1
|
Lu D, Zhang W, Chen K, Feng X. Dual effects of GABA A R agonist anesthetics in neurodevelopment and vulnerable brains: From neurotoxic to therapeutic effects. Neural Regen Res 2026; 21:81-95. [PMID: 39665822 PMCID: PMC12094567 DOI: 10.4103/nrr.nrr-d-24-00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/30/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024] Open
Abstract
Debates regarding the specific effects of general anesthesia on developing brains have persisted for over 30 years. A consensus has been reached that prolonged, repeated, high-dose exposure to anesthetics is associated with a higher incidence of deficits in behavior and executive function, while single exposure has a relatively minor effect on long-term neurological function. In this review, we summarize the dose-dependent neuroprotective or neurotoxic effects of gamma-aminobutyric acid type A receptor agonists, a representative group of sedatives, on developing brains or central nervous system diseases. Most preclinical research indicates that anesthetics have neurotoxic effects on the developing brain through various signal pathways. However, recent studies on low-dose anesthetics suggest that they may promote neurodevelopment during this critical period. These findings are incomprehensible for the general "dose-effect" principles of pharmacological research, which has attracted researchers' interest and led to the following questions: What is the threshold for the dual effects exerted by anesthetics such as propofol and sevoflurane on the developing brain? To what extent can their protective effects be maximized? What are the underlying mechanisms involved in these effects? Consequently, this issue has essentially become a "mathematical problem." After summarizing the dose-dependent effects of gamma-aminobutyric acid type A receptor agonist sedatives in both the developing brain and the brains of patients with central nervous system diseases, we believe that all such anesthetics exhibit specific threshold effects unique to each drug. These effects range from neuroprotection to neurotoxicity, depending on different brain functional states. However, the exact values of the specific thresholds for different drugs in various brain states, as well as the underlying mechanisms explaining why these thresholds exist, remain unclear. Further in-depth exploration of these issues could significantly enhance the therapeutic translational value of these anesthetics.
Collapse
Affiliation(s)
- Dihan Lu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wen Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Keyu Chen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Wei Z, Liu C, Chen J, Yao Y, Qin D. Krüppel-like factor 4 transcription factor in blood-brain barrier endothelial cells: A potential role in Alzheimer's disease. Animal Model Exp Med 2025; 8:819-828. [PMID: 40135492 DOI: 10.1002/ame2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Alzheimer's disease is the most prevalent chronic neurodegenerative disorder worldwide, with no sufficient cure. Ongoing research is focused on developing new therapies aimed at preventing or delaying the onset of symptoms, slowing disease progression, and improving cognitive and behavioral outcomes in individuals affected by Alzheimer's disease. Among the various pathological changes associated with this condition, blood-brain barrier (BBB) leakage plays a crucial role as it serves as a vital boundary for maintaining central nervous system (CNS) health. Preserving the integrity and functionality of the BBB is essential to protect the brain from amyloid-β accumulation, neuroinflammation, and neuronal degeneration. This review summarizes models of Alzheimer's disease characterized by BBB leakage over time. More importantly, we introduce Krüppel-like factor 4 (KLF4), a transcription factor involved in vascular systems, and discuss its relevance to Alzheimer's disease. By elucidating the functions of KLF4 within both vascular and CNSs, this review highlights its potential role in modulating BBB integrity in Alzheimer's pathology, which may contribute to therapeutic strategies for managing this debilitating condition.
Collapse
Affiliation(s)
- Ziying Wei
- Medical Research Center, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunhua Liu
- Medical Research Center, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianyu Chen
- Medical Research Center, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuxiao Yao
- Medical Research Center, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dajiang Qin
- Medical Research Center, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Antonova VV, Silachev DN, Plotnikov EY, Pevzner IB, Ivanov ME, Boeva EA, Kalabushev SN, Yadgarov MY, Cherpakov RA, Grebenchikov OA, Kuzovlev AN. Positive Effects of Argon Inhalation After Traumatic Brain Injury in Rats. Int J Mol Sci 2024; 25:12673. [PMID: 39684384 DOI: 10.3390/ijms252312673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
The noble gas argon is one of the most promising neuroprotective agents for hypoxic-reperfusion injuries of the brain. However, its effect on traumatic injuries has been insufficiently studied. The aim of this study was to analyze the effect of the triple inhalation of the argon-oxygen mixture Ar 70%/O2 30% on physical and neurological recovery and the degree of brain damage after traumatic brain injury and to investigate the possible molecular mechanisms of the neuroprotective effect. The experiments were performed in male Wistar rats. A controlled brain injury model was used to investigate the effects of argon treatment and the underlying molecular mechanisms. The results of the study showed that animals with craniocerebral injuries that were treated with argon inhalation exhibited better physical recovery rates, better neurological status, and less brain damage. Argon treatment significantly reduced the expression of the proinflammatory markers TNFα and CD68 caused by TBI, increased the expression of phosphorylated protein kinase B (pAKT), and promoted the expression of the transcription factor Nrf2 in intact animals. Treatment with an argon-oxygen breathing mixture after traumatic brain injury has a neuroprotective effect by suppressing the inflammatory response and activating the antioxidant and anti-ischemic system.
Collapse
Affiliation(s)
- Viktoriya V Antonova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Denis N Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Irina B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Mikhail E Ivanov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ekaterina A Boeva
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Sergey N Kalabushev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Mikhail Ya Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Rostislav A Cherpakov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Oleg A Grebenchikov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Artem N Kuzovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| |
Collapse
|
4
|
Qiu Y, Zeng Y, Zhang C, Lv X, Ling Y, Si Y, Guo T, Ni Y, Zhang J, Xu C, Wang Z, Hu J. A ROS-responsive loaded desferoxamine (DFO) hydrogel system for traumatic brain injury therapy. Biomed Mater 2024; 19:025016. [PMID: 38215474 DOI: 10.1088/1748-605x/ad1dfd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/12/2024] [Indexed: 01/14/2024]
Abstract
Traumatic brain injury (TBI) produces excess iron, and increased iron accumulation in the brain leads to lipid peroxidation and reactive oxygen species (ROSs), which can exacerbate secondary damage and lead to disability and death. Therefore, inhibition of iron overload and oxidative stress has a significant role in the treatment of TBI. Functionalized hydrogels with iron overload inhibiting ability and of oxidative stress inhibiting ability will greatly contribute to the repair of TBI. Herein, an injectable, post-traumatic microenvironment-responsive, ROS-responsive hydrogel encapsulated with deferrioxamine mesylate (DFO) was developed. The hydrogel is rapidly formed via dynamic covalent bonding between phenylboronic acid grafted hyaluronic acid (HA-PBA) and polyvinyl alcohol (PVA), and phenylboronate bonds are used to respond to and reduce ROS levels in damaged brain tissue to promote neuronal recovery. The release of DFO from HA-PBA/PVA hydrogels in response to ROS further promotes neuronal regeneration and recovery by relieving iron overload and thus eradicating ROS. In the Feeney model of Sprague Dawley rats, HA-PBA/PVA/DFO hydrogel treatment significantly improved the behavior of TBI rats and reduced the area of brain contusion in rats. In addition, HA-PBA/PVA/DFO hydrogel significantly reduced iron overload to reduce ROS and could effectively promote post-traumatic neuronal recovery. Its effects were also explored, and notably, HA-PBA/PVA/DFO hydrogel can reduce iron overload as well as ROS, thus protecting neurons from death. Thus, this injectable, biocompatible and ROS-responsive drug-loaded hydrogel has great potential for the treatment of TBI. This work suggests a novel method for the treatment of secondary brain injury by inhibiting iron overload and the oxidative stress response after TBI.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yu Zeng
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Chun Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Xiaorui Lv
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yating Ling
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yu Si
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Tao Guo
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yinying Ni
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Jingwen Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Changgen Xu
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Ziyu Wang
- Health Clinical Laboratories, Health BioMed Co., Ltd, Ningbo, Zhejiang 315042, People's Republic of China
| | - Jiabo Hu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| |
Collapse
|
5
|
Zhao Q, Zhang J, Li H, Li H, Xie F. Models of traumatic brain injury-highlights and drawbacks. Front Neurol 2023; 14:1151660. [PMID: 37396767 PMCID: PMC10309005 DOI: 10.3389/fneur.2023.1151660] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
Traumatic brain injury (TBI) is the leading cause for high morbidity and mortality rates in young adults, survivors may suffer from long-term physical, cognitive, and/or psychological disorders. Establishing better models of TBI would further our understanding of the pathophysiology of TBI and develop new potential treatments. A multitude of animal TBI models have been used to replicate the various aspects of human TBI. Although numerous experimental neuroprotective strategies were identified to be effective in animal models, a majority of strategies have failed in phase II or phase III clinical trials. This failure in clinical translation highlights the necessity of revisiting the current status of animal models of TBI and therapeutic strategies. In this review, we elucidate approaches for the generation of animal models and cell models of TBI and summarize their strengths and limitations with the aim of exploring clinically meaningful neuroprotective strategies.
Collapse
Affiliation(s)
- Qinghui Zhao
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Jianhua Zhang
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Huige Li
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Hongru Li
- Zhumadian Central Hospital, Zhumadian, China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| |
Collapse
|
6
|
Wang QS, Xiao RJ, Peng J, Yu ZT, Fu JQ, Xia Y. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal KLF4 Alleviated Ischemic Stroke Through Inhibiting N6-Methyladenosine Modification Level of Drp1 by Targeting lncRNA-ZFAS1. Mol Neurobiol 2023; 60:3945-3962. [DOI: 10.1007/s12035-023-03301-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 03/06/2023] [Indexed: 04/03/2023]
|
7
|
Gu T, Xu C, Meng X, Gao D, Jiang G, Yin A, Liu Q, Zhang L. Sevoflurane Preconditioning Alleviates Posttraumatic Stress Disorder-Induced Apoptosis in the Hippocampus via the EZH2-Regulated Akt/mTOR Axis and Improves Synaptic Plasticity. J Mol Neurosci 2023; 73:225-236. [PMID: 36930428 DOI: 10.1007/s12031-023-02114-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a persistent and severe psychological and mental disorder resulting from experiences of serious trauma or stress and is suffered by many individuals. Previous studies have shown that pretreatment with sevoflurane is efficient in reducing the incidence of PTSD. However, we require a more comprehensive understanding of the specific mechanisms by which sevoflurane works. Enhancer of zeste homolog 2 (EZH2) has been reported to be regulated by sevoflurane, and to improve patient cognition. In this study, we aimed to explore the mechanisms of sevoflurane and the role of EZH2 in PTSD cases. We explored the effects of sevoflurane and EPZ-6438 (inhibitor of EZH2) on rat behavior, followed by an investigation of EZH2 mRNA and protein expression. The effects of sevoflurane and EZH2 on neuronal survival were assessed by western blotting and TUNEL staining, while western blotting was used to examine the expression of PSD95 and the AKT/mTOR proteins. Sevoflurane preconditioning restored EZH2 expression and significantly inhibited apoptosis by regulating phosphorylation of the AKT/mTOR pathway. Synaptic plasticity was also significantly improved. These results suggest that pretreatment with sevoflurane could play an important role in PTSD prevention by regulating EZH2 expression.
Collapse
Affiliation(s)
- Tingting Gu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chang Xu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaozhou Meng
- Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China
| | - Dapeng Gao
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guanghao Jiang
- Department of Anesthesiology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Anqi Yin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China. .,Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Green TRF, Murphy SM, Rowe RK. Comparisons of quantitative approaches for assessing microglial morphology reveal inconsistencies, ecological fallacy, and a need for standardization. Sci Rep 2022; 12:18196. [PMID: 36307475 PMCID: PMC9616881 DOI: 10.1038/s41598-022-23091-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2022] Open
Abstract
Microglial morphology is used to measure neuroinflammation and pathology. For reliable inference, it is critical that microglial morphology is accurately quantified and that results can be easily interpreted and compared across studies and laboratories. The process through which microglial morphology is quantified is a key methodological choice and little is known about how this choice may bias conclusions. We applied five of the most commonly used ImageJ-based methods for quantifying the microglial morphological response to a stimulus to identical photomicrographs and individual microglial cells isolated from these photomicrographs, which allowed for direct comparisons of results generated using these approaches. We found a lack of comparability across methods that analyzed full photomicrographs, with significant discrepancies in results among the five methods. Quantitative methods to analyze microglial morphology should be selected based on several criteria, and combinations of these methods may give the most biologically accurate representation of microglial morphology.
Collapse
Affiliation(s)
- Tabitha R. F. Green
- grid.134563.60000 0001 2168 186XDepartment of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ USA ,grid.266190.a0000000096214564Department of Integrative Physiology, University of Colorado, 2860 Wilderness Place, Boulder, CO 80301 USA
| | - Sean M. Murphy
- grid.134563.60000 0001 2168 186XDepartment of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ USA
| | - Rachel K. Rowe
- grid.134563.60000 0001 2168 186XDepartment of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ USA ,grid.266190.a0000000096214564Department of Integrative Physiology, University of Colorado, 2860 Wilderness Place, Boulder, CO 80301 USA
| |
Collapse
|
9
|
Zhu Y, Zhang M, Wang J, Wang Q. Knockdown of UAF1 alleviates sevoflurane-induced cognitive impairment and neurotoxicity in rats by inhibiting pro-inflammatory signaling and oxidative stress. J Toxicol Sci 2022; 47:349-357. [PMID: 36047109 DOI: 10.2131/jts.47.349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Evidence has shown that suppression of the activation of NLRP3 inflammasome could ameliorate surgery/sevoflurane (SEV)-induced post-operative cognitive dysfunction (POCD). However, the underlying mechanisms remain unclear. UAF1 acts as a binding partner of USP1, which inhibits the ubiquitination-mediated degradation of NLRP3, indicating that UAF1 may be implicated in POCD through regulating the NLRP3 inflammasome. Here, we studied the role of UAF1/NLRP3 in SEV-induced cognitive impairment and neurotoxicity in rats. Neonatal rats were randomly divided into control, SEV, SEV+AAV-shNC and SEV+AAV-shUAF1 (UAF1-downregulated) groups. Morris water maze (MWM) test was applied to assess cognitive impairment. TUNEL staining, qRT-PCR and ELISA were used to assess the apoptosis and inflammation markers, respectively. The levels of superoxide dismutase (SOD), catalase (CAT) and malondialdehyde (MDA) were quantified to determine oxidative stress. The results showed that SEV treatment led to significant cognitive impairment, increased apoptosis in hippocampal tissues, upregulation of MDA and inflammatory factors (TNF-α, IL-1β, IL-18), as well as a decrease in SOD and CAT levels. All of the above observations were reversed by UAF1 downregulation. Furthermore, depletion of UAF1 neutralized SEV-mediated increase in p-NLRP3, p-IκBα and p-p65 levels. Altogether, the current study demonstrated that knockdown of UAF1 could alleviate SEV-induced cognitive impairment and neurotoxicity in rats by inhibiting pro-inflammatory signaling and oxidative stress.
Collapse
Affiliation(s)
- Yingjun Zhu
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, China
| | - Min Zhang
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, China
| | - Jiayu Wang
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, China
| | - Qingxiu Wang
- Department of Anesthesiology, The Affiliated Shanghai East Hospital of Tongji University, China
| |
Collapse
|
10
|
Chen C, Zuo J, Zhang H. Sevoflurane Post-treatment Mitigates Oxygen-glucose Deprivationinduced Pyroptosis of Hippocampal Neurons by Regulating the Mafb/DUSP14 Axis. Curr Neurovasc Res 2022; 19:245-254. [PMID: 35927915 DOI: 10.2174/1567202619666220802104426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 06/18/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ischemic brain injury often results in irreversible pyroptosis of neurons. Sevoflurane (Sevo) post-treatment exerts an alleviative role in neuroinflammation. OBJECTIVES This work evaluated the mechanism of Sevo post-treatment in oxygen-glucose deprivation (OGD)-induced pyroptosis of rat hippocampal neurons. METHODS Rat hippocampal neuron cell line H19-7 cells were treated with OGD, followed by posttreatment of 2% Sevo. The expression patterns of Mafb ZIP Transcription Factor B (Mafb) and dual- specificity phosphatase 14 (DUSP14) were determined via quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting methods. H19-7 cell viability and the release of lactate dehydrogenase (LDH) were examined via the cell counting kit-8 and LDH assay kits. Levels of pyroptosis-related proteins and cytokines NOD-like receptor family, pyrin domain containing 3 (NLRP3), N-term cleaved Gasdermin-D (GSDMD-N), cleaved-caspase-1, interleukin (IL)-1β, and IL-18 were also examined. The binding relation between Mafb and the DUSP14 promoter was detected. Besides, the roles of Mafb/DUSP14 in OGD-induced pyroptosis of rat hippocampal neurons were investigated through functional rescue experiments. RESULTS Mafb and DUSP14 expression levels were decreased in OGD-induced hippocampal neurons. Sevo post-treatment up-regulated Mafb and DUSP14, facilitated H19-7 cell viability, inhibited LDH release, and reduced levels of NLRP3, GSDMD-N, cleaved-caspase-1, IL-1β, and IL-18. Mafb increased DUSP14 expression via binding to the DUSP14 promoter. Repressing Mafb or DUSP14 exacerbated pyroptosis of hippocampal neurons. CONCLUSION Sevo post-treatment increased Mafb and DUSP14 expressions, which repressed OGDinduced pyroptosis of hippocampal neurons.
Collapse
Affiliation(s)
- Chen Chen
- Department of Anesthesiology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, Guangdong 518036, P.R. China
| | - Jianwei Zuo
- Department of Sports Medicine, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, Guangdong 518036, P.R. China
| | - Huimei Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|