Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Mol Med ; 30(1): 39, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38493090

ABSTRACT

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


Subject(s)
Anesthetics, Inhalation , Cognitive Dysfunction , Methyl Ethers , Aged , Animals , Humans , Infant , Rats , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/metabolism , Cognition , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Methyl Ethers/pharmacology , Methyl Ethers/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sevoflurane/metabolism , Sevoflurane/pharmacology , tau Proteins/metabolism , Fibroblast Growth Factor 2/metabolism
2.
Synapse ; 78(1): e22286, 2024 01.
Article in English | MEDLINE | ID: mdl-38287474

ABSTRACT

In this study, we aimed to validate the hypothesis that the interplay between sevoflurane, oxidative stress and ferroptosis is crucial for the pathogenesis of sevoflurane-induced cognitive impairment in aged individuals. The mice with sevoflurane-induced cognitive impairment were used to explore the effects of sevoflurane on oxidative stress, iron homeostasis, and cognitive function in aged mice. Iron content and oxidative stress markers were analyzed in hippocampal tissue homogenates using specific assays. Additionally, the levels of iron death-related markers (Fth1 and Gpx4) were assessed by real-time PCR and Western blotting. Morris Water Maze and novel object recognition (NOR) tests were conducted to evaluate cognitive function. Sevoflurane exposure in aged mice resulted in a significant increase in iron overloading in the hippocampus, followed by a subsequent stabilization. Oxidative stress levels were elevated in the hippocampal tissue of sevoflurane-exposed mice, and a significant correlation was observed between iron death and oxidative stress. Liproxstatin-1, a ferroptosis inhibitor, effectively ameliorated the decline in memory and learning abilities induced by sevoflurane anesthesia. Liproxstatin-1 treatment reduced iron overload and oxidative stress in the hippocampal tissue of aged mice. The expression of Fth1 and Gpx4, iron death-related markers, was downregulated following Liproxstatin-1 intervention. Our findings suggest that sevoflurane anesthesia disrupts iron homeostasis, leading to increased oxidative stress and cognitive impairment in aged mice. These results highlight the potential of targeting iron-mediated processes to mitigate sevoflurane-induced cognitive impairment in the aging population.


Subject(s)
Anesthesia , Cognitive Dysfunction , Ferroptosis , Quinoxalines , Spiro Compounds , Animals , Mice , Sevoflurane/adverse effects , Sevoflurane/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Oxidative Stress , Anesthesia/adverse effects , Cognition , Iron/adverse effects , Iron/metabolism , Hippocampus/metabolism
3.
Adv Sci (Weinh) ; 11(10): e2305989, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38145349

ABSTRACT

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


Subject(s)
Microglia , Neurodegenerative Diseases , Mice , Animals , Sevoflurane/metabolism , Sevoflurane/pharmacology , Microglia/metabolism , Lipid Metabolism , Biomimetics , Signal Transduction , Neurodegenerative Diseases/metabolism
4.
BMC Med ; 21(1): 510, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38129829

ABSTRACT

BACKGROUND: Exposure to general anesthesia influences neuronal functions during brain development. Recently, interneurons were found to be involved in developmental neurotoxicity by anesthetic exposure. But the underlying mechanism and long-term consequences remain elusive. METHODS: Pregnant mice received 2.5% sevoflurane for 6-h on gestational day 14.5. Pentylenetetrazole (PTZ)-induced seizure, anxiety- and depression-like behavior tests were performed in 30- and 60-day-old male offspring. Cortical interneurons were labeled using Rosa26-EYFP/-; Nkx2.1-Cre mice. Immunofluorescence and electrophysiology were performed to determine the cortical interneuron properties. Q-PCR and in situ hybridization (ISH) were performed for the potential mechanism, and the finding was further validated by in utero electroporation (IUE). RESULTS: In this study, we found that maternal sevoflurane exposure increased epilepsy susceptibility by using pentylenetetrazole (PTZ) induced-kindling models and enhanced anxiety- and depression-like behaviors in adolescent offspring. After sevoflurane exposure, the highly ordered cortical interneuron migration was disrupted in the fetal cortex. In addition, the resting membrane potentials of fast-spiking interneurons in the sevoflurane-treated group were more hyperpolarized in adolescence accompanied by an increase in inhibitory synapses. Both q-PCR and ISH indicated that CXCL12/CXCR4 signaling pathway downregulation might be a potential mechanism under sevoflurane developmental neurotoxicity which was further confirmed by IUE and behavioral tests. Although the above effects were obvious in adolescence, they did not persist into adulthood. CONCLUSIONS: Our findings demonstrate that maternal anesthesia impairs interneuron migration through the CXCL12/CXCR4 signaling pathway, and influences the interneuron properties, leading to the increased epilepsy susceptibility in adolescent offspring. Our study provides a novel perspective on the developmental neurotoxicity of the mechanistic link between maternal use of general anesthesia and increased susceptibility to epilepsy.


Subject(s)
Epilepsy , Pentylenetetrazole , Humans , Pregnancy , Female , Mice , Animals , Male , Sevoflurane/metabolism , Sevoflurane/pharmacology , Pentylenetetrazole/toxicity , Pentylenetetrazole/metabolism , Maternal Exposure/adverse effects , Interneurons/metabolism , Epilepsy/chemically induced
5.
BMC Gastroenterol ; 23(1): 279, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37568083

ABSTRACT

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


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Wnt Signaling Pathway/genetics , Sevoflurane/pharmacology , Sevoflurane/metabolism , beta Catenin/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Cell Proliferation/genetics , Bile Ducts, Intrahepatic , Bile Duct Neoplasms/pathology , Cell Movement , Apoptosis/genetics
6.
Toxicol Sci ; 195(2): 257-269, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37494465

ABSTRACT

Anesthesia with sevoflurane contributes to perioperative neurocognitive disorder (PND), which is characterized by the deficiency in study and memory. T-Box transcription factor 2 (Tbx2), which is involved in the development of hippocampus neurons, was upregulated in the hippocampus of rats exposed to sevoflurane. Our study aimed to explore the role of Tbx2 in sevoflurane-induced cognitive disorder and hippocampus neuron damages. The expression of Tbx2 in hippocampus was upregulated after sevoflurane exposure, which was accompanied by the accumulation of reactive oxygen species and lipid peroxidation, as well as the loss of neurons in hippocampus. In vitro, silencing Tbx2 suppressed oxidative stress and ferroptosis induced by sevoflurane, whereas exogenous overexpression of Tbx2 exacerbated these processes. Importantly, Tbx2 knockdown improved sevoflurane-induced cognitive disorder in aged rats, as evidenced by the increases in behavioral indexes. Mechanistically, the expression of brain-derived neurotrophic factor (BDNF), as well as the downstream nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling, was repressed by Tbx2. Mimicking the activation of BDNF with 7,8-dihydroxyflavone rescued the effects of Tbx2 overexpression on oxidative stress and ferroptosis in vitro, indicating that the BDNF/Nrf2/HO-1 signaling may mediate the role of Tbx2 in sevoflurane-induced cognitive disorder and neuron damages. In summary, Tbx2 may contribute to neuronal damages via enhancing the oxidative stress and ferroptosis caused by sevoflurane. BDNF/Nrf2/HO-1 signaling mediates the role of Tbx2 in sevoflurane-induced cognitive disorder. Knockdown of Tbx2 improves sevoflurane-induced cognitive impairment. Our finding provides a novel insight for PND treatment.


Subject(s)
Cognitive Dysfunction , Ferroptosis , Rats , Animals , Sevoflurane/toxicity , Sevoflurane/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Rats, Sprague-Dawley , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Neurons , Hippocampus
7.
Exp Biol Med (Maywood) ; 248(7): 641-655, 2023 04.
Article in English | MEDLINE | ID: mdl-37309741

ABSTRACT

General anesthetics are potent neurotoxins when given during early development, causing apoptotic deletion of substantial number of neurons and persistent neurocognitive and behavioral deficits in animals and humans. The period of intense synaptogenesis coincides with the peak of susceptibility to deleterious effects of anesthetics, a phenomenon particularly pronounced in vulnerable brain regions such as subiculum. With steadily accumulating evidence confirming that clinical doses and durations of anesthetics may permanently alter the physiological trajectory of brain development, we set out to investigate the long-term consequences on dendritic morphology of subicular pyramidal neurons and expression on genes regulating the complex neural processes such as neuronal connectivity, learning, and memory. Using a well-established model of anesthetic neurotoxicity in rats and mice neonatally exposed to sevoflurane, a volatile general anesthetic commonly used in pediatric anesthesia, we report that a single 6 h of continuous anesthesia administered at postnatal day (PND) 7 resulted in lasting dysregulation in subicular mRNA levels of cAMP responsive element modulator (Crem), cAMP responsive element-binding protein 1 (Creb1), and Protein phosphatase 3 catalytic subunit alpha, a subunit of calcineurin (Ppp3ca) (calcineurin) when examined during juvenile period at PND28. Given the critical role of these genes in synaptic development and neuronal plasticity, we deployed a set of histological measurements to investigate the implications of anesthesia-induced dysregulation of gene expression on morphology and complexity of surviving subicular pyramidal neurons. Our results indicate that neonatal exposure to sevoflurane induced lasting rearrangement of subicular dendrites, resulting in higher orders of complexity and increased branching with no significant effects on the soma of pyramidal neurons. Correspondingly, changes in dendritic complexity were paralleled by the increased spine density on apical dendrites, further highlighting the scope of anesthesia-induced dysregulation of synaptic development. We conclude that neonatal sevoflurane induced persistent genetic and morphological dysregulation in juvenile rodents, which could indicate heightened susceptibility toward cognitive and behavioral disorders we are beginning to recognize as sequelae of early-in-life anesthesia.


Subject(s)
Anesthetics, Inhalation , Methyl Ethers , Humans , Child , Animals , Rats , Mice , Sevoflurane/toxicity , Sevoflurane/metabolism , Calcineurin/metabolism , Calcineurin/pharmacology , Animals, Newborn , Anesthetics, Inhalation/toxicity , Methyl Ethers/toxicity , Hippocampus/metabolism
8.
J Transl Med ; 21(1): 397, 2023 06 18.
Article in English | MEDLINE | ID: mdl-37331963

ABSTRACT

BACKGROUND: Preclinical studies in acute respiratory distress syndrome (ARDS) have suggested that inhaled sevoflurane may have lung-protective effects and clinical trials are ongoing to assess its impact on major clinical outcomes in patients with ARDS. However, the underlying mechanisms of these potential benefits are largely unknown. This investigation focused on the effects of sevoflurane on lung permeability changes after sterile injury and the possible associated mechanisms. METHODS: To investigate whether sevoflurane could decrease lung alveolar epithelial permeability through the Ras homolog family member A (RhoA)/phospho-Myosin Light Chain 2 (Ser19) (pMLC)/filamentous (F)-actin pathway and whether the receptor for advanced glycation end-products (RAGE) may mediate these effects. Lung permeability was assessed in RAGE-/- and littermate wild-type C57BL/6JRj mice on days 0, 1, 2, and 4 after acid injury, alone or followed by exposure at 1% sevoflurane. Cell permeability of mouse lung epithelial cells was assessed after treatment with cytomix (a mixture of TNFɑ, IL-1ß, and IFNγ) and/or RAGE antagonist peptide (RAP), alone or followed by exposure at 1% sevoflurane. Levels of zonula occludens-1, E-cadherin, and pMLC were quantified, along with F-actin immunostaining, in both models. RhoA activity was assessed in vitro. RESULTS: In mice after acid injury, sevoflurane was associated with better arterial oxygenation, decreased alveolar inflammation and histological damage, and non-significantly attenuated the increase in lung permeability. Preserved protein expression of zonula occludens-1 and less increase of pMLC and actin cytoskeletal rearrangement were observed in injured mice treated with sevoflurane. In vitro, sevoflurane markedly decreased electrical resistance and cytokine release of MLE-12 cells, which was associated with higher protein expression of zonula occludens-1. Improved oxygenation levels and attenuated increase in lung permeability and inflammatory response were observed in RAGE-/- mice compared to wild-type mice, but RAGE deletion did not influence the effects of sevoflurane on permeability indices after injury. However, the beneficial effect of sevoflurane previously observed in wild-type mice on day 1 after injury in terms of higher PaO2/FiO2 and decreased alveolar levels of cytokines was not found in RAGE-/- mice. In vitro, RAP alleviated some of the beneficial effects of sevoflurane on electrical resistance and cytoskeletal rearrangement, which was associated with decreased cytomix-induced RhoA activity. CONCLUSIONS: Sevoflurane decreased injury and restored epithelial barrier function in two in vivo and in vitro models of sterile lung injury, which was associated with increased expression of junction proteins and decreased actin cytoskeletal rearrangement. In vitro findings suggest that sevoflurane may decrease lung epithelial permeability through the RhoA/pMLC/F-actin pathway.


Subject(s)
Actins , Respiratory Distress Syndrome , Animals , Mice , Sevoflurane/pharmacology , Sevoflurane/metabolism , Sevoflurane/therapeutic use , Actins/metabolism , Receptor for Advanced Glycation End Products/metabolism , Mice, Inbred C57BL , Lung/pathology , Respiratory Distress Syndrome/pathology , Cytokines/metabolism , Permeability , Models, Theoretical
9.
J Neuroimmunol ; 379: 578070, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37148600

ABSTRACT

Microglia-mediated neuroinflammatory responses play a key role in perioperative neurocognitive disorders (PND). Triggering receptor expressed on myeloid cells-1 (TREM1) has been shown to be a key regulator of inflammation. However, its role in PND remains largely unknown. This study aimed to evaluate the role of TREM1 in sevoflurane-induced PND. We applied AAV knockdown TREM1 in hippocampal microglia in aging mice. The mice were then subjected to neurobehavioral and biochemical testing after the intervention of sevoflurane. We found that sevoflurane inhalation can cause PND in mice, increase hippocampal TREM1 expression, polarize microglia to M1 type, upregulate TNF-α and IL-1ß expression (pro-inflammatory), and inhibit TGF-ß and IL-10 expression (anti-inflammatory). Knocking down TREM1 can improve sevoflurane-induced cognitive dysfunction, reduce M1 type marker iNOS, and increase M2 type marker ARG, improving the neuroinflammation. TREM1 is a target for sevoflurane-induced PND prevention.


Subject(s)
Inflammation , Microglia , Mice , Animals , Microglia/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Sevoflurane/adverse effects , Sevoflurane/metabolism , Inflammation/metabolism , Neurocognitive Disorders/metabolism
10.
Neuropathology ; 43(6): 431-440, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37037475

ABSTRACT

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


Subject(s)
Cognitive Dysfunction , Methyl Ethers , Animals , Mice , Animals, Newborn , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Hippocampus , Methyl Ethers/adverse effects , Methyl Ethers/metabolism , Mice, Inbred C57BL , PPAR gamma/metabolism , PPAR gamma/pharmacology , Sevoflurane/adverse effects , Sevoflurane/metabolism
11.
J Pharmacol Exp Ther ; 385(2): 135-145, 2023 05.
Article in English | MEDLINE | ID: mdl-36828631

ABSTRACT

The purpose of this study was to investigate the effects of the volatile anesthetic agents isoflurane and sevoflurane, at clinically relevant concentrations, on the fluidity of lipid membranes and permeability of the blood-brain barrier (BBB). We analyzed the in vitro effects of isoflurane or ketamine using erythrocyte ghosts (sodium fluorescein permeability), monolayers of brain microvascular endothelial cells ([13C]sucrose and fluorescein permeability), or liposomes (fluorescence anisotropy). Additionally, we determined the effects of 30-minute exposure of mice to isoflurane on the brain tight junction proteins. Finally, we investigated in vivo brain uptake of [13C]mannitol and [13C]sucrose after intravenous administration in mice under anesthesia with isoflurane, sevoflurane, or ketamine/xylazine in addition to the awake condition. Isoflurane at 1-mM and 5-mM concentrations increased fluorescein efflux from the erythrocyte ghosts in a concentration-dependent manner. Similarly, in endothelial cell monolayers exposed to 3% (v/v) isoflurane, permeability coefficients rose by about 25% for fluorescein and 40% for [13C]sucrose, whereas transendothelial resistance and cell viability remained unaffected. Although isoflurane caused a significant decrease in liposomes anisotropy values, ketamine/xylazine did not show any effects. Brain uptake clearance (apparent Kin) of the passive permeability markers in vivo in mice approximately doubled under isoflurane or sevoflurane anesthesia compared with either ketamine/xylazine anesthesia or the awake condition. In vivo exposure of mice to isoflurane did not change any of the brain tight junction proteins. Our data support membrane permeabilization rather than loosening of intercellular tight junctions as an underlying mechanism for increased permeability of the endothelial cell monolayers and the BBB in vivo. SIGNIFICANCE STATEMENT: The blood-brain barrier controls the entry of endogenous substances and xenobiotics from the circulation into the central nervous system. Volatile anesthetic agents like isoflurane alter the lipid structure of cell membranes, transiently facilitating the brain uptake of otherwise poorly permeable, hydrophilic small molecules. Clinical implications may arise when potentially neurotoxic drugs gain enhanced access to the central nervous system under inhalational anesthetics.


Subject(s)
Anesthetics, Inhalation , Anesthetics , Isoflurane , Ketamine , Mice , Animals , Isoflurane/pharmacology , Blood-Brain Barrier/metabolism , Sevoflurane/metabolism , Sevoflurane/pharmacology , Endothelial Cells/metabolism , Xylazine/metabolism , Xylazine/pharmacology , Liposomes , Anesthetics/pharmacology , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/metabolism , Tight Junctions/metabolism , Permeability , Tight Junction Proteins/metabolism , Fluoresceins , Lipids
12.
Cardiovasc Toxicol ; 23(2): 86-92, 2023 02.
Article in English | MEDLINE | ID: mdl-36800141

ABSTRACT

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


Subject(s)
Ferroptosis , Myocytes, Cardiac , Animals , Rats , Sevoflurane/metabolism , Sevoflurane/pharmacology , Hypoxia , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Homeostasis
13.
BMC Med ; 21(1): 7, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36600274

ABSTRACT

BACKGROUND: Perioperative neurocognitive disorders (PND) with a high incidence frequently occur in elderly surgical patients closely associated with prolonged anesthesia-induced neurotoxicity. The neuromorphopathological underpinnings of anesthesia-induced neurotoxicity have remained elusive. METHODS: Prolonged anesthesia with sevoflurane was used to establish the sevoflurane-induced neurotoxicity (SIN) animal model. Morris water maze, elevated plus maze, and open field test were employed to track SIN rats' cognitive behavior and anxiety-like behaviors. We investigated the neuropathological basis of SIN through techniques such as transcriptomic, electrophysiology, molecular biology, scanning electron microscope, Golgi staining, TUNEL assay, and morphological analysis. Our work further clarifies the pathological mechanism of SIN by depleting microglia, inhibiting neuroinflammation, and C1q neutralization. RESULTS: This study shows that prolonged anesthesia triggers activation of the NF-κB inflammatory pathway, neuroinflammation, inhibition of neuronal excitability, cognitive dysfunction, and anxiety-like behaviors. RNA sequencing found that genes of different types of synapses were downregulated after prolonged anesthesia. Microglial migration, activation, and phagocytosis were enhanced. Microglial morphological alterations were also observed. C1qa, the initiator of the complement cascade, and C3 were increased, and C1qa tagging synapses were also elevated. Then, we found that the "Eat Me" complement pathway mediated microglial synaptic engulfment in the hippocampus after prolonged anesthesia. Afterward, synapses were remarkably lost in the hippocampus. Furthermore, dendritic spines were reduced, and their genes were also downregulated. Depleting microglia ameliorated the activation of neuroinflammation and complement and rescued synaptic loss, cognitive dysfunction, and anxiety-like behaviors. When neuroinflammatory inhibition or C1q neutralization occurred, complement was also decreased, and synaptic elimination was interrupted. CONCLUSIONS: These findings illustrated that prolonged anesthesia triggered neuroinflammation and complement-mediated microglial synaptic engulfment that pathologically caused synaptic elimination in SIN. We have demonstrated the neuromorphopathological underpinnings of SIN, which have direct therapeutic relevance for PND patients.


Subject(s)
Anesthesia , Cognitive Dysfunction , Neuroinflammatory Diseases , Animals , Rats , Anesthesia/adverse effects , Anxiety/etiology , Anxiety/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Complement C1q/metabolism , Hippocampus/metabolism , Microglia/drug effects , Microglia/physiology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/complications , Sevoflurane/adverse effects , Sevoflurane/metabolism
14.
Cardiovasc Drugs Ther ; 37(6): 1053-1064, 2023 Dec.
Article in English | MEDLINE | ID: mdl-35171385

ABSTRACT

OBJECTIVE: As some articles have highlighted the role of microRNA-92a (miR-92a) in myocardial ischemia-reperfusion injury (MI/RI), this article aimed to investigate the effect of miR-92a on Sevoflurane (Sevo)-treated MI/RI via regulation of Krüppel-like factor 4 (KLF4). METHODS: An MI/RI rat model was established by ligating the left anterior descending coronary artery. The cardiac function, pathological changes of myocardial tissues, inflammatory response, oxidative stress and cardiomyocyte apoptosis in MI/RI rats were determined. KLF4 and miR-92a expression was detected in the myocardial tissue of rats, and the target relationship between miR-92a and KLF4 was confirmed. RESULTS: Sevo treatment alleviated myocardial damage, inflammatory response, oxidative stress response, and cardiomyocyte apoptosis, and improved cardiac function in MI/RI rats. miR-92a increased and KLF4 decreased in the myocardial tissue of MI/RI rats. KLF4 was targeted by miR-92a. Downregulation of miR-92a or upregulation of KLF4 further enhanced the effect of Sevo treatment on MI/RI. CONCLUSION: This study suggests that depletion of miR-92a promotes upregulation of KLF4 to improve cardiac function, reduce cardiomyocyte apoptosis and further enhance the role of Sevo treatment in alleviating MI/RI.


Subject(s)
MicroRNAs , Myocardial Reperfusion Injury , Rats , Animals , MicroRNAs/metabolism , Sevoflurane/pharmacology , Sevoflurane/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Kruppel-Like Factor 4 , Myocardium/pathology , Myocytes, Cardiac , Apoptosis
15.
Cereb Cortex ; 33(5): 1955-1971, 2023 02 20.
Article in English | MEDLINE | ID: mdl-35584785

ABSTRACT

Sevoflurane exposure in the neonatal period causes long-term developmental neuropsychological dysfunction, including memory impairment and anxiety-like behaviors. However, the molecular mechanisms underlying such effects have not been fully elucidated. In this study, we investigated the effect of neonatal exposure to sevoflurane on neurobehavioral profiles in adolescent rats, and applied an integrated approach of lipidomics and proteomics to investigate the molecular network implicated in neurobehavioral dysfunction. We found that neonatal exposure to sevoflurane caused cognitive impairment and social behavior deficits in adolescent rats. Lipidomics analyses revealed that sevoflurane significantly remodeled hippocampal lipid metabolism, including lysophatidylcholine (LPC) metabolism, phospholipid carbon chain length and carbon chain saturation. Through a combined proteomics analysis, we found that neonatal exposure to sevoflurane significantly downregulated the expression of lysophosphatidylcholine acyltransferase 1 (LPCAT1), a key enzyme in the regulation of phospholipid metabolism, in the hippocampus of adolescent rats. Importantly, hippocampal LPCAT1 overexpression restored the dysregulated glycerophospholipid (GP) metabolism and alleviated the learning and memory deficits caused by sevoflurane. Collectively, our evidence that neonatal exposure to sevoflurane downregulates LPCAT1 expression and dysregulates GP metabolism in the hippocampus, which may contribute to the neurobehavioral dysfunction in the adolescent rats.


Subject(s)
Anesthetics, Inhalation , Animals , Rats , Sevoflurane/metabolism , Sevoflurane/pharmacology , Animals, Newborn , Anesthetics, Inhalation/pharmacology , Rats, Sprague-Dawley , Maze Learning , Memory Disorders/metabolism , Hippocampus/metabolism , Phospholipids/metabolism
16.
Pediatr Res ; 93(4): 838-844, 2023 03.
Article in English | MEDLINE | ID: mdl-35804157

ABSTRACT

BACKGROUND: The aim of the study was to determine the effects of repeated anesthesia exposure across postnatal development. METHODS: Seventy-two newborn Sprague-Dawley rats were randomly divided into Sev group and Con-aged group. Sev groups were exposed to 2.6% sevoflurane for 2 h on postnatal day (P) 7, P14, and P21; the Con groups only received carrier gas for 2 h. Learning and memory were evaluated using the MWM test at P31 (juvenile), P91 (adult), and 18 months postnatally (aged). The relative expression of APP and Mapt mRNA was detected by RT-PCR, while Aß, tau, and P-tau protein levels were analyzed by immunohistochemistry. RESULTS: After repeated inhalation of sevoflurane, MWM test performance was significantly decreased in the Sev-aged group compared to the Con-aged group (P > 0.05). The relative expression of APP and Mapt mRNA was not significantly different between groups in each growth period (P > 0.05). The tau expression in the juvenile hippocampal CA1, CA3, and dentate gyrus regions increased markedly in the Sev group, while P-tau only increased in the hippocampal CA3 region in the Sev-adult group. The expression of tau, P-tau, and Aß in the hippocampal regions was upregulated in the Sev-aged group. CONCLUSIONS: Multiple exposures to sevoflurane across postnatal development can induce or aggravate cognitive impairment in old age. IMPACT: Whether multiple sevoflurane exposures across postnatal development cause cognitive impairment in childhood, adulthood, or old age, as well as the relationship between sevoflurane and the hippocampal Aß, tau, and P-tau proteins, remains unknown. This study's results demonstrate that multiple exposures to sevoflurane across postnatal development do not appear to affect cognitive function in childhood and adulthood; however, multiple exposures may lead to a cognitive function deficit in old age. The underlying mechanism may involve overexpression of the tau, P-tau, and Aß proteins in the hippocampus.


Subject(s)
Anesthetics, Inhalation , Cognitive Dysfunction , Methyl Ethers , Rats , Animals , Sevoflurane/adverse effects , Sevoflurane/metabolism , Rats, Sprague-Dawley , Methyl Ethers/toxicity , Methyl Ethers/metabolism , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/metabolism , Maze Learning , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/psychology , Cognition , Hippocampus/metabolism
17.
J Clin Pharmacol ; 63(1): 91-104, 2023 01.
Article in English | MEDLINE | ID: mdl-35943164

ABSTRACT

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


Subject(s)
Cytochrome P-450 CYP2E1 , Hypnotics and Sedatives , Polymorphism, Single Nucleotide , Sevoflurane , Humans , Case-Control Studies , Cytochrome P-450 CYP2E1/metabolism , Genotype , Hypnotics and Sedatives/metabolism , Hypnotics and Sedatives/pharmacokinetics , Sevoflurane/metabolism , Sevoflurane/pharmacokinetics
18.
Int Immunopharmacol ; 113(Pt A): 109380, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36330914

ABSTRACT

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


Subject(s)
Lung Neoplasms , Prostatic Neoplasms , Male , Mice , Animals , Humans , Sevoflurane/pharmacology , Sevoflurane/metabolism , Interleukin-6/metabolism , Lung Neoplasms/pathology , Macrophages/metabolism , Prostatic Neoplasms/pathology
19.
Pharm Biol ; 60(1): 1915-1924, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36205592

ABSTRACT

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


Subject(s)
Iron Overload , Neurotoxicity Syndromes , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Chalcones , Cognition , Hippocampus , Iron Overload/metabolism , Male , NF-E2-Related Factor 2/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Oxidative Stress , Rats , Rats, Sprague-Dawley , Sevoflurane/metabolism , Sevoflurane/toxicity
20.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(8): 1108-1119, 2022 Aug 28.
Article in English, Chinese | MEDLINE | ID: mdl-36097779

ABSTRACT

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


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Sirtuin 3 , Adenosine Triphosphate/metabolism , Animals , Male , Mitochondria/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , NAD/metabolism , Rats , Rats, Sprague-Dawley , Sevoflurane/metabolism , Sirtuin 3/metabolism , Voltage-Dependent Anion Channel 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...