 1.5 times of MAC equivalent, and the measurements were repeated every 15 min for 120 min. After 60 min, nicardipine CRI at 40 µg/kg/h with a loading dose was added to the dexmedetomidine CRI. Dexmedetomidine infusions significantly decreased the sevoflurane MAC but nicardipine did not significantly alter the MAC either with or without dexmedetomidine CRI in dogs. Dexmedetomidine dose-dependently decreased the cardiac index and increased the systemic vascular resistance index; these effects were fully counteracted by concomitant nicardipine CRI. Nicardipine CRI can be useful for controlling the cardiovascular depression elicited by dexmedetomidine in anesthetized dogs without affecting the anesthetic potency of sevoflurane.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Nicardipine , Sevoflurane , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/administration & dosage , Dogs , Sevoflurane/pharmacology , Sevoflurane/administration & dosage , Nicardipine/pharmacology , Nicardipine/administration & dosage , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Male , Cross-Over Studies , Female , Pulmonary Alveoli/drug effects , Infusions, Intravenous/veterinary , Heart Rate/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Blood Pressure/drug effects
18.
Cell Signal ; 119: 111195, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38688381

ABSTRACT

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.


Subject(s)
Membrane Proteins , Microglia , Necroptosis , Neurons , Nucleotidyltransferases , Sevoflurane , Signal Transduction , Microglia/metabolism , Microglia/drug effects , Animals , Signal Transduction/drug effects , Sevoflurane/pharmacology , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Necroptosis/drug effects , Neurons/metabolism , Neurons/drug effects , Nucleotidyltransferases/metabolism , Cell Line , Tumor Necrosis Factor-alpha/metabolism
19.
Medicine (Baltimore) ; 103(16): e37552, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640331

ABSTRACT

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.


Subject(s)
Anesthetics, Inhalation , Propofol , Child , Infant , Humans , Sevoflurane/pharmacology , Remifentanil , Anesthetics, Inhalation/pharmacology , Evoked Potentials, Motor/physiology , Anesthesia, General , Anesthetics, Intravenous/pharmacology
20.
Medicine (Baltimore) ; 103(16): e37699, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640306

ABSTRACT

RATIONALE: Malignant hyperthermia (MH) is a rare yet serious medical complication that typically arises following general anesthesia or the administration of specific anesthetics. Due to the infrequency of MH, anesthesiologists often lack sufficient expertise in identifying and managing it, leading to misdiagnosis and inappropriate treatment. There is an urgent need to enhance the diagnosis and management of MH through the utilization of relevant tools. PATIENT CONCERNS: In this case, a 52-year-old woman underwent radical cervical cancer surgery under general anesthesia, with no family or significant medical history. She experienced a gradual increase in end-tidal carbon dioxide (ETCO2) to a maximum of 75 mm Hg and a rise in body temperature from 36.5 to 37.5 °C in a very short period, as well as a blood gas analysis showing a pH of 7.217. DIAGNOSIS: The anesthesiologist immediately used The WeChat applet-based National Remote Emergency System for Malignant Hyperthermia (MH-NRES), and the score was 40, which indicated that the patient was very likely to have MH. INTERVENTIONS: We immediately discontinued sevoflurane and switched total intravenous anesthesia to maintain general anesthesia, with a rapid intravenous infusion of dantrolene sodium. OUTCOMES: The ETCO2 and the temperature quickly dropped to normal, followed by successful completion of the surgery, and the patient was discharged 8 days after surgery. LESSONS: The experience can provide a basis use of MH-NRES and improve the ability of anesthesiologists to deal with intraoperative MH as well as increase the survival probability of patients.


Subject(s)
Malignant Hyperthermia , Uterine Cervical Neoplasms , Female , Humans , Middle Aged , Malignant Hyperthermia/diagnosis , Malignant Hyperthermia/etiology , Malignant Hyperthermia/therapy , Uterine Cervical Neoplasms/surgery , Uterine Cervical Neoplasms/complications , Dantrolene/therapeutic use , Sevoflurane , Anesthesia, General/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...