1
|
Tong J, Gao J, Qi Y, Gao Z, Wang Q, Liu Y, Yuan T, Ren M, Yang G, Li Z, Li J, Sun H, Zhao X, Leung YY, Mu Y, Xu J, Lu C, Peng S, Ge L. GABA AR-PPT1 palmitoylation homeostasis controls synaptic transmission and circuitry oscillation. Transl Psychiatry 2024; 14:488. [PMID: 39695089 DOI: 10.1038/s41398-024-03206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
The infantile neuronal ceroid lipofuscinosis, also called CLN1 disease, is a fatal neurodegenerative disease caused by mutations in the CLN1 gene encoding palmitoyl protein thioesterase 1 (PPT1). Identifying the depalmitoylation substrates of PPT1 is crucial for understanding CLN1 disease. In this study, we found that GABAAR, the critical synaptic protein essential for inhibitory neurotransmission, is a substrate of PPT1. PPT1 depalmitoylates GABAAR α1 subunit at Cystein-260, while binding to Cystein-165 and -179. Mutations of PPT1 or its GABAAR α1 subunit binding site enhanced inhibitory synaptic transmission and strengthened oscillations powers but disrupted phase coupling in CA1 region and impaired learning and memory in 1- to 2-months-old PPT1-deficient and Gabra1em1 mice. Our study highlights the critical role of PPT1 in maintaining GABAAR palmitoylation homeostasis and reveals a previously unknown molecular pathway in CLN1 diseases induced by PPT1 mutations.
Collapse
Affiliation(s)
- Jia Tong
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jingjing Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yawei Qi
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Ziyan Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Qianqian Wang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yang Liu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Tiangang Yuan
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Minglong Ren
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Guixia Yang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Zhaoyue Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jin Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Hongyuan Sun
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Xing Zhao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yeung-Yeung Leung
- Division of Brain Sciences, Imperial College Faculty of Medicine, Du Cane Road, London, UK
| | - Yonghui Mu
- Basic Medical College, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jiamin Xu
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Chengbiao Lu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
- He'nan International Joint Laboratory for Non-invasive Neural Modulation, Department of Physiology and Pathology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Shiyong Peng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Lihao Ge
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| |
Collapse
|
2
|
Jiang W, Zhang M, Cao R, Wang X, Zuo Y. Different ethanol exposure durations affect cytochrome P450 2E1-mediated sevoflurane metabolism in rat liver. BMC Anesthesiol 2024; 24:321. [PMID: 39256673 PMCID: PMC11384694 DOI: 10.1186/s12871-024-02704-5] [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: 03/03/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Chronic alcohol users often exhibit an increased minimum alveolar concentration (MAC) of sevoflurane, yet the specific mechanism remains unclear. It has been reported that ethanol exposure can upregulate the protein expression and enzyme activity of cytochrome P450 2E1 (CYP2E1). CYP2E1 is a key enzyme that converts 2-5% of sevoflurane into equimolar amounts of hexafluoroisopropanol (HFIP) and F-. This study aims to explore whether ethanol exposure could alter sevoflurane metabolism through CYP2E1 modulation, potentially explaining the increased MAC observed in alcohol users. METHODS Eighty adult male Sprague-Dawley (SD) rats were randomly divided into two groups and received either 50% ethanol (dose: 3 g/kg) or 0.9% saline twice daily by gavage. After 1, 2, 3, and 4 weeks of gavage, ten rats were randomly selected from each group to undergo 1-hour anesthesia with 2.3% sevoflurane. Blood samples were collected after anesthesia to measure the concentration of free HFIP using gas chromatography. Additionally, the left lobe tissue of the liver was collected for the analysis of CYP2E1 protein expression by Western blot and CYP2E1 enzyme activity by colorimetric assay. Correlations between these parameters were analyzed using Pearson's correlation. RESULTS In the ethanol group, CYP2E1 expression, activity, and the concentration of free HFIP were significantly higher at all time points compared to the control group (P < 0.05), except for protein expression in the first week (P > 0.05). Within-group comparisons indicated no significant changes in any of the parameters for the control group (P > 0.05). In the ethanol group, there was no difference in free HFIP concentration between the first and second weeks (P > 0.05), but a significant increase was observed in the third and fourth weeks (P < 0.01); protein expression and enzyme activity significantly varied over time, especially showing a notable increase from the first to the third and fourth weeks (P < 0.05). Correlation analysis revealed strong positive correlations between free HFIP concentration and CYP2E1 activity (r = 0.7898), free HFIP concentration and CYP2E1 expression (r = 0.8418), and CYP2E1 activity and expression (r = 0.8740), all with P < 0.001. CONCLUSIONS Ethanol exposure increased both the expression and enzymatic activity of CYP2E1, consequently enhancing the metabolism of sevoflurane.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Min Zhang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Rui Cao
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xinghao Wang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Youbo Zuo
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
3
|
Zhao T, Liu M, Gu C, Wang X, Wang Y. Retraction Note: Activation of c-Src tyrosine kinase mediated the degradation of occludin in ventilator-induced lung injury. Respir Res 2024; 25:55. [PMID: 38267926 PMCID: PMC10809482 DOI: 10.1186/s12931-024-02692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Affiliation(s)
- Tao Zhao
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Mengjie Liu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Changping Gu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Xin Wang
- Department of Anesthesiology, Jinan Fifth People's Hospital, Ji'nan, Shandong, China
| | - Yuelan Wang
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| |
Collapse
|
4
|
Lu P, Liang F, Dong Y, Xie Z, Zhang Y. Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration. Int J Mol Sci 2023; 24:ijms24076746. [PMID: 37047719 PMCID: PMC10095407 DOI: 10.3390/ijms24076746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
Collapse
Affiliation(s)
- Pan Lu
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
5
|
Salaün JP, Chagnot A, Cachia A, Poirel N, Datin-Dorrière V, Dujarrier C, Lemarchand E, Rolland M, Delalande L, Gressens P, Guillois B, Houdé O, Levard D, Gakuba C, Moyon M, Naveau M, Orliac F, Orliaguet G, Hanouz JL, Agin V, Borst G, Vivien D. Consequences of General Anesthesia in Infancy on Behavior and Brain Structure. Anesth Analg 2023; 136:240-250. [PMID: 36638508 DOI: 10.1213/ane.0000000000006233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND One in 7 children will need general anesthesia (GA) before the age of 3. Brain toxicity of anesthetics is controversial. Our objective was to clarify whether exposure of GA to the developing brain could lead to lasting behavioral and structural brain changes. METHODS A first study was performed in mice. The behaviors (fear conditioning, Y-maze, and actimetry) and brain anatomy (high-resolution magnetic resonance imaging) of 6- to 8-week-old Swiss mice exposed or not exposed to GA from 4 to 10 days old were evaluated. A second study was a complementary analysis from the preexisting APprentissages EXécutifs et cerveau chez les enfants d'âge scolaire (APEX) cohort to assess the replicability of our data in humans. The behaviors (behavior rating inventory of executive function, emotional control, and working memory score, Backward Digit Span, and Raven 36) and brain anatomy (high-resolution magnetic resonance imaging) were compared in 102 children 9 to 10 years of age exposed or not exposed to a single GA (surgery) during infancy. RESULTS The animal study revealed chronic exacerbated fear behavior in the adult mice (95% confidence interval [CI], 4-80; P = .03) exposed to postnatal GA; this was associated with an 11% (95% CI, 7.5-14.5) reduction of the periaqueductal gray matter (P = .046). The study in humans suggested lower emotional control (95% CI, 0.33-9.10; P = .06) and a 6.1% (95% CI, 4.3-7.8) reduction in the posterior part of the right inferior frontal gyrus (P = .019) in the children who had been exposed to a single GA procedure. CONCLUSIONS The preclinical and clinical findings of these independent studies suggest lasting effects of early life exposure to anesthetics on later emotional control behaviors and brain structures.
Collapse
Affiliation(s)
- Jean-Philippe Salaün
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Department of Anesthesiology and Critical Care Medicine, CHU Caen, Caen University Hospital, Caen, France
| | - Audrey Chagnot
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Arnaud Cachia
- Université de Paris, LaPsyDé, CNRS, Paris, France.,Institut Universitaire de France, Paris, France
| | - Nicolas Poirel
- Université de Paris, LaPsyDé, CNRS, Paris, France.,Institut Universitaire de France, Paris, France.,GIP Cyceron, Caen, France
| | - Valérie Datin-Dorrière
- Université de Paris, LaPsyDé, CNRS, Paris, France.,GIP Cyceron, Caen, France.,Department of Neonatology, CHU Caen, Caen University Hospital, Caen, France
| | - Cléo Dujarrier
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Eloïse Lemarchand
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Marine Rolland
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Department of Anesthesiology and Critical Care Medicine, CHU Caen, Caen University Hospital, Caen, France
| | | | | | | | - Olivier Houdé
- Université de Paris, LaPsyDé, CNRS, Paris, France.,Institut Universitaire de France, Paris, France.,GIP Cyceron, Caen, France
| | - Damien Levard
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Clément Gakuba
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Department of Anesthesiology and Critical Care Medicine, CHU Caen, Caen University Hospital, Caen, France
| | - Marine Moyon
- Université de Paris, LaPsyDé, CNRS, Paris, France
| | - Mikael Naveau
- CNRS, GIP Cyceron, Normandie Université, Caen, France
| | - François Orliac
- Université de Paris, LaPsyDé, CNRS, Paris, France.,GIP Cyceron, Caen, France
| | - Gilles Orliaguet
- Department of Pediatric Anesthesia and Intensive Care, Necker-Enfants Malades University Hospital, AP-HP, Centre - Université de Paris, France, Université de Paris, Paris, France
| | - Jean-Luc Hanouz
- Department of Anesthesiology and Critical Care Medicine, CHU Caen, Caen University Hospital, Caen, France.,Caen Normandy University, Unicaen, Caen, France
| | - Véronique Agin
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Grégoire Borst
- Université de Paris, LaPsyDé, CNRS, Paris, France.,Institut Universitaire de France, Paris, France
| | - Denis Vivien
- From the Normandie Universite UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Department of Clinical Research, CHU Caen, Caen University Hospital, Caen, France
| |
Collapse
|
6
|
Cai M, Gao X, Yu S. Tripartite motif 72 inhibits apoptosis and mitochondrial dysfunction in neural stem cells induced by anesthetic sevoflurane by activating PI3K/AKT pathway. CHINESE J PHYSIOL 2023; 66:36-42. [PMID: 36814155 DOI: 10.4103/cjop.cjop-d-22-00082] [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: 02/22/2023] Open
Abstract
Anesthetics exposure induces neurocognitive deficits during brain development and impairs self-renewal and differentiation of neural stem cells (NSCs). Tripartite motif 72 (TRIM72, also known as mitsugumin 53, MG53) is involved in tissue repair and plasma membrane damage repair. The neuroprotective effect of TRIM72 against sevoflurane-induced neurotoxicity of NSCs was investigated in this study. First, human NSCs were exposed to different concentrations of sevoflurane. Results showed that TRIM72 was downregulated in sevoflurane-treated NSCs. Exposure to sevoflurane reduced cell viability in NSCs. Second, sevoflurane-treated NSCs were stimulated with recombinant human TRIM72 (rhTRIM72). Treatment with rhTRIM72 enhanced the cell viability in sevoflurane-treated NSCs. Moreover, treatment with a rhTRIM72-attenuated sevoflurane-induced increase in caspase-3 activity in NSCs. Third, JC-1 aggregates were deceased and JC-1 monomer was increased in sevoflurane-treated NSCs, which were reversed by rhTRIM72. Furthermore, rhTRIM72 also weakened sevoflurane-induced decrease in superoxide dismutase and glutathione peroxidase and increase in malondialdehyde and reactive oxygen species in NSCs. Finally, reduced phosphorylation levels of protein kinase B (AKT) and phosphatidylinositol 3-kinase (PI3K) in sevoflurane-treated NSCs were upregulated by rhTRIM72. In conclusion, TRIM72 inhibited cell apoptosis and reduced the mitochondria membrane potential of sevoflurane-treated NSCs through activation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Minmin Cai
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiang Gao
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shenghui Yu
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
7
|
Lee JA, Bae DH, Choi WH, Cho CH, Bang YS, Yoo J. Effects of Sevoflurane Exposure on Fetal Brain Development Using Cerebral Organoids. J Mol Neurosci 2022; 72:2440-2450. [PMID: 36478139 DOI: 10.1007/s12031-022-02080-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
Sevoflurane is a safe and well-known inhaled anesthetic. Given that sevoflurane can be delivered to developing fetuses through the mother, it is critical to determine whether this agent affects fetal neurodevelopment. Recent research has sought to determine whether sevoflurane affects fetal brain development when the mother is exposed during the second to third trimester of pregnancy, considered to be the crucial period for the development of nervous system. However, even though the first trimester is a critical period for fetal organogenesis and the most susceptible time to teratogen exposure, research regarding the effects of sevoflurane on organogenesis, especially on brain development, is insufficient. In the present study, human embryonic stem cells (hESC)-derived cerebral organoids were exposed to sevoflurane during the time corresponding to the first trimester to investigate the effect of early sevoflurane exposure on fetal brain development, specifically the processes of neuronal differentiation and maturation. Organoid size exposed to the intermediate concentration of sevoflurane did not differ from control, immunofluorescence demonstrated that sevoflurane temporarily decreased the size of SOX2 + /N-cad + ventricular zone structures only during the mid-time point, and upregulated expression of TUJ1 and MAP2 only during the early time point. However, all markers returned to normal levels, and organoids formed normal cortical structures at the late time point. Our results suggest that maternal sevoflurane exposure during the first trimester of pregnancy can cause abnormal neuronal differentiation in the fetal brain. However, considering the recovery observed in later periods, sevoflurane exposure might not have lasting impacts on fetal brain development.
Collapse
Affiliation(s)
- Jae A Lee
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Dong Hyuck Bae
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Woo Hee Choi
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea
| | - Chang-Hoon Cho
- R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea
| | - Yun-Sic Bang
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, CHA Bundang Medical Center, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| | - Jongman Yoo
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea. .,R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
8
|
Ji D, Karlik J. Neurotoxic Impact of Individual Anesthetic Agents on the Developing Brain. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1779. [PMID: 36421228 PMCID: PMC9689007 DOI: 10.3390/children9111779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 11/14/2022] [Indexed: 08/04/2023]
Abstract
Concerns about the safety of anesthetic agents in children arose after animal studies revealed disruptions in neurodevelopment after exposure to commonly used anesthetic drugs. These animal studies revealed that volatile inhalational agents, propofol, ketamine, and thiopental may have detrimental effects on neurodevelopment and cognitive function, but dexmedetomidine and xenon have been shown to have neuroprotective properties. The neurocognitive effects of benzodiazepines have not been extensively studied, so their effects on neurodevelopment are undetermined. However, experimental animal models may not truly represent the pathophysiological processes in children. Multiple landmark studies, including the MASK, PANDA, and GAS studies have provided reassurance that brief exposure to anesthesia is not associated with adverse neurocognitive outcomes in infants and children, regardless of the type of anesthetic agent used.
Collapse
|
9
|
Zhang J, Chang Q, Rizzello L, Wu Y. Research progress on the effects and mechanisms of anesthetics on neural stem cells. IBRAIN 2022; 8:453-464. [PMID: 37786590 PMCID: PMC10528967 DOI: 10.1002/ibra.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 10/04/2023]
Abstract
Exposure to anesthetic drugs has been proven to seriously affect developing animals in terms of neural stem cells' (NSCs') proliferation, differentiation, and apoptosis. This can severely hamper the development of physiological learning and memory skills. Studies on the effects of anesthetics on NSCs' proliferation and differentiation are thus reviewed here, with the aim to highlight which specific drug mechanisms are the least harmful to NSCs. PubMed has been used as the preferential searching database of relevant literature to identify studies on the effects and mechanisms of NSCs' proliferation and differentiation. It was concluded that propofol and sevoflurane may be the safest options for NSCs during pregnancy and in pediatric clinical procedures, while dexmedetomidine has been found to reduce opioid-related damage in NSCs. It was also found that the growth environment may impact neurodevelopment even more than narcotic drugs. Nonetheless, the current scientific literature available further highlights how more extensive clinical trials are absolutely required for corroborating the conclusion drawn here.
Collapse
Affiliation(s)
- Ji Zhang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Quan‐Yuan Chang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Loris Rizzello
- Department of Pharmaceutical SciencesUniversity of MilanMilanItaly
- National Institute of Molecular Genetics (INGM)MilanItaly
| | - You Wu
- Department of Family PlanningThe Affiliated Hospital of Zunyi Medical UniversityGuizhouZunyiChina
| |
Collapse
|
10
|
Feng Y, Wang K, Wang N, Jia P, Zhang L, Yuan H, Lu P, Lu Y, Zhang H, Li R, Zhang Y, Li Q, Zhang P. Tetramethylpyrazine protects neural stem cells against sevoflurane-induced toxicity through Akt/GSK-3β pathway. Metab Brain Dis 2022; 37:2457-2466. [PMID: 35838869 DOI: 10.1007/s11011-022-01008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022]
Abstract
Sevoflurane, a commonly used anesthetic, has been found to cause neural stem cell (NSC) injury, thereby contributing to neurocognitive impairment following general anesthesia. Tetramethylpyrazine (TMP), one of the most widely used medicinal compounds isolated from a traditional Chinese herb, possess neuroprotective activity. However, its effect on sevoflurane-induced NSC injury remains unclear. NSCs were pretreated with indicated concentrations of TMP for 2 h and then exposed to sevoflurane for 6 h. Cell injury was measured using lactate dehydrogenase (LDH) release assay. Cell viability and proliferation were detected by cell counting kit-8 (CCK-8) assay and 5-bromo-2'-deoxyuridine (BrdU) labeling, respectively. Apoptotic cells were detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The levels of cleaved caspase-3, phosphorylated protein kinase B (Akt) and phosphorylated glycogen synthase kinase-3β (GSK-3β) were detected by western blotting. Our results showed exposure to sevoflurane decreased the viability and proliferation of NSCs, while TMP preserved NSC viability and proliferation after sevoflurane exposure. In addition, the expression of cleaved caspase-3 and TUNEL positive cells were markedly decreased in TMP-treated NSCs compared with the control. Furthermore, pretreatment with TMP significantly increased the levels of phosphorylated Akt and GSK-3β in sevoflurane-injured NSCs. However, an upstream inhibitor of Akt, LY294002 abolished the protective of TMP on the cell viability of NSCs. In conclusion, these findings indicate that TMP protects NSCs from sevoflurane-induced toxicity through Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Yan Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
- Department of Anesthesiology, Xi'an People's Hospital (Xi'an Fourth Hospital), 710004, Xi'an, Shaanxi, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Lei Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
- Department of Anesthesiology, Xi'an People's Hospital (Xi'an Fourth Hospital), 710004, Xi'an, Shaanxi, China
| | - Haozheng Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Qianqian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, 710004, Xi'an, Shaanxi, China.
| |
Collapse
|
11
|
Shen Y, Zhou T, Liu X, Liu Y, Li Y, Zeng D, Zhong W, Zhang M. Sevoflurane-Induced miR-211-5p Promotes Neuronal Apoptosis by Inhibiting Efemp2. ASN Neuro 2021; 13:17590914211035036. [PMID: 34730432 PMCID: PMC8819752 DOI: 10.1177/17590914211035036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Sevoflurane exposure can result in serious neurological side effects including neuronal
apoptosis and cognitive impairment. Although the microRNA miR-211-5p is profoundly
upregulated following sevoflurane exposure in neonatal rodent models, the impact of
miR-211-5p on neuronal apoptosis and cognitive impairment postsevoflurane exposure has not
yet been elucidated. Here, we found that sevoflurane upregulated miR-211-5p and
downregulated EGF-Containing Fibulin Extracellular Matrix Protein 2 (Efemp2, Fibulin-4)
levels in vitro and in vivo. Sevoflurane's effect on miR-211-5p expression was based on
enhancing primary miR-211 transcription. miR-211-5p targets Efemp2's mRNA 3′-untranslated
region, reducing Efemp2 expression. RNA immunoprecipitation revealed significant
enrichment of the miR-211-5p:Efemp2 mRNA dyad in the RNA-induced silencing complex.
miR-211-5p mimics downregulated Efemp2, leading to phosphorylation of Smad2 and Smad3,
upregulation of pro-apoptotic Bim, and mitochondrial release of allograft inflammatory
factor 1 and cytochrome C. In contrast, miR-211-5p hairpin inhibitor (AntimiR-211-5p)
negatively regulated this apoptotic pathway and reduced neuronal apoptosis in an
Efemp2-dependent manner. Sevoflurane-exposed mice administered AntimiR-211-5p displayed
reduced cortical apoptosis levels and near-term cognitive impairment. In conclusion,
sevoflurane-induced miR-211-5p promotes neuronal apoptosis via Efemp2 inhibition. Summary
statement: This study revealed the significance of sevoflurane-induced increases in
miR-211-5p on the promotion of neuronal apoptosis via inhibition of Efemp2 and its
downstream targets.
Collapse
Affiliation(s)
- Yousu Shen
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Tao Zhou
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Xiaobing Liu
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Yanlong Liu
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Yaqi Li
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Dewu Zeng
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Wensheng Zhong
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| | - Mingsheng Zhang
- Department of Anaesthesiology, 159384Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Jiangxi, China
| |
Collapse
|
12
|
Apai C, Shah R, Tran K, Pandya Shah S. Anesthesia and the Developing Brain: A Review of Sevoflurane-induced Neurotoxicity in Pediatric Populations. Clin Ther 2021; 43:762-778. [PMID: 33674065 DOI: 10.1016/j.clinthera.2021.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/14/2020] [Accepted: 01/25/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE For over 150 years of anesthetic practice, it was believed that the effects of general anesthetics were temporary and not adverse. A growing number of studies over the past 2 decades, however, have identified structural and cognitive abnormalities, especially in the developing brain. Despite the growing evidence of anesthetic-induced neurotoxicity in animal studies, the evidence to date in humans has been inconsistent and unclear. Sevoflurane, a commonly used inhalational agent in pediatric anesthesia, is an agent of choice for inhalational induction due to its rapid activity and low blood-gas solubility. With evaluation of the current literature, improved considerations can be made regarding the widespread use of sevoflurane as an anesthetic. METHODS PubMed database was searched for article published between 1969 through 2020. The reference lists of identified articles were searched manually for additional papers eligible for inclusion. This review addressed the tolerability of sevoflurane in specific populations, particularly pediatrics, and is divided into 3 parts: (1) the history of sevoflurane use in anesthetic practice and the pharmacokinetic properties that make it advantageous in pediatric populations; (2) proposed mechanisms of anesthesia-induced neurotoxicity; and (3) considerations due to potential adverse effects of sevoflurane in both short and long procedures. FINDINGS There is reason for concern regarding the neurotoxic effects of sevoflurane in both the pediatric and elderly populations, as spatial memory loss, developmental deficits, and an enhanced risk for Alzheimer disease have been linked with the use of this popular inhalational agent. IMPLICATIONS The duration and dose of sevoflurane may need to be altered, especially in longer procedures in pediatric populations. This may change how sevoflurane is administered, thus indicating a greater demand for an understanding of its limitations as an anesthetic agent.
Collapse
Affiliation(s)
- Carol Apai
- Department of Anesthesiology, New Jersey Medical School, Division of Biomedical and Health Sciences, Rutgers University, Newark, NJ, USA
| | - Rohan Shah
- Department of Anesthesiology, New Jersey Medical School, Division of Biomedical and Health Sciences, Rutgers University, Newark, NJ, USA
| | - Khoa Tran
- Department of Anesthesiology, Keck Hospital, Keck Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Shridevi Pandya Shah
- Department of Anesthesiology, New Jersey Medical School, Division of Biomedical and Health Sciences, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
13
|
Wang Y, Yin CP, Tai YL, Zhao ZJ, Hou ZY, Wang QJ. Apoptosis inhibition is involved in improvement of sevoflurane-induced cognitive impairment following normobaric hyperoxia preconditioning in aged rats. Exp Ther Med 2021; 21:203. [PMID: 33500697 PMCID: PMC7818554 DOI: 10.3892/etm.2021.9636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Sevoflurane, a commonly used anesthetic agent has been confirmed to induce cognitive impairment in aged rats. Normobaric hyperoxia preconditioning has been demonstrated to induce neuroprotection in rats. The present study aimed to determine whether normobaric hyperoxia preconditioning could ameliorate cognitive deficit induced by sevoflurane and the possible mechanism by which it may exert its effect. A total of 66, 20-month-old male Sprague-Dawley rats were randomly divided into 3 groups (n=22 each): Rats in the control (C) and sevoflurane anesthesia (S) groups received no normobaric hyperoxia preconditioning before sevoflurane exposure, rats in the normobaric hyperoxia pretreatment (HO) group received normobaric hyperoxia preconditioning before sevoflurane exposure (95% oxygen for 4 continuous h daily for 6 consecutive days). The anesthesia rats (S and HO groups), were exposed to 2.5% sevoflurane for 5 h, while the sham anesthesia rats (C group) were exposed to no sevoflurane. The neurobehavioral assessment was performed using a Morris water maze test, the expressions of the apoptosis proteins were determined using western blot analysis, and the apoptosis rate and cytosolic calcium concentration were measured by flow cytometry. Normobaric hyperoxia preconditioning improved prolonged escape latency and raised the number of platform crossings induced by sevoflurane in the Morris water maze test, increased the level of bcl-2 protein, and decreased the level of bax and active caspase-3 protein, the apoptosis rate and cytosolic calcium concentration in the hippocampus 24 h after sevoflurane exposure. The findings of the present study may imply that normobaric hyperoxia preconditioning attenuates sevoflurane-induced spatial learning and memory impairment, and this effect may be partly related to apoptosis inhibition in the hippocampus. In conclusion, normobaric hyperoxia preconditioning may be a promising strategy against sevoflurane-induced cognitive impairment by inhibiting the hippocampal neuron apoptosis.
Collapse
Affiliation(s)
- Ying Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China.,Department of Anesthesiology, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Chun-Ping Yin
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yan-Lei Tai
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Zi-Jun Zhao
- Department of Anesthesiology, Hebei Chest Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Zhi-Yong Hou
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Qiu-Jun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
14
|
Zhang Y, Li M, Cui E, Zhang H, Zhu X, Zhou J, Yan M, Sun J. Dexmedetomidine attenuates sevoflurane‑induced neurocognitive impairment through α2‑adrenoceptors. Mol Med Rep 2020; 23:38. [PMID: 33179100 PMCID: PMC7684862 DOI: 10.3892/mmr.2020.11676] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 07/28/2020] [Indexed: 12/25/2022] Open
Abstract
It has been reported that sevoflurane induces neurotoxicity in the developing brain. Dexmedetomidine is an α2 adrenoceptor agonist used for the prevention of sevoflurane‑induced agitation in children in clinical practice. The aim of the present study was to determine whether dexmedetomidine could prevent sevoflurane‑induced neuroapoptosis, neuroinflammation, oxidative stress and neurocognitive impairment. Additionally, the involvement of α2 adrenoceptors in the neuroprotective effect of dexmedetomidine was assessed. Postnatal day (P)6 C57BL/6 male mice were randomly divided into four groups (n=6 in each group). Mice were pretreated with dexmedetomidine, either alone or together with yohimbine, an α2 adrenoceptor inhibitor, then exposed to 3% sevoflurane in 25% oxygen. Control mice either received normal saline alone or with sevoflurane exposure. Following sevoflurane exposure, the expression of cleaved caspase‑3 was detected by immunohistochemistry in hippocampal tissue sections. In addition, the levels of tumor necrosis factor‑α (TNF‑α), interleukin (IL)‑1β, IL‑6 and malondialdehyde, as well as superoxide dismutase (SOD) activity in the hippocampus were measured. At P35, the learning and memory abilities were assessed in each mouse using a Morris water maze test. Dexmedetomidine significantly decreased the expression of activated caspase‑3 following sevoflurane exposure. Moreover, dexmedetomidine significantly decreased the levels of TNF‑α, IL‑1β and IL‑6 in the hippocampus. SOD activity also increased in a dose‑dependent manner in dexmedetomidine‑treated mice. MDA decreased in a dose‑dependent manner in dexmedetomidine‑treated mice. Lastly, sevoflurane‑induced learning and memory impairment was reversed by dexmedetomidine treatment. By contrast, co‑administration of yohimbine significantly attenuated the neuroprotective effects of dexmedetomidine. These findings suggested that dexmedetomidine exerted a neuroprotective effect against sevoflurane‑induced apoptosis, inflammation, oxidative stress and neurocognitive impairment, which was mediated, at least in part, by α2 adrenoceptors.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Mao Li
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Enhui Cui
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Hao Zhang
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Xiaozhong Zhu
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Jing Zhou
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Ming Yan
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Jian Sun
- Department of Anesthesiology, The Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
15
|
Wang Q, Tian X, Lu Q, Liu K, Gong J. Study on the ameliorating effect of miR-221-3p on the nerve cells injury induced by sevoflurane. Int J Neurosci 2020; 132:181-191. [PMID: 32900248 DOI: 10.1080/00207454.2020.1806267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Sevoflurane is a widely used anesthetics, however, it has been reported that sevoflurane has neurotoxic effects. Studies have shown that miR-221-3p can ameliorate neuron damage. This study was to investigate whether miR-221-3p could reduce the neurotoxic effect of sevoflurane on nerve cells. MATERIALS AND METHODS The rat hippocampal neuron cells were treated with sevoflurane or cultured normally. And we constructed neuron cells that overexpressed or low expression of miR-221-3p in the presence or absence of sevoflurane. The cells were transfected with CDKN1B or siCDKN1B, and co-transfected with miR-221-3p mimic and CDKN1B or miR-221-3p inhibitor and siCDKN1B. Cell viability and apoptosis were detected by CCK-8 and flow cytometer. Target gene of miR-221-3p were predicted by TargetScan and luciferase reporter assay. The expressions of related genes were detected by western blotting and quantitative real-time polymerase chain reaction. RESULTS Sevoflurane decreased miR-221-3p level and increased CDKN1B level, inhibited cell viability and promoted apoptosis. Overexpress of miR-221-3p decreased CDKN1B level, up-regulated cell viability and inhibited apoptosis, and reversed the effects of sevoflurane on cell viability and apoptosis, while the effects low expression of miR-221-3p was contrary. CDKN1B was the target gene of miR-221-3p, which inhibited cell viability and promoted apoptosis, and reversed the effects of miR-221-3p mimic, whereas siCDKN1B did the opposite effects. CONCLUSIONS Sevoflurane can cause nerve cell injury, and miR-221-3p may promote cell activity and inhibit apoptosis by inhibiting CDKN1B expression, thereby ameliorating cell injury induced by sevoflurane.
Collapse
Affiliation(s)
- Qirui Wang
- Department of Anesthesiology, Zhenhai District People's Hospital of Ningbo, Ningbo, Zhejiang, China
| | - Xin Tian
- Department of Anesthesiology, Zhenhai District People's Hospital of Ningbo, Ningbo, Zhejiang, China
| | - Qijuan Lu
- Department of Anesthesiology, Zhenhai District People's Hospital of Ningbo, Ningbo, Zhejiang, China
| | - Kun Liu
- Department of Anesthesiology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jiekun Gong
- Department of Anesthesiology, Zhenhai District People's Hospital of Ningbo, Ningbo, Zhejiang, China
| |
Collapse
|
16
|
Yang F, Zhao H, Zhang K, Wu X, Liu H. Research progress and treatment strategies for anesthetic neurotoxicity. Brain Res Bull 2020; 164:37-44. [PMID: 32798600 DOI: 10.1016/j.brainresbull.2020.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/20/2020] [Accepted: 08/05/2020] [Indexed: 12/23/2022]
Abstract
Every year, a large number of infants and young children worldwide are administered general anesthesia. Whether general anesthesia adversely affects the intellectual development and cognitive function of children at a later date remains controversial. Many animal experiments have shown that general anesthetics can cause nerve damage during development, affect synaptic plasticity, and induce apoptosis, and finally affect learning and memory function in adulthood. The neurotoxicity of pediatric anesthetics (PAN) has received extensive attention in the field of anesthesia, which has been listed as a potential problem affecting public health by NFDA of the United States. Previous studies on rodents and non-human primates indicate that inhalation of anesthetics early after birth can induce long-term and sustained impairment of learning and memory function, as well as changes in brain function. Many anti-oxidant drugs, dexmedetomidine, as well as a rich living environment and exercise have been proven to reduce the neurotoxicity of anesthetics. In this paper, we summarize the research progress, molecular mechanisms and current intervention measures of anesthetic neurotoxicity.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Sanhao Street 36, Shenyang, 110004, China.
| | - Hai Zhao
- Clinical Skills Center, Shenyang Medical College, Huanghe Street 146, Shenyang, 110034, China.
| | - Kaiyuan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Sanhao Street 36, Shenyang, 110004, China.
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Sanhao Street 36, Shenyang, 110004, China.
| | - Hongtao Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Sanhao Street 36, Shenyang, 110004, China.
| |
Collapse
|
17
|
Zhang L, Xue Z, Yan J, Jiang H. LncRNA Rik-203 Contributes to Sevoflurane Induced Neurotoxicity? Front Med (Lausanne) 2020; 7:353. [PMID: 32793611 PMCID: PMC7387574 DOI: 10.3389/fmed.2020.00353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Background: The anesthetics inhibit neural differentiation, induce neuron loss and cognitive impairment in young animals. However, the underlying mechanisms of anesthesia on neural differentiation are unknown. Methods: Embryonic stem cells (ESCs) and mice received sevoflurane anesthesia. RNA sequencing; gene expression of mRNAs, LncRNAs and miRNAs; over-expression and RNA interference of genes; flow cytometry; real-time quantity PCR and Western blot were used in the studies. RNA pull-down assay and PCR were employed to detect any miRNA that attached to Rik-203. The binding of miRNA with mRNA of BDNF was presented by the luciferase assay. Results: Here we found that LncRNA Riken-203(Rik-203) was highly expressed in mice brain and was upregulated during neural differentiation. Sevoflurane decreased the amount of Rik-203 in mice brain. Knockdown of Rik-203 repressed the neural differentiation derived from mouse embryonic stem cell and downregulated the neural progenitor cells markers Sox1 and Nestin. RNA pull-down showed that miR-466l-3p was highly bound to Rik-203. Inhibition of miR-466l-3p restored the neural differentiation repressed by Rik-203 knockdown. Brain derived neurotrophic factor (BDNF), which was downregulated by sevoflurane, was also directly targeted by miR-466l-3p. Overexpression of BDNF restored the neural differentiation repressed by miR-466l-3p and Rik-203 knockdown. Conclusion: Our study suggested that sevoflurane related LncRNARik-203 facilitates neural differentiation by inhibiting miR-466l-3p's ability to reduce BDNF levels.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyu Xue
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Li T, Huang Z, Wang X, Zou J, Tan S. Role of the GABAA receptors in the long-term cognitive impairments caused by neonatal sevoflurane exposure. Rev Neurosci 2020; 30:869-879. [PMID: 31145696 DOI: 10.1515/revneuro-2019-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023]
Abstract
Sevoflurane is a widely used inhalational anesthetic in pediatric surgeries, which is considered reasonably safe and reversible upon withdrawal. However, recent preclinical studies suggested that peri-neonatal sevoflurane exposure may cause developmental abnormalities in the brain. The present review aimed to present and discuss the accumulating experimental data regarding the undesirable effects of sevoflurane on brain development as revealed by the laboratory studies. First, we summarized the long-lasting side effects of neonatal sevoflurane exposure on cognitive functions. Subsequently, we presented the structural changes, namely, neuroapoptosis, neurogenesis and synaptogenesis, following sevoflurane exposure in the immature brain. Finally, we also discussed the potential mechanisms underlying subsequent cognitive impairments later in life, which are induced by neonatal sevoflurane exposure and pointed out potential strategies for mitigating sevoflurane-induced long-term cognitive impairments. The type A gamma-amino butyric acid (GABAA) receptor, the main targets of sevoflurane, is excitatory rather than inhibitory in the immature neurons. The excitatory effects of the GABAA receptors have been linked to increased neuroapoptosis, elevated serum corticosterone levels and epigenetic modifications following neonatal sevoflurane exposure in rodents, which might contribute to sevoflurane-induced long-term cognitive abnormalities. We proposed that the excitatory GABAA receptor-mediated HPA axis activity might be a novel mechanism underlying sevoflurane-induced long-term cognitive impairments. More studies are needed to investigate the effectiveness and mechanisms by targeting the excitatory GABAA receptor as a prevention strategy to alleviate cognitive deficits induced by neonatal sevoflurane exposure in future.
Collapse
Affiliation(s)
- Tao Li
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Zeyi Huang
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Xianwen Wang
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Ju Zou
- Department of Parasitology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Sijie Tan
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| |
Collapse
|
19
|
Sevoflurane Impairs Short-Term Memory by Affecting PSD-95 and AMPA Receptor in the Hippocampus of a Mouse Model. Behav Neurol 2019; 2019:1068260. [PMID: 31772680 PMCID: PMC6854262 DOI: 10.1155/2019/1068260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/23/2019] [Accepted: 09/10/2019] [Indexed: 11/18/2022] Open
Abstract
Objective To explore the effects of sevoflurane on the latency and error times of the passive avoidance and levels of PSD-95 and AMPA receptors in the hippocampus. We evaluated the effects of sevoflurane on short-term memory in adult mice and explored the possible mechanism. Methods 144 Kunming mice (2-3 months, 30-35 g) were randomly divided into two groups A (n = 64) and B (n = 80) and received the dark-avoidance (DA) and step-down avoidance (SA) tests, respectively. The groups DA and SA were further divided into control (inhaled 40% O2 2 h) and sevoflurane (3.3% sevoflurane and 40% O2 2 h) subgroups. Before inhalation intervention, all mice were trained to be familiar with the Morris water maze (MWM). According to the test points of behavioral indicators, 8 mice were randomly selected from each subgroup at point 12 h (T1), 24 h (T2), 48 h (T3), and 72 h (T4) after inhalation intervention. The step-through latency and error times were measured in 5 min. After the behavioral test, the mice were killed and the tissues of the hippocampus were taken for hematoxylin and eosin (H&E) staining. The expression level of PSD-95 and AMPA receptors in the hippocampus was detected by immunohistochemistry and Western Blot. The changes of synaptic transmission were measured via electrophysiology analysis of hippocampal slices. Results The mice in the control subgroups found the platform in a shorter pathway than those in the sevoflurane subgroups during an MWM test. The step-through latency of T1 and T2 in the sevoflurane subgroup was shorter than baseline time, and the error times were increased in 5 min and higher than baseline time when compared with the control subgroup (P < 0.05) in the A and B groups. Compared with the control subgroup, the expression level of PSD-95 and AMPA receptors in the hippocampus was decreased at T1 and T2 in the sevoflurane subgroup (P < 0.05). The nerve cells were partially swelling. Electrophysiology analysis showed that the levels of PSD-95 and AMPA receptor expression were associated with synaptic transmission. Conclusion Sevoflurane impaired short-term memory in adult mice by inhibiting the expression of PSD-95 and AMPA receptors in the hippocampus, which led to the decrease in synaptic transmission.
Collapse
|
20
|
Chen P, Shang A, Wang W, Yang J. Astragaloside suppresses tumor necrosis factor receptor‐associated factor 5 signaling pathway and alleviates neurodegenerative changes in retinal pigment epithelial cells induced by isoflurane. J Cell Biochem 2018; 120:1028-1037. [PMID: 30277612 DOI: 10.1002/jcb.27599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Pei‐Jun Chen
- Department of Anesthesiology The First Affiliated Hospital of Soochow University Suzhou China
- Department of Anesthesiology The Sixth People’s Hospital of Yancheng City Yancheng China
| | - An‐Quan Shang
- Department of Laboratory Medicine Tongji Hospital of Tongji University School of Medicine Shanghai China
| | - Wei‐Wei Wang
- Department of Pathology The Sixth People’s Hospital of Yancheng City Yancheng China
| | - Jian‐Ping Yang
- Department of Anesthesiology The First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
21
|
Kashem MA, Sultana N, Balcar VJ. Exposure of Rat Neural Stem Cells to Ethanol Affects Cell Numbers and Alters Expression of 28 Proteins. Neurochem Res 2018; 43:1841-1854. [PMID: 30043189 DOI: 10.1007/s11064-018-2600-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/17/2018] [Accepted: 07/20/2018] [Indexed: 11/28/2022]
Abstract
Developing brain cells express many proteins but little is known of how their protein composition responds to chronic exposure to alcohol and/or how such changes might relate to alcohol toxicity. We used cultures derived from embryonic rat brain (previously shown to contain mostly neural stem cells; rat NSC, rNSC), exposed them to ethanol (25-100 mM) for up to 96 h and studied how they reacted. Ethanol (50 and 100 mM) reduced cell numbers indicating either compromised cell proliferation, cytotoxicity or both. Increased lipid peroxidation was consistent with the presence of oxidative stress accompanying alcohol-induced cytotoxicity. Proteomics revealed 28 proteins as altered by ethanol (50 mM for 96 h). Some were constituents of cytoskeleton, others were involved in transcription/translation, signal transduction and oxidative stress. Nucleophosmin (NPM1) and dead-end protein homolog 1 (DND1) were further studied by immunological techniques in cultured neurons and astrocytes (derived from brain tissue at embryonic ages E15 and E20, respectively). In the case of DND1 (but not NPM1) ethanol induced similar pattern of changes in both types of cells. Given the critical role of the protein NPM1 in cell proliferation and differentiation, its reduced expression in the ethanol-exposed rNSC could, in part, explain the lower cells numbers. We conclude that chronic ethanol profoundly alters protein composition of rNSC to the extent that their functioning-including proliferation and survival-would be seriously compromised. Translated to humans, such changes could point the way towards mechanisms underlying the fetal alcohol spectrum disorder and/or alcoholism later in life.
Collapse
Affiliation(s)
- Mohammed A Kashem
- Laboratory of Neurochemistry, Bosch Institute and Discipline of Anatomy and Histology, School of Medical Sciences, Sydney Medical School, The University of Sydney, Anderson Stuart Building F13, Sydney, NSW, 2006, Australia
| | - Nilufa Sultana
- Laboratory of Neurochemistry, Bosch Institute and Discipline of Anatomy and Histology, School of Medical Sciences, Sydney Medical School, The University of Sydney, Anderson Stuart Building F13, Sydney, NSW, 2006, Australia
| | - Vladimir J Balcar
- Laboratory of Neurochemistry, Bosch Institute and Discipline of Anatomy and Histology, School of Medical Sciences, Sydney Medical School, The University of Sydney, Anderson Stuart Building F13, Sydney, NSW, 2006, Australia.
| |
Collapse
|
22
|
Cheng Y, Jiang Y, Zhang L, Wang J, Chai D, Hu R, Li C, Sun Y, Jiang H. Mesenchymal stromal cells attenuate sevoflurane-induced apoptosis in human neuroglioma H4 cells. BMC Anesthesiol 2018; 18:84. [PMID: 30021512 PMCID: PMC6052698 DOI: 10.1186/s12871-018-0553-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 06/27/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Inhalation of sevoflurane can induce neuronal apoptosis, cognitive impairment and abnormal behaviors. Bone marrow mesenchymal stem cells (MSCs) can secret neurotrophic factors and cytokines to protect from oxidative stress-related neuronal apoptosis. However, whether MSCs can protect from sevoflurane-induced neuronal apoptosis and the potential mechanisms are unclear. METHODS A non-contact co-culture of MSCs with human neuroglioma H4 cells (H4 cells) was built. H4 cells were co-cultured with MSCs or without MSCs (control) for 24 h. The co-cultured H4 cells were exposed to 4% sevoflurane for 6 h. The levels of caspase-3, reactive oxygen species (ROS), adenosine triphosphate (ATP), and the release of cytochrome C were determined by Western blot and fluorescence assay. RESULTS Sevoflurane exposure significantly elevated the levels of cleaved caspase 3 and Bax in H4 cells. However, these phenomena were significantly offset by the co-culture with MSCs in H4 cells. Co-culture with MSCs before, but not after, sevoflurane exposure, significantly attenuated sevoflurane-induced ROS production in H4 cells. MSCs prevented sevoflurane-mediated release of cytochrome C from the mitochondria and production of ATP in H4 cells. CONCLUSIONS Our study indicated that soluble factors secreted by MSCs attenuated the sevoflurane-induced oxidative stress and apoptosis of neuronal cells by preserving their mitochondrial function.
Collapse
Affiliation(s)
- Yanyong Cheng
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Yunfeng Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Jiayi Wang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Dongdong Chai
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Rong Hu
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Chunzhu Li
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| |
Collapse
|
23
|
Effect of multiple neonatal sevoflurane exposures on hippocampal apolipoprotein E levels and learning and memory abilities. Pediatr Neonatol 2018; 59:154-160. [PMID: 28890046 DOI: 10.1016/j.pedneo.2017.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/21/2017] [Accepted: 08/22/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Sevoflurane anesthesia is widely used in pediatric patients. In this study, we investigated whether early multiple exposures to sevoflurane induced cognitive dysfunction by altering the hippocampal expression of ApoE later in development. METHODS Sprague-Dawley rats were exposed to 2.6% sevoflurane at postnatal day 7 (P7), P14, and P21 for 2 h. The ability of learning and memory was assessed using the Morris water maze at P37 and P97. The hippocampal volume was measured by magnetic resonance imaging (MRI) at P37 and P97. The hippocampal expression of ApoE was assessed by immunohistochemical analyses and real-time polymerase chain reaction (PCR). RESULTS Behavioral testing revealed that the ability of learning and memory in the sevoflurane-exposed rats was decreased compared with the control animals; however, there was no significant difference (P > 0.05). The MRI results showed a significant decrease in the left hippocampal volume, left maximum hippocampal length, and right maximum hippocampal length in the sevoflurane young group compared with the control young group (P < 0.05). The brain volume, left maximum hippocampal length, right hippocampal volume, and maximum brain length were significantly lower in the sevoflurane adult group than in the control adult group (P < 0.05). In young animals, the ApoE expression in the hippocampal CA1 and CA3 regions and the ApoE mRNA level were significantly higher compared with the control group (P < 0.05), but not in the dentate gyrus region (P > 0.05). Among the adult animals, there was no significant difference between the groups in any parameter tested (P > 0.05). CONCLUSION Multiple exposures to sevoflurane during the neonatal period decreased the volume of the hippocampus and increased the hippocampal expression of ApoE. The differential expression level of ApoE in different hippocampal subdivisions suggested that the expression of ApoE was regionally specific and reversible.
Collapse
|
24
|
Sevoflurane preconditioning promotes activation of resident CSCs by transplanted BMSCs via miR-210 in a rat model for myocardial infarction. Oncotarget 2017; 8:114637-114647. [PMID: 29383108 PMCID: PMC5777720 DOI: 10.18632/oncotarget.23062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/14/2017] [Indexed: 11/26/2022] Open
Abstract
Objective To assess the effect of sevoflurane preconditioning (SFpre) on bone marrow mesenchymal stem cells (BMSCs) for the treatment of acute myocardial infarction. Results 24 hours after the transplantation, decreased apoptosis of implanted BMSCs and up-regulation of cytokines expression were found within the ischemic area in SFpreBMSCs group compared with BMSCs group (P < 0.05). 4 weeks later, SFpreBMSCs group showed more viable implanted BMSCs, CSC-derived cardiomyocytes, and higher vessel and myocardial density within the infarcted region and improved cardiac function, compared with control and BMSCs groups (P < 0.05). Compared with untreated BMSCs, promoted migration, inhibited apoptosis, increased cytokine secretion, and enhanced activation to CSCs were detected in SFpreBMSCs exposed to profound hypoxia and serum deprivation, via up-regulating miR-210 expression (P < 0.05). Conclusions Sevoflurane preconditioning can protect BMSCs against hypoxia by activating miR-210 expression and promote their paracrine functions and effects on resident CSCs. Methods After the preconditioning, rat BMSCs (SFpreBMSCs group) were transplanted into rat AMI models, while BMSCs group received unconditioned BMSCs. Apoptosis and paracrine functions of the transplanted BMSCs, angiogenesis, resident cardiac stem cells (CSCs) derived myocardial regeneration, cardiac function and remodeling were assessed at various time points. In vitro experiments were performed to determine the expression of miR-210 in BMSCs exposed to sevoflurane and the effect of sevoflurane on BMSCs’ migration, apoptosis and secretion of cytokines under hypoxic condition, as well as cytokine-induced CSCs activation.
Collapse
|
25
|
Yang Z, Lv J, Li X, Meng Q, Yang Q, Ma W, Li Y, Ke ZJ. Sevoflurane decreases self-renewal capacity and causes c-Jun N-terminal kinase-mediated damage of rat fetal neural stem cells. Sci Rep 2017; 7:46304. [PMID: 28393934 PMCID: PMC5385884 DOI: 10.1038/srep46304] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/14/2017] [Indexed: 01/09/2023] Open
Abstract
Increasing studies have demonstrated that sevoflurane can induce neurotoxicity in the developing brains. JNK normally promotes apoptosis. It was hypothesized that sevoflurane affected the proliferation and differentiation of FNSCs and induced cell apoptosis, which caused the learning and memory deficits via JNK pathway. Sevoflurane at a concentration of 1.2% did not induce damage on the FNSCS. However, concentrations of 2.4% and 4.8% decreased the cell viability, as shown by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and increased apoptosis, as shown by flow cytometry. The 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay demonstrated that 4.8% sevoflurane reduced the proliferation of FNSCs. Compared with the control group, the 4.8% sevoflurane group showed a decrease in the proportion of undifferentiated FNSCs at 6-h exposure; 4.8% sevoflurane could increase the p-JNK/JNK ratio. JNK inhibition by the specific inhibitor SP600125 enhanced partially the cell viability. Cumulatively, 4.8% sevoflurane induced significant damage on FNSCs; it decreased cell proliferation and proportion of undifferentiated cells as well. JNK pathway might play a key role in the decrease in survival of FNSCs induced by an inhaled anesthetic. The present findings might raise the possibility that JNK inhibition has therapeutic potential in protecting FNSCs from the adverse effects of the inhaled anesthetic.
Collapse
Affiliation(s)
- Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 200030, Shanghai, China
| | - Jingjing Lv
- Department of Anesthesiology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui, China
| | - Xingxing Li
- Department of Anesthesiology, First Affiliated Hospital of AnHui Medical University, Hefei, 230022, Anhui, China
| | - Qiong Meng
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 200030, Shanghai, China
| | - Qiling Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 200030, Shanghai, China
| | - Wei Ma
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 200030, Shanghai, China
| | - Yuanhai Li
- Department of Anesthesiology, First Affiliated Hospital of AnHui Medical University, Hefei, 230022, Anhui, China
| | - Zun Ji Ke
- Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
26
|
Fang F, Song R, Ling X, Peng M, Xue Z, Cang J. Multiple sevoflurane anesthesia in pregnant mice inhibits neurogenesis of fetal hippocampus via repressing transcription factor Pax6. Life Sci 2017; 175:16-22. [DOI: 10.1016/j.lfs.2017.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/24/2017] [Accepted: 03/05/2017] [Indexed: 10/20/2022]
|
27
|
Anesthetic neurotoxicity: Apoptosis and autophagic cell death mediated by calcium dysregulation. Neurotoxicol Teratol 2016; 60:59-62. [PMID: 27856359 DOI: 10.1016/j.ntt.2016.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 11/02/2016] [Accepted: 11/12/2016] [Indexed: 12/13/2022]
Abstract
A number of findings suggested that general anesthetics induced neural cell death by apoptosis in various animal models. Although clinical evidence regarding the correlation between anesthetic exposures at young age and subsequent cognitive impairments remains unclear, repeated or consistent exposures to general anesthetics may be a potential harmful risk in developing human brains. The mechanisms underlying the anesthetic neurotoxicity have received extensive attention recently. We will attempt a brief review to summarize current understanding on the role of both apoptosis and autophagic cell death mediated by calcium dysregulation in anesthetic neurotoxicity.
Collapse
|
28
|
Chen X, Zhou X, Lu D, Yang X, Zhou Z, Chen X, Chen Y, He W, Feng X. Aberrantly expressed long noncoding RNAs are involved in sevoflurane-induced developing hippocampal neuronal apoptosis: a microarray related study. Metab Brain Dis 2016; 31:1031-40. [PMID: 27234990 DOI: 10.1007/s11011-016-9838-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/16/2016] [Indexed: 12/23/2022]
Abstract
The commonly used volatile anesthetic sevoflurane has been shown to induce widespread apoptosis in the developing brain, yet the underlying molecular mechanisms are not fully understood. Accumulating research has demonstrated that long noncoding RNAs (lncRNAs) regulate multiple biological processes, including neural development, differentiation and apoptosis. They are aberrantly expressed in multiple neurodegenerative diseases. In this study, we employed a lncRNA-mRNA microarray analysis to determine whether and how lncRNAs are involved in sevoflurane-induced hippocampal neuronal apoptosis in neonatal mice. Our data showed that a single 6-h sevoflurane exposure of P7 mice resulted in significant morphological changes and apoptosis in the hippocampus. Moreover, the microarray simultaneously revealed 817 lncRNAs and 856 of their potential coding targets that related to apoptosis, of which 31 lncRNAs (19 up and 12 down) and 25 mRNAs were significantly differentially expressed (P < 0.05) after sevoflurane exposure. Importantly, we found that Bcl2l11 (BIM), which potentiates mitochondria-dependent apoptosis and its nearby enhancer-like lncRNA ENSMUST00000136025, were both more highly expressed in sevoflurane-treated samples compared with control samples. Subsequent qRT-PCR results confirmed the changes. Further CNC network indicated that lncRNA ENSMUST00000136025 was positively correlated with Bim. Moreover, sevoflurane induced a significant increase of pro-apoptotic protein BIM and Bax but a reduction of anti-apoptotic proteins Bcl-2 in the hippocampus. Our study first demonstrates that aberrantly expressed lncRNAs play a role in sevoflurane-induced hippocampal apoptosis. We noted that up-regulated ENSMUST00000136025 highly likely induced the over-expression of BIM, which eventually promoted mitochondria-mediated apoptosis. Such findings further broaden the understanding of molecular mechanisms responsible for sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fujian Provincial Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xue Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Dihan Lu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xiaoyu Yang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Zhibin Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xi Chen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Yanqing Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fujian Provincial Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
| | - Wen He
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
29
|
Yang L, Ge Y, Lin S, Fang X, Zhou L, Gao J. Sevoflurane inhibits the self-renewal of mouse embryonic stem cells via the GABAAR-ERK signaling pathway. Mol Med Rep 2016; 14:2119-26. [DOI: 10.3892/mmr.2016.5466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 06/20/2016] [Indexed: 11/06/2022] Open
|
30
|
Polydatin Protects Bone Marrow Stem Cells against Oxidative Injury: Involvement of Nrf 2/ARE Pathways. Stem Cells Int 2015; 2016:9394150. [PMID: 27022401 PMCID: PMC4684894 DOI: 10.1155/2016/9394150] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/20/2015] [Accepted: 08/02/2015] [Indexed: 01/14/2023] Open
Abstract
Polydatin, a glucoside of resveratrol, has been reported to possess potent antioxidative effects. In the present study, we aimed to investigate the effects of polydatin in bone marrow-derived mesenchymal stem cells (BMSCs) death caused by hydrogen peroxide (H2O2), imitating the microenvironment surrounding transplanted cells in the injured spinal cord in vitro. In our study, MTT results showed that polydatin effectively prevented the decrease of cell viability caused by H2O2. Hochest 33258, Annexin V-PI, and Western blot assay showed H2O2-induced apoptosis in BMSCs, which was attenuated by polydatin. Further studies indicated that polydatin significantly protects BMSCs against apoptosis due to its antioxidative effects and the regulation of Nrf 2/ARE pathway. Taken together, our results indicate that polydatin could be used in combination with BMSCs for the treatment of spinal cord injury by improving the cell survival and oxidative stress microenvironments.
Collapse
|