1
|
Gao T, Huang Z. Novel insights into sevoflurane-induced developmental neurotoxicity mechanisms. Epigenomics 2024; 16:1231-1252. [PMID: 39316776 PMCID: PMC11485883 DOI: 10.1080/17501911.2024.2395250] [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: 03/13/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: This study explores Sevoflurane (Sevo)-induced neurotoxicity mechanisms in neonates through transcriptome sequencing and models.Methods: Seven-day-old mice were exposed to 3% Sevo, and hippocampal tissue was collected for analysis of differentially expressed lncRNAs and mRNAs compared with normal mice. MiR-152-3p was selected, and the interaction between H19, USP30, and miR-152-3p was explored in BV2 microglial cells and mouse hippocampal neurons.Results: Sevo disrupts mitochondrial autophagy via USP30 upregulation, exacerbating neurotoxicity and activating NLRP1 inflammasome-mediated inflammation.Conclusion: Sevo neurotoxicity is mediated through the H19/miR-152-3p/USP30 axis, implicating microglial regulation of neuronal pyroptosis.
Collapse
Affiliation(s)
- Tingting Gao
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, P.R. China
| | - Zeqing Huang
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, P.R. China
| |
Collapse
|
2
|
Shi Q, Wang X, Pradhan AK, Fenzl T, Rammes G. The Effects of Sevoflurane and Aβ Interaction on CA1 Dendritic Spine Dynamics and MEGF10-Related Astrocytic Synapse Engulfment. Int J Mol Sci 2024; 25:7393. [PMID: 39000499 PMCID: PMC11242502 DOI: 10.3390/ijms25137393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
General anesthetics may accelerate the neuropathological changes related to Alzheimer's disease (AD), of which amyloid beta (Aβ)-induced toxicity is one of the main causes. However, the interaction of general anesthetics with different Aβ-isoforms remains unclear. In this study, we investigated the effects of sevoflurane (0.4 and 1.2 maximal alveolar concentration (MAC)) on four Aβ species-induced changes on dendritic spine density (DSD) in hippocampal brain slices of Thy1-eGFP mice and multiple epidermal growth factor-like domains 10 (MEGF10)-related astrocyte-mediated synaptic engulfment in hippocampal brain slices of C57BL/6 mice. We found that both sevoflurane and Aβ downregulated CA1-dendritic spines. Moreover, compared with either sevoflurane or Aβ alone, pre-treatment with Aβ isoforms followed by sevoflurane application in general further enhanced spine loss. This enhancement was related to MEGF10-related astrocyte-dependent synaptic engulfment, only in AβpE3 + 1.2 MAC sevoflurane and 3NTyrAβ + 1.2 MAC sevoflurane condition. In addition, removal of sevoflurane alleviated spine loss in Aβ + sevoflurane. In summary, these results suggest that both synapses and astrocytes are sensitive targets for sevoflurane; in the presence of 3NTyrAβ, 1.2 MAC sevoflurane alleviated astrocyte-mediated synaptic engulfment and exerted a lasting effect on dendritic spine remodeling.
Collapse
Affiliation(s)
- Qinfang Shi
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (Q.S.); (A.K.P.); (T.F.)
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingxing Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Arpit Kumar Pradhan
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (Q.S.); (A.K.P.); (T.F.)
- Graduate School of Systemic Neuroscience, Ludwig Maximilian University of Munich, 82152 Munich, Germany
| | - Thomas Fenzl
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (Q.S.); (A.K.P.); (T.F.)
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (Q.S.); (A.K.P.); (T.F.)
| |
Collapse
|
3
|
Ge Y, Ming L, Xu D. Sevoflurane-induced cognitive effect on α7-nicotine receptor and M 1 acetylcholine receptor expression in the hippocampus of aged rats. Neurol Res 2024; 46:593-604. [PMID: 38747300 DOI: 10.1080/01616412.2024.2338031] [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: 02/07/2024] [Accepted: 03/28/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Sevoflurane treatment increases the incidence of postoperative cognitive dysfunction (POCD), and patients with POCD show a decline in cognitive abilities compared to preoperative levels. OBJECTIVES This study aimed to investigate whether the activation of α7 nicotinic acetylcholine receptor (α7nAChR) and the expression of M1 acetylcholine receptor (mAChR M1) in the hippocampus affects the cognitive function of aged rats. METHODS Forty-eight Sprague-Dawley (SD) rats of 1-week- and 12-months-old were divided into eight groups: four groups for α7nAChR and four groups for mAChR M1, respectively. All SD rats received 1.0-02% sevoflurane for α7nAChR and 1.0-02% sevoflurane for mAChR M1 for 2-6 h, respectively. The Y-maze test was used to assess the ability to learn and memory after receiving sevoflurane for 7 days at the same moment portion. RT-PCR was used to determine the expression of α7nAChR and mAChR M1 in the hippocampus of rats. RESULTS The α7nAChR mitigated the formation of sevoflurane-induced memory impairment by modulating the translocation of NR2B from the intracellular reservoir to the cell surface reservoir within the hippocampus. Next, sevoflurane-induced decline of cognitive function and significantly decreased mAChR M1 expression at mRNA levels. CONCLUSION α7nAChR regulates the trafficking of NR2B in the hippocampus of rats via the Src-family tyrosine kinase (SFK) pathway. This regulation is associated with cognitive deficits induced by sevoflurane in hippocampal development. Sevoflurane affects the cognitive function of rats by suppressing the mAChR M1 expression at mRNA levels in the hippocampus.
Collapse
Affiliation(s)
- Yuan Ge
- Department of Anesthesiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Ming
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Dedong Xu
- Department of Anesthesiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
Jiao L, Jing Z, Zhang W, Su X, Yan H, Tian S. Codon Pattern and Context Analysis in Genes Triggering Alzheimer's Disease and Latent Tau Protein Aggregation Post-Anesthesia Exhibited Unique Molecular Patterns Associated with Functional Aspects. J Alzheimers Dis 2024; 97:1645-1660. [PMID: 38306048 DOI: 10.3233/jad-231142] [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] [Indexed: 02/03/2024]
Abstract
Background Previous reports have demonstrated post-operative dementia and Alzheimer's disease (AD), and increased amyloid-β levels and tau hyperphosphorylation have been observed in animal models post-anesthesia. Objective After surgical interventions, loss in memory has been observed that has been found linked with genes modulated after anesthesia. Present study aimed to study molecular pattern present in genes modulated post anesthesia and involved in characters progressing towards AD. Methods In the present study, 17 transcript variants belonging to eight genes, which have been found to modulate post-anesthesia and contribute to AD progression, were envisaged for their compositional features, molecular patterns, and codon and codon context-associated studies. Results The sequences' composition was G/C rich, influencing dinucleotide preference, codon preference, codon usage, and codon context. The G/C nucleotides being highly occurring nucleotides, CpGdinucleotides were also preferred; however, CpG was highly disfavored at p3-1 at the codon junction. The nucleotide composition of Cytosine exhibited a unique feature, and unlike other nucleotides, it did not correlate with codon bias. Contrarily, it correlated with the sequence lengths. The sequences were leucine-rich, and multiple leucine repeats were present, exhibiting the functional role of neuroprotection from neuroinflammation post-anesthesia. Conclusions The analysis pave the way to elucidate unique molecular patterns in genes modulated during anesthetic treatment and might help ameliorate the ill effects of anesthetics in the future.
Collapse
Affiliation(s)
- Liyuan Jiao
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ziye Jing
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenjie Zhang
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuesen Su
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hualei Yan
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shouyuan Tian
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
5
|
Wu Y, Yang Z, Su S, Xu X, Li Y, Li X, Gao Y, Sun D, Wan S, Pen M, Jin W, Ke C. Differential epitranscriptome and proteome modulation in the brain of neonatal mice exposed to isoflurane or sevoflurane. Cell Biol Toxicol 2023; 39:2133-2148. [PMID: 35249202 DOI: 10.1007/s10565-022-09701-9] [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: 11/07/2021] [Accepted: 02/11/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Repeated neonatal exposure to anesthetics may disturb neurodevelopment and cause neuropsychological disorders. The m6A modification participates in the gene regulation of neurodevelopment in mouse fetuses exposed to anesthetics. This study aims to explore the underlying molecular mechanisms of neurotoxicity after early-life anesthesia exposure. METHODS Mice were exposed to isoflurane (1.5%) or sevoflurane (2.3%) for 2 h daily during postnatal days (PND) 7-9. Sociability, spatial working memory, and anxiety-like behavior were assessed on PND 30-35. Synaptogenesis, epitranscriptome m6A, and the proteome of brain regions were evaluated on PND 21. RESULTS Both isoflurane and sevoflurane produced abnormal social behaviors at the juvenile age, with different sociality patterns in each group. Synaptogenesis in the hippocampal area CA3 was increased in the sevoflurane-exposed mice. Both anesthetics led to numerous persistent m6A-induced alterations in the brain, associated with critical metabolic, developmental, and immune functions. The proteins altered by isoflurane exposure were mainly associated with epilepsy, ataxia, and brain development. As for sevoflurane, the altered proteins were involved in social behavior. CONCLUSIONS Social interaction, the modulation patterns of the m6A modification, and protein expression were altered in an isoflurane or sevoflurane-specific way. Possible molecular pathways involved in brain impairment were revealed, as well as the mechanism underlying behavioral deficits following repeated exposure to anesthetics in newborns.
Collapse
Affiliation(s)
- Yanqiong Wu
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200030, China
| | - Shanchun Su
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xueqin Xu
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yang Li
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaohui Li
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yan Gao
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dongsheng Sun
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shengjun Wan
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Mingjin Pen
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Weiling Jin
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Changbin Ke
- Institute of Anesthesiology & Pain (IAP), Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
6
|
Li S, Hou Q, Wang R, Hou Y, Wang Q, Zhang B, Ni C, Zheng H. Sevoflurane upregulates neuron death process-related Ddit4 expression by NMDAR in the hippocampus. Aging (Albany NY) 2023; 15:5698-5712. [PMID: 37348034 PMCID: PMC10333074 DOI: 10.18632/aging.204822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a serious and common complication induced by anesthesia and surgery. Neuronal apoptosis induced by general anesthetic neurotoxicity is a high-risk factor. However, a comprehensive analysis of general anesthesia-regulated gene expression patterns and further research on molecular mechanisms are lacking. Here, we performed bioinformatics analysis of gene expression in the hippocampus of aged rats that received sevoflurane anesthesia in GSE139220 from the GEO database, found a total of 226 differentially expressed genes (DEGs) and investigated hub genes according to the number of biological processes in which the genes were enriched and performed screening by 12 algorithms with cytoHubba in Cytoscape. Among the screened hub genes, Agt, Cdkn1a, Ddit4, and Rhob are related to the neuronal death process. We further confirmed that these genes, especially Ddit4, were upregulated in the hippocampus of aged mice that received sevoflurane anesthesia. NMDAR, the core target receptor of sevoflurane, rather than GABAAR, mediates the sevoflurane regulation of DDIT4 expression. Our study screened sevoflurane-regulated DEGs and focused on the neuronal death process to reveal DDIT4 as a potential target mediated by NMDAR, which may provide a new target for the treatment of sevoflurane neurotoxicity.
Collapse
Affiliation(s)
- Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Runjia Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiang Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
7
|
Zhao X, Li Z, Guo Y, Liu Q, Qing M, Sheng Y, Chen Y, Xie L, Zhou Z. Alfaxalone Alleviates Neurotoxicity and Cognitive Impairment Induced by Isoflurane Anesthesia in Offspring Rats. Anesth Analg 2023; 136:1206-1216. [PMID: 36947459 DOI: 10.1213/ane.0000000000006420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
BACKGROUND The anesthetic isoflurane can cause neurotoxicity in fetuses and offspring of rats, affecting their neurodevelopment. However, the underlying mechanisms and therapeutic targets of isoflurane-induced neurotoxicity remain to be identified. Alfaxalone (ALF) is a steroid anesthetic. Steroids have been reported to have neuroprotective effects. This study aimed to investigate whether ALF could alleviate the isoflurane-induced neurotoxicity in fetuses and offspring of rats. METHODS On gestation day 15 (G15), the pregnant SD rats were randomly assigned to 4 groups: control 1 (CTL1) + control 2 (CTL2), isoflurane (ISO) + CTL2, CTL1 + ALF, and ISO + ALF. To analyze the changes in the expression levels of inflammatory cytokines, apoptotic factors, and synaptophysin, the brain tissues from the G15 fetuses and offspring at postnatal day 7 (P7), postnatal day 14 (P14), and postnatal day 31 (P31) were collected. The newborn neurons in the rats' offspring at P7, P14, and P31 were counted using immunofluorescence techniques. The Morris water maze (MWM) test was performed to assess the learning and memory abilities of P31 offspring rats. RESULTS ALF significantly alleviated the isoflurane-induced increase in the expression levels of inflammatory cytokines and apoptotic factors, such as interleukin (IL)-6 (ISO + CTL2 versus ISO + ALF: 5.133 ± 0.739 versus 1.093 ± 0.213, P < .001) and Caspase-3 (6.457 ± 0.6 versus 1.062 ± 0.1, P < .001) in the G15 fetuses. In P31 offspring rats, the expression levels of synaptophysin (0.719 ± 0.04 versus 1.068 ± 0.072, P < .001) and the number of newborn neurons in the dentate gyrus of the hippocampus were significantly lower in the ISO + CTL2 group as compared to those in the ISO + ALF group (118 ± 6 versus 140 ± 7, P < .001). These changes also occurred in the rat offspring at P7 and P14. In the MWM test, the escape latency of CTL1 + ALF group rats was significantly lower than that of ISO + ALF group rats (41 ± 6 versus 31 ± 7, P < .001) at P31. CONCLUSIONS Based on these findings, this study suggested that isoflurane exposure during pregnancy in rats could cause neuroinflammation and death of embryos as well as impairment of cognitive function in the offspring rats. ALF can be used to counteract the negative effects of isoflurane.
Collapse
Affiliation(s)
- Xingkai Zhao
- From the Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Jiang Y, Zhou Y, Tan S, Xu C, Ma J. Role of posttranslational modifications in memory and cognitive impairments caused by neonatal sevoflurane exposure. Front Pharmacol 2023; 14:1113345. [PMID: 36992831 PMCID: PMC10040769 DOI: 10.3389/fphar.2023.1113345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
With the advancement of technology, increasingly many newborns are receiving general anesthesia at a young age for surgery, other interventions, or clinical assessment. Anesthetics cause neurotoxicity and apoptosis of nerve cells, leading to memory and cognitive impairments. The most frequently used anesthetic in infants is sevoflurane; however, it has the potential to be neurotoxic. A single, short bout of sevoflurane exposure has little impact on cognitive function, but prolonged or recurrent exposure to general anesthetics can impair memory and cognitive function. However, the mechanisms underlying this association remain unknown. Posttranslational modifications (PTMs), which can be described roughly as the regulation of gene expression, protein activity, and protein function, have sparked enormous interest in neuroscience. Posttranslational modifications are a critical mechanism mediating anesthesia-induced long-term modifications in gene transcription and protein functional deficits in memory and cognition in children, according to a growing body of studies in recent years. Based on these recent findings, our paper reviews the effects of sevoflurane on memory loss and cognitive impairment, discusses how posttranslational modifications mechanisms can contribute to sevoflurane-induced neurotoxicity, and provides new insights into the prevention of sevoflurane-induced memory and cognitive impairments.
Collapse
Affiliation(s)
- Yongliang Jiang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People’s Hospital of Chengdu, Chengdu, China
| | - Siwen Tan
- Outpatient Department, West China Hospital of Sichuan University, Chengdu, China
| | - Chongxi Xu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junpeng Ma
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Junpeng Ma,
| |
Collapse
|
9
|
mTORC1-Dependent and GSDMD-Mediated Pyroptosis in Developmental Sevoflurane Neurotoxicity. Mol Neurobiol 2023; 60:116-132. [PMID: 36224321 DOI: 10.1007/s12035-022-03070-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/05/2022] [Indexed: 12/30/2022]
Abstract
Developmental sevoflurane exposure leads to neuronal cell death, and subsequent learning and memory cognitive defects. The underlyi\ng mechanism remains to be elucidated. Gasdermin D (GSDMD)-mediated pyroptosis is a form of inflammatory cell death and participates in a variety of neurodegenerative diseases. Several studies illustrated that dysregulation of mTOR activity is involved in pyroptotic cell death. The current study was designed to interrogate the role of GSDMD-mediated pyroptosis and mTOR activity in developmental sevoflurane exposure. We found that inhibition of GSDMD pore formation with Disulfiram (DSF) or Necrosulfonamide (NSA) significantly attenuated sevoflurane neurotoxicity in vitro. In addition, treatment with DSF or NSA also mitigated damage-associated molecular patterns (DAMPs) release and subsequent plasma membrane rupture (PMR) induced by sevoflurane challenge. Further investigation showed that the overactivation of mTOR signaling is involved in sevoflurane induced pyroptosis both in vivo and in vitro. Intriguingly, we found that the DAMPs release and subsequent PMR triggered by developmental sevoflurane priming were compromised by knocking down the expression of mTORC1 component Raptor, but not mTORC2 component Rictor. Moreover, sevoflurane induced pyroptosis could also be restored by suppressing mTOR activity or knocking down the expressions of Ras-related small GTPases RagA or RagC. Finally, administration of DSF or NSA dramatically improved the spatial and emotional cognitive disorders without alternation of locomotor activity. Taken together, these results indicate that mTORC1-dependent and GSDMD-mediated pyroptosis contributes to the developmental sevoflurane neurotoxicity. Characterizing these processes may provide experimental evidence for the possible prevention of developmental sevoflurane neurotoxicity.
Collapse
|
10
|
Li W, Yi Q, Shi H. Hippocampal gene expression patterns in Sevoflurane anesthesia associated neurocognitive disorders: A bioinformatic analysis. Front Neurol 2022; 13:1084874. [PMID: 36561300 PMCID: PMC9763458 DOI: 10.3389/fneur.2022.1084874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background Several studies indicate general anesthetics can produce lasting effects on cognitive function. The commonly utilized anesthetic agent Sevoflurane has been implicated in neurodegenerative processes. The present study aimed to identify molecular underpinnings of Sevoflurane anesthesia linked neurocognitive changes by leveraging publically available datasets for bioinformatics analysis. Methods A Sevoflurane anesthesia related gene expression dataset was obtained. Sevoflurane related genes were obtained from the CTD database. Neurocognitive disorders (NCD) related genes were downloaded from DisGeNET and CTD. Intersecting differentially expressed genes between Sevoflurane and NCD were identified as cross-talk genes. A protein-protein interaction (PPI) network was constructed. Hub genes were selected using LASSO regression. Single sample gene set enrichment analysis; functional network analysis, pathway correlations, composite network analysis and drug sensitivity analysis were performed. Results Fourteen intersecting cross-talk genes potentially were identified. These were mainly involved in biological processes including peptidyl-serine phosphorylation, cellular response to starvation, and response to gamma radiation, regulation of p53 signaling pathway, AGE-RAGE signaling pathway and FoxO signaling. Egr1 showed a central role in the PPI network. Cdkn1a, Egr1, Gadd45a, Slc2a1, and Slc3a2 were identified as important or hub cross-talk genes. Among the interacting pathways, Interleukin-10 signaling and NF-kappa B signaling enriched among Sevoflurane-related DEGs were highly correlated with HIF-1 signaling enriched in NCD-related genes. Composite network analysis showed Egr1 interacted with AGE-RAGE signaling and Apelin signaling pathways, Cdkn1a, and Gadd45a. Cdkn1a was implicated in in FoxO signaling, PI3K-Akt signaling, ErbB signaling, and Oxytocin signaling pathways, and Gadd45a. Gadd45a was involved in NF-kappa B signaling and FoxO signaling pathways. Drug sensitivity analysis showed Egr1 was highly sensitive to GENIPIN. Conclusion A suite of bioinformatics analysis revealed several key candidate hippocampal genes and associated functional signaling pathways that could underlie Sevoflurane associated neurodegenerative processes.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Anesthesiology, The Second Affiliated Hospital of the Shandong First Medical University, Taian, China
| | - Qijun Yi
- Department of Oncology, The Second Affiliated Hospital of the Shandong First Medical University, Taian, China
| | - Huijian Shi
- Department of Anesthesiology, The Second Affiliated Hospital of the Shandong First Medical University, Taian, China,*Correspondence: Huijian Shi
| |
Collapse
|
11
|
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: 3.5] [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
|
12
|
Xu R, Zhu Y, Jia J, Li WX, Lu Y. RIPK1/RIPK3-Mediated Necroptosis is Involved in Sevoflurane-Induced Neonatal Neurotoxicity in the Rat Hippocampus. Cell Mol Neurobiol 2022; 42:2235-2244. [PMID: 33991280 DOI: 10.1007/s10571-021-01098-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
Recent studies have shown that exposure to sevoflurane in developing brains causes neuronal apoptosis and cognitive dysfunction. "Necroptosis" is a novel pathway of necrosis. We introduced the caspase-specific inhibitor Z-VAD in addition to the receptor-interacting protein kinase 1 (RIPK1) inhibitor Nec-1, to ascertain the existence and importance of necroptosis. Sprague-Dawley rat pups postnatal day 7 were randomly assigned into one of five groups: control, sevoflurane + Z-VAD, sevoflurane + Nec-1, sevoflurane + Z-VAD + Nec-1 and 3% sevoflurane group. Neuronal apoptosis was evaluated by hematoxylin and eosin staining. The MTT assay was performed to evaluate cell viability. Immunofluorescence was employed to measure expression of RIPK1 and RIPK3. Western blots showing expression of RIPK1, RIPK3 and phosphorylation of mixed lineage kinase domain-like (p-MLKL) were used to explore the role of necroptosis. Binding of RIPK1/RIPK3 was detected via co-immunoprecipitation. Finally, the Morris water maze test was used to determine cognitive function. Exposure to 3% sevoflurane for 6 h induced neurotoxicity and inhibited cell viability. Neuron viability was low in the SEV, SEV + Z-VAD and SEV + Nec-1 groups. The study revealed that RIPK1 and RIPK3 protein expression increased significantly, but there was no significant differences between the SEV and SEV + Z-VAD groups. The expression of p-MLKL significantly increased in the SEV and SEV + Z-VAD groups, but not in the SEV + Nec-1 group or SEV + Z-VAD + Nec-1 group compared to the control group. Co-immunoprecipitation results showed that sevoflurane exposure enhanced binding of RIPK1/RIPK3 protein significantly. Blockade of apoptosis and necroptosis alleviated sevoflurane-induced cognitive impairment. Sevoflurane exposure elicited neurotoxicity within neonatal hippocampal neurons and tissues. Blockade of apoptosis or necroptosis alone did not attenuate sevoflurane-induced neurotoxicity (SIN). RIPK1/RIPK3-mediated necroptosis was involved in SIN in hippocampal neurons. SIN could be attenuated only by inhibiting both apoptosis and necroptosis.
Collapse
Affiliation(s)
- Rui Xu
- Department of Anesthesiology, The Eye, Ear, Nose and Throat Hospital, Fudan University, Fenyang Road #83, Shanghai, 200031, People's Republic of China
| | - Yun Zhu
- Department of Oro-Maxillofacial Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jie Jia
- Department of Anesthesiology, The Eye, Ear, Nose and Throat Hospital, Fudan University, Fenyang Road #83, Shanghai, 200031, People's Republic of China
| | - Wen Xian Li
- Department of Anesthesiology, The Eye, Ear, Nose and Throat Hospital, Fudan University, Fenyang Road #83, Shanghai, 200031, People's Republic of China.
| | - Yi Lu
- Department of Anesthesiology, The Eye, Ear, Nose and Throat Hospital, Fudan University, Fenyang Road #83, Shanghai, 200031, People's Republic of China.
| |
Collapse
|
13
|
Chen H, Peng Y, Wang L, Wang X. Sevoflurane attenuates cognitive dysfunction and NLRP3-dependent caspase-1/11-GSDMD pathway-mediated pyroptosis in the hippocampus via upregulation of SIRT1 in a sepsis model. Arch Physiol Biochem 2022; 128:1413-1420. [PMID: 32538180 DOI: 10.1080/13813455.2020.1773860] [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] [Indexed: 10/24/2022]
Abstract
Septic encephalopathy (SE) is a devastating consequence of sepsis, a hyper-triggered host response against infectious challenge, which ultimately leads to brain damage. The present study examined whether sevoflurane (SVF), a volatile anaesthetic, can counteract the perturbation of homeostasis in a caecal ligation and puncture (CLP)-induced mouse model of SE. SVF enhances neurocognition in terms of spatial memory improvement via counter-regulation of activated oxidative-inflammatory stress and pyroptotic processes in SE. Further, the beneficial effects of SVF against SE are mediated by activation of silent information regulator 1 (SIRT1)-mediated reduction of reactive oxygen species (ROS) level, regulation of thioredoxin (TXN) and thioredoxin interacting protein (TIP) levels, reduction of inflammatory-pyroptotic signalling (NLRP3, caspase 1/11, GSDMD, TLR4 and TRIF) proteins, as well as a reduction of inflammatory cytokine (IL-1β and IL-18) levels. These findings suggest that SVF may have therapeutic potential for the treatment of SE and associated cognitive malfunction.
Collapse
Affiliation(s)
- Hao Chen
- Department of Anesthesiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yi Peng
- Department of Anesthesiology, Shijiazhuang First Hospital, Shijiazhuang, China
| | - Li Wang
- Department of Anesthesiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Wang
- Department of Anesthesiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Wong-Kee-You AMB, Loveridge-Easther C, Mueller C, Simon N, Good WV. The impact of early exposure to general anesthesia on visual and neurocognitive development. Surv Ophthalmol 2022; 68:539-555. [PMID: 35970232 DOI: 10.1016/j.survophthal.2022.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022]
Abstract
Every year millions of children are exposed to general anesthesia while undergoing surgical and diagnostic procedures. In the field of ophthalmology, 44,000 children are exposed to general anesthesia annually for strabismus surgery alone. While it is clear that general anesthesia is necessary for sedation and pain minimization during surgical procedures, the possibility of neurotoxic impairments from its exposure is of concern. In animals there is strong evidence linking early anesthesia exposure to abnormal neural development. but in humans the effects of anesthesia are debated. In humans many aspects of vision develop within the first year of life, making the visual system vulnerable to early adverse experiences and potentially vulnerable to early exposure to general anesthesia. We attempt to address whether the visual system is affected by early postnatal exposure to general anesthesia. We first summarize key mechanisms that could account for the neurotoxic effects of general anesthesia on the developing brain and review existing literature on the effects of early anesthesia exposure on the visual system in both animals and humans and on neurocognitive development in humans. Finally, we conclude by proposing future directions for research that could address unanswered questions regarding the impact of general anesthesia on visual development.
Collapse
Affiliation(s)
| | - Cam Loveridge-Easther
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA; University of Auckland, Auckland, New Zealand
| | - Claudia Mueller
- Sutter Health, San Francisco, CA, USA; Stanford Children's Health, Palo Alto, CA, USA
| | | | - William V Good
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA.
| |
Collapse
|
15
|
HDAC6 Inhibition Alleviates Anesthesia and Surgery-Induced Less Medial Prefrontal-Dorsal Hippocampus Connectivity and Cognitive Impairment in Aged Rats. Mol Neurobiol 2022; 59:6158-6169. [PMID: 35882756 DOI: 10.1007/s12035-022-02959-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022]
Abstract
To investigate the underlying mechanisms of postoperative cognitive dysfunction and the impairment of medial prefrontal cortex-hippocampus connectivity. Postoperative cognitive dysfunction frequently affects elderly following surgery. The role of inter-brain-region connectivity abnormality after anesthesia and surgery on postoperative cognitive dysfunction development remains unclear. Medial prefrontal cortex-hippocampus connectivity of aged and adult rats was evaluated by injecting neurotracer biotinylated dextranamine (BDA) into bilateral hippocampus 3 days before partial hepatectomy, and biotinylated dextranamine positive cells of medial prefrontal cortex 2 days after hepatectomy were counted. HDAC6 shRNA was injected into medial prefrontal cortex and hippocampus bilaterally before hepatectomy or an HDAC6 activity inhibitor Tubastatin A was administered systemically after hepatectomy. Neuroinflammation and HDAC6 down-target ac-tubulin in medial prefrontal cortex and hippocampus were detected. Learning and memory of rats were evaluated by Barnes Maze task during 2-5 days after surgery and delayed matching-to-place water maze task during 10-23 days after surgery. Compared to the age-matched normal controls, anesthesia and surgery significantly decreased BDA-positive neurons in medial prefrontal cortex of aged rats, but not young adult rats. Local HDAC6 knockdown and systemic HDAC6 inhibition both increased BDA-positive neurons number of medial prefrontal cortex, alleviated learning and memory impairment in the Barnes Maze task and water maze task, decreased HDAC6 expression, inflammatory cytokines, astrocyte and microglial activation, and increased ac-tubulin expression in aged rats which received surgery. Our data indicated that anesthesia and surgery impaired medial prefrontal cortex-hippocampus connectivity and cognition which was associated with HDAC6 overexpression.
Collapse
|
16
|
Scutellaria baicalensis and its constituents baicalin and baicalein as antidotes or protective agents against chemical toxicities: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1297-1329. [PMID: 35676380 DOI: 10.1007/s00210-022-02258-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022]
Abstract
Scutellaria baicalensis (SB), also known as the Chinese skullcap, has a long history of being used in Chinese medicine to treat a variety of conditions ranging from microbial infections to metabolic syndrome and malignancies. Numerous studies have reported that treatment with total SB extract or two main flavonoids found in its root and leaves, baicalin (BA) and baicalein (BE), can prevent or alleviate the detrimental toxic effects of exposure to various chemical compounds. It has been shown that BA and BE are generally behind the protective effects of SB against toxicants. This paper aimed to review the protective and therapeutic effects of SB and its main components BA and BE against chemical compounds that can cause intoxication after acute or chronic exposure and seriously affect different vital organs including the brain, heart, liver, and kidneys. In this review paper, we had a look into a total of 221 in vitro and in vivo studies from 1995 to 2021 from the scientific databases PubMed, Scopus, and Web of Science which reported protective or therapeutic effects of BA, BE, or SB against drugs and chemicals that one might be exposed to on a professional or accidental basis and compounds that are primarily used to simulate disease models. In conclusion, the protective effects of SB and its flavonoids can be mainly attributed to increase in antioxidants enzymes, inhibition of lipid peroxidation, reduction of inflammatory cytokines, and suppression of apoptosis pathway.
Collapse
|
17
|
Fu N, Zhu R, Zeng S, Li N, Zhang J. Effect of Anesthesia on Oligodendrocyte Development in the Brain. Front Syst Neurosci 2022; 16:848362. [PMID: 35664684 PMCID: PMC9158484 DOI: 10.3389/fnsys.2022.848362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Oligodendrocytes (OLs) participate in the formation of myelin, promoting the propagation of action potentials, and disruption of their proliferation and differentiation leads to central nervous system (CNS) damage. As surgical techniques have advanced, there is an increasing number of children who undergo multiple procedures early in life, and recent experiments have demonstrated effects on brain development after a single or multiple anesthetics. An increasing number of clinical studies showing the effects of anesthetic drugs on the development of the nervous system may mainly reside in the connections between neurons, where myelin development will receive more research attention. In this article, we review the relationship between anesthesia exposure and the brain and OLs, provide new insights into the development of the relationship between anesthesia exposure and OLs, and provide a theoretical basis for clinical prevention of neurodevelopmental risks of general anesthesia drugs.
Collapse
|
18
|
Linassi F, De Laurenzis A, Maran E, Gadaldi A, Spano' L, Gerosa G, Pittarello D, Zanatta P, Carron M. Influence of Previous General Anesthesia on Cognitive Impairment: An Observational Study Among 151 Patients. Front Hum Neurosci 2022; 16:810046. [PMID: 35652004 PMCID: PMC9148968 DOI: 10.3389/fnhum.2022.810046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/14/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Preoperative neurocognitive disorder (preO-NCD) is a common condition affecting 14–51. 7% of the elderly population. General anesthesia has already been associated with the one-year post-operative neurocognitive disorder (PostO-NCD), specifically, a deficit in executive function, measured by the Trail Making Test B (TMT-B), but its long-term effects on cognitive function have not been investigated. We aimed to detect preO-NCD prevalence in patients scheduled for cardiac surgery and further investigate the possible role of previous general anesthesia (pGA) in general preoperative cognitive status [measured via the Montreal Cognitive Assessment (MoCA)] and/or in executive functioning (measured via TMT-B). Methods In this observational, prospective study, 151 adult patients scheduled for elective cardiac surgery underwent MoCA and TMT-B. Data on age, education, pGA, comorbidities, and laboratory results were collected. Results We discovered a general cognitive function impairment of 79.5% and an executive function impairment of 22%. Aging is associated with an increased likelihood (OR 2.99, p = 0.047) and education with a decreased likelihood (OR 0.35, p = 0.0045) of general cognitive impairment, but only education was significantly associated with a decreased likelihood (OR 0.22, p = 0.021) of executive function impairment. While pGA did not significantly affect preO-NCD, a noteworthy interaction between aging and pGA was found, resulting in a synergistic effect, increasing the likelihood of executive function impairment (OR 9.740, p = 0.0174). Conclusion We found a high prevalence of preO-NCD in patients scheduled for cardiac surgery. General cognitive function impairment is highly associated with advancing age (not pGA). However, older patients with at least one pGA appeared to be at an increased risk of preO-NCD, especially executive function impairment, suggesting that TMT-B should be associated with MoCA in the preoperative cognitive evaluation in this population.
Collapse
Affiliation(s)
- Federico Linassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- *Correspondence: Federico Linassi
| | - Alessandro De Laurenzis
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| | - Eleonora Maran
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| | - Alessandra Gadaldi
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| | - Leonardo Spano'
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| | - Gino Gerosa
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Demetrio Pittarello
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| | - Paolo Zanatta
- Department of Anesthesia and Intensive Care, Integrated University Hospital of Verona, Verona, Italy
| | - Michele Carron
- Department of Medicine, Anaesthesiology and Intensive Care, University of Padova, Padova, Italy
| |
Collapse
|
19
|
Risk of Dementia According to Surgery Type: A Nationwide Cohort Study. J Pers Med 2022; 12:jpm12030468. [PMID: 35330467 PMCID: PMC8955036 DOI: 10.3390/jpm12030468] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/19/2022] Open
Abstract
The relationship between dementia and surgery remains unclear. Research to elucidate the relationship between them is scarce, and conducting epidemiological research is complicated. This study aimed to investigate the incidence and risk of dementia according to the surgery type. We performed a retrospective propensity score-matched cohort study using nationwide representative cohort sample data from the Korean National Health Insurance Service in South Korea between 2003 and 2004. Incidence rates for dementia were obtained by dividing the number of patients with dementia by person-years at risk. To identify the risk of dementia according to the type of surgery, we investigated the hazard ratio by each surgery type. The incidence rates of dementia in control, musculoskeletal, and two or more surgeries groups were 9.66, 13.47, and 13.36 cases per 1000 person-years, respectively. The risk of dementia in the musculoskeletal and two or more surgeries groups was 1.44-fold higher (95% confidence interval (95% CI), 1.22–1.70) and 1.42-fold higher (95% CI, 1.17–1.72) than that in the control group, respectively. Patients who underwent musculoskeletal surgery and two or more surgeries had a higher risk of dementia; however, there was no association with the type of anesthesia administered.
Collapse
|
20
|
Liu H, Meng X, Li Y, Chen S, Ji Y, Song S, Ji F, Jin X. Neonatal exposure to sevoflurane impairs preference for social novelty in C57BL/6 female mice at early-adulthood. Biochem Biophys Res Commun 2022; 593:129-136. [PMID: 35063768 DOI: 10.1016/j.bbrc.2022.01.022] [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: 12/23/2021] [Accepted: 01/08/2022] [Indexed: 11/28/2022]
Abstract
Social interaction deficit is core symptom of children with autism, owing to interaction of genetic predisposition and environmental toxins. Sevoflurane could induce neurotoxicity in developing brain in rodent models. This study aims to investigate whether sevoflurane anesthesia in neonatal period could impair social behaviors in male and female mice. Twenty-eight male and thirty-one female mice were randomly assigned to receive 3.0% sevoflurane or 60% oxygen on postnatal day 6. They were tested for social interaction behaviors at one- and two-month-old. In addition, the cortex and hippocampus of neonatal mice undergoing sevoflurane anesthesia were harvested for immunoblotting analysis. As a result, both male and female mice undergoing sevoflurane anesthesia showed strong sociability and weak preference for social novelty at juvenile age. In addition, the male mice developed normal preference for social novelty at early-adulthood; However, the female mice remained weak preference for social novelty. Furthurmore, sevoflurane anesthesia could decrease the levels of PSD95 but not Neuroligin-1 in the hippocampus but not cortex of neonatal mice. In conclusion, sevoflurane anesthesia in neonatal period could disturb development of social memory and impair preference for social novelty in female mice at early-adulthood, with the potential mechanism of decreasing PSD95 expression in the hippocampus of C57BL/6 mice.
Collapse
Affiliation(s)
- Huayue Liu
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, PR China; Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, PR China
| | - Xiaowen Meng
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, PR China; Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, PR China
| | - Yixuan Li
- Suzhou Medical College of Soochow University, Suzhou, 215123, PR China
| | - Shiwen Chen
- Suzhou Medical College of Soochow University, Suzhou, 215123, PR China
| | - Yumeng Ji
- Suzhou Medical College of Soochow University, Suzhou, 215123, PR China
| | - Shaoyong Song
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, PR China; Department of Pain Medicine, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215124, PR China
| | - Fuhai Ji
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, PR China; Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, PR China.
| | - Xin Jin
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, PR China; Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, PR China.
| |
Collapse
|
21
|
Liu L, Zhang X, Wang C, Wu X, Long B. Hypercholesterolemia aggravates sevoflurane-induced cognitive impairment in aged rats by inducing neurological inflammation and apoptosis. J Biochem Mol Toxicol 2022; 36:e23009. [PMID: 35174938 DOI: 10.1002/jbt.23009] [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: 09/24/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/30/2022]
Abstract
We aimed to explore the effects of hypercholesterolemia on sevoflurane-induced cognitive impairment in aged rats and the underlying mechanism(s). Aged rats were administrated with high-fat diet, sevoflurane, or both. Thereafter, the plasma levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) were evaluated. The Morris water maze task was performed to evaluate the spatial learning and memory ability of rats. Moreover, Nissl and Evans blue staining were conducted to test nerve damage and detect the blood-brain barrier permeability, respectively. The percentage of apoptotic cells was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. The messenger RNA expression of inflammatory factors and protein expression of microglial activation markers and apoptosis-related proteins were tested by real-time polymerase chain reaction, enzyme-linked immunosorbent assay, or western blot analysis, respectively. High-fat diet induced high levels of TC, TG, and LDL but decreased levels of HDL. However, sevoflurane had no effects on these levels. In contrast, sevoflurane significantly induced the impairment of learning and memory, nerve damage, neuroinflammatory damage, and neuronal apoptosis. Hypercholesterolemia exacerbated the sevoflurane-induced impairment in aged rats. These results suggested that hypercholesterolemia aggravates sevoflurane-induced cognitive impairment in aged rats, possibly by inducing neurological inflammation and apoptosis.
Collapse
Affiliation(s)
- Lidan Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cong Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Long
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
22
|
Tian D, Xing Y, Gao W, Zhang H, Song Y, Tian Y, Dai Z. Sevoflurane Aggravates the Progress of Alzheimer’s Disease Through NLRP3/Caspase-1/Gasdermin D Pathway. Front Cell Dev Biol 2022; 9:801422. [PMID: 35127716 PMCID: PMC8807556 DOI: 10.3389/fcell.2021.801422] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is the most common form of dementia worldwide. Previous studies have reported that sevoflurane, a frequently used anesthetic, can induce cognitive impairment in preclinical and clinical settings. However, the mechanism underlying the development of this neurotoxicity is currently unclear. Methods: Seven-month-old APP/PS1 mice were placed in an anesthesia induction box containing 3% sevoflurane in 100% O2 for 6 h, while BV2 cells were cultured with 4% sevoflurane for 6 h. Pyroptosis and tau protein expression in excised hippocampus tissues and cells were measured using Western blotting and immunofluorescence assay. Caspase-1 and NLRP3 were knocked out in BV2 microglia using CRISPR/Cas9 technology to determine whether they mediate the effects induced by sevoflurane. Results: Sevoflurane directly activated caspase-1 to induce pyroptosis in the mouse model of AD via NLRP3 and AIM2 activation. In addition, sevoflurane mediated cleavage of gasdermin D (GSDMD) but not gasdermin E (GSDME), promoted the biosynthesis of downstream interleukin-1β and interleukin-18, and increased β-amyloid (Aβ) deposition and tau phosphorylation. The nontoxic caspase-1 small-molecule inhibitor VX-765 significantly inhibited this activation process in microglia, while NLRP3 deletion suppressed sevoflurane-induced caspase-1 cleavage and subsequently pyroptosis, as well as tau pathology. Furthermore, silencing caspase-1 alleviated the sevoflurane-induced release of IL-1β and IL-18 and inhibited tau-related enzymes in microglia. Conclusion: This study is the first to report that clinical doses of sevoflurane aggravate the progression of AD via the NLRP3/caspase-1/GSDMD axis. Collectively, our findings elucidate the crucial mechanisms of NLRP3/caspase-1 in pyroptosis and tau pathogenesis induced by sevoflurane and suggest that VX-765 could represent a novel therapeutic intervention for treating AD.
Collapse
Affiliation(s)
- Di Tian
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Anesthesiology, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Yanmei Xing
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Anesthesiology, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Wenli Gao
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Anesthesiology, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Hongyan Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Yifeng Song
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Ya Tian
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
| | - Zhongliang Dai
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Anesthesiology, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen, China
- *Correspondence: Zhongliang Dai,
| |
Collapse
|
23
|
Sun M, Xie Z, Zhang J, Leng Y. Mechanistic insight into sevoflurane-associated developmental neurotoxicity. Cell Biol Toxicol 2022; 38:927-943. [PMID: 34766256 PMCID: PMC9750936 DOI: 10.1007/s10565-021-09677-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023]
Abstract
With the development of technology, more infants receive general anesthesia for surgery, other interventions, or clinical examination at an early stage after birth. However, whether general anesthetics can affect the function and structure of the developing infant brain remains an important, complex, and controversial issue. Sevoflurane is the most-used anesthetic in infants, but this drug is potentially neurotoxic. Short or single exposure to sevoflurane has a weak effect on cognitive function, while long or repeated exposure to general anesthetics may cause cognitive dysfunction. This review focuses on the mechanisms by which sevoflurane exposure during development may induce long-lasting undesirable effects on the brain. We review neural cell death, neural cell damage, impaired assembly and plasticity of neural circuits, tau phosphorylation, and neuroendocrine effects as important mechanisms for sevoflurane-induced developmental neurotoxicity. More advanced technologies and methods should be applied to determine the underlying mechanism(s) and guide prevention and treatment of sevoflurane-induced neurotoxicity. 1. We discuss the mechanisms underlying sevoflurane-induced developmental neurotoxicity from five perspectives: neural cell death, neural cell damage, assembly and plasticity of neural circuits, tau phosphorylation, and neuroendocrine effects.
2. Tau phosphorylation, IL-6, and mitochondrial dysfunction could interact with each other to cause a nerve damage loop.
3. miRNAs and lncRNAs are associated with sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Mingyang Sun
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China 730000 ,Department of Anesthesiology and Perioperative Medicine, Center for Clinical Single Cell Biomedicine, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan People’s Republic of China 450003
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, Center for Clinical Single Cell Biomedicine, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan People’s Republic of China 450003
| | - Yufang Leng
- Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu People’s Republic of China 730000
| |
Collapse
|
24
|
Cong L, Cong Y, Feng N, Liang W, Wu Y. Up-regulated microRNA-132 reduces the cognition-damaging effect of sevoflurane on Alzheimer's disease rats by inhibiting FOXA1. Genomics 2021; 113:3644-3652. [PMID: 34400241 DOI: 10.1016/j.ygeno.2021.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 07/30/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Some studies have implied the damaging effect of sevoflurane (sevo) on cognitive function in Alzheimer's disease (AD). This research was conducted to explore the effect of microRNA (miR)-132/forkhead-box A1 (FOXA1) axis on cognitive ability of sevo-treated AD rats. METHODS The condensed-matter Aβ1-40-induced AD rats were injected with miR-132- or FOXA1-related plasmids, followed by inhalation with 3% sevo. Then, the cognitive functions of AD rats were assessed. miR-132 and FOXA1 levels in hippocampal tissues of AD rats, and their interaction were identified. RESULTS miR-132 expression was reduced and FOXA1 mRNA and protein levels were elevated in AD rats. miR-132 targeted FOXA1. Sevo treatment impaired cognitive function in AD rats. Elevated miR-132 or inhibited FOXA1 attenuated sevo-mediated injury in AD rats. Overexpressed FOXA1 rescued the effect of elevated miR-132 in AD rats with sevo treatment. CONCLUSION Up-regulated miR-132 reduces the cognition-damaging effect of sevo on AD rats by inhibiting FOXA1.
Collapse
Affiliation(s)
- Lin Cong
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang, People's Republic of China
| | - Yuena Cong
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang, People's Republic of China
| | - Nianping Feng
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang, People's Republic of China
| | - Weiwei Liang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang, People's Republic of China
| | - Yun Wu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang, People's Republic of China.
| |
Collapse
|
25
|
Hofmann C, Sander A, Wang XX, Buerge M, Jungwirth B, Borgstedt L, Kreuzer M, Kopp C, Schorpp K, Hadian K, Wotjak CT, Ebert T, Ruitenberg M, Parsons CG, Rammes G. Inhalational Anesthetics Do Not Deteriorate Amyloid-β-Derived Pathophysiology in Alzheimer's Disease: Investigations on the Molecular, Neuronal, and Behavioral Level. J Alzheimers Dis 2021; 84:1193-1218. [PMID: 34657881 DOI: 10.3233/jad-201185] [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: 11/15/2022]
Abstract
BACKGROUND Studies suggest that general anesthetics like isoflurane and sevoflurane may aggravate Alzheimer's disease (AD) neuropathogenesis, e.g., increased amyloid-β (Aβ) protein aggregation resulting in synaptotoxicity and cognitive dysfunction. Other studies showed neuroprotective effects, e.g., with xenon. OBJECTIVE In the present study, we want to detail the interactions of inhalational anesthetics with Aβ-derived pathology. We hypothesize xenon-mediated beneficial mechanisms regarding Aβ oligomerization and Aβ-mediated neurotoxicity on processes related to cognition. METHODS Oligomerization of Aβ 1-42 in the presence of anesthetics has been analyzed by means of TR-FRET and silver staining. For monitoring changes in neuronal plasticity due to anesthetics and Aβ 1-42, Aβ 1-40, pyroglutamate-modified amyloid-(AβpE3), and nitrated Aβ (3NTyrAβ), we quantified long-term potentiation (LTP) and spine density. We analyzed network activity in the hippocampus via voltage-sensitive dye imaging (VSDI) and cognitive performance and Aβ plaque burden in transgenic AD mice (ArcAβ) after anesthesia. RESULTS Whereas isoflurane and sevoflurane did not affect Aβ 1-42 aggregation, xenon alleviated the propensity for aggregation and partially reversed AβpE3 induced synaptotoxic effects on LTP. Xenon and sevoflurane reversed Aβ 1-42-induced spine density attenuation. In the presence of Aβ 1-40 and AβpE3, anesthetic-induced depression of VSDI-monitored signaling recovered after xenon, but not isoflurane and sevoflurane removal. In slices pretreated with Aβ 1-42 or 3NTyrAβ, activity did not recover after washout. Cognitive performance and plaque burden were unaffected after anesthetizing WT and ArcAβ mice. CONCLUSION None of the anesthetics aggravated Aβ-derived AD pathology in vivo. However, Aβ and anesthetics affected neuronal activity in vitro, whereby xenon showed beneficial effects on Aβ 1-42 aggregation, LTP, and spine density.
Collapse
Affiliation(s)
- Carolin Hofmann
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Annika Sander
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xing Xing Wang
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martina Buerge
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bettina Jungwirth
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Anesthesiology, University Hospital Ulm, Ulm, Germany
| | - Laura Borgstedt
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Kreuzer
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claudia Kopp
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Neuronal Plasticity, Munich, Germany.,Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Tim Ebert
- Max Planck Institute of Psychiatry, Neuronal Plasticity, Munich, Germany
| | | | | | - Gerhard Rammes
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
26
|
Xu M, Feng J, Tang M, Guo Q, Zhan J, Zhu F, Lei H, Kang Q. Blocking retrograde axonal transport of autophagosomes contributes to sevoflurane-induced neuron apoptosis in APP/PS1 mice. Acta Neurol Belg 2021; 121:1207-1215. [PMID: 32385796 PMCID: PMC8443487 DOI: 10.1007/s13760-020-01359-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Autophagy, a crucial pathway for the degradation of proteins in eukaryotic cells, is linked to the development of Alzheimer's disease (AD), and the accumulated autophagosomes in the cells resulting in the death of cells. Sevoflurane can impair spatial learning and memory in mice with AD and lead to the apoptosis of nerve cells; however, the underlying mechanisms remain unknown. We aim to explore the effects and underlying mechanisms of sevoflurane in APPswe/PS1ΔE9 double-transgenic mice. 51 heterozygous APPswe/PS1ΔE9 double-transgenic mice were involved and divided into three groups, including control group, sham group and sevoflurane group. Morris water maze experiment was used to test the learning and memory abilities of mice, flow cytometry was conducted to detect apoptosis and mitochondrial membrane potential of brain cells in mice, transmission electron microscopy was used to observe the number of autophagosomes at the axon in mice, and western blot was carried out to detect the expression of Bax, Bcl-2, LC3II, P62, KIF3B and DIC proteins of brain cells in mice. In our study, we found that significantly longer escape latencies, fewer crossings of the platform and shorter time spent in the target quadrant of the morris water maze experiment in the sevoflurane group. Flow cytometry showed cellular apoptosis was increased and the membrane potential of the mitochondria was reduced of brain cells in the sevoflurane group. Transmission electron microscopy displayed that there was a remarkable upregulation of autophagosomes at the axon of brain cells in mice after treatment of sevoflurane. Western blot demonstrated that the expression of Bax, LC3II, P62 and KIF3B proteins were elevated, and the expression of Bcl-2 and DIC proteins were reduced in the sevoflurane group. Sevoflurane impaired acquisition learning and memory function, promoted the apoptosis of hippocampal neurons in APPswe/PS1ΔE9 double-transgenic mice, and the mechanism might be related to the activation of autophagy along with the disruption of autophagosomes retrograde transport in axons.
Collapse
Affiliation(s)
- Mingliang Xu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Mingxi Tang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Qingxi Guo
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Jian Zhan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Fuzu Zhu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Hong Lei
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Qingmei Kang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.
| |
Collapse
|
27
|
Inhibiting PDE7A Enhances the Protective Effects of Neural Stem Cells on Neurodegeneration and Memory Deficits in Sevoflurane-Exposed Mice. eNeuro 2021; 8:ENEURO.0071-21.2021. [PMID: 34135002 PMCID: PMC8266220 DOI: 10.1523/eneuro.0071-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/03/2022] Open
Abstract
Sevoflurane is widely used in general anesthesia, especially for children. However, prolonged exposure to sevoflurane is reported to be associated with adverse effects on the development of brain in infant monkey. Neural stem cells (NSCs), with potent proliferation, differentiation, and renewing ability, provide an encouraging tool for basic research and clinical therapies for neurodegenerative diseases. We aim to explore the functional effects of injecting NSCs with phosphodiesterase 7A (PDE7A) knock-down in infant mice exposed to sevoflurane. The effects of PDE7A in NSCs proliferation and differentiation were determined by cell counting kit-8 (CCK-8) assay and differentiation-related gene expression assay, respectively. The effects of NSCs with modified PDE7A on mice’s long-term memory and learning ability were assessed by behavioral assays. Our data demonstrated that depleting PDE7A promoted, whereas forcing PDE7A suppressed the activation of cAMP/cAMP-response element binding protein (CREB) signaling as well as cell proliferation and neuronal differentiation of NSCs. Inhibition of PDE7A in NSCs exhibited profound improved effects on long-term memory and learning ability of mice exposed to sevoflurane. Our results for the first time show that knock-down of PDE7A improves the neurogenesis of NSCs in vitro and in vivo, and is beneficial for alleviating sevoflurane-induced brain damage in infant mice.
Collapse
|
28
|
Neonatal administration of a subanaesthetic dose of JM-1232(-) in mice results in no behavioural deficits in adulthood. Sci Rep 2021; 11:12874. [PMID: 34145371 PMCID: PMC8213711 DOI: 10.1038/s41598-021-92344-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 06/08/2021] [Indexed: 11/09/2022] Open
Abstract
In animal models, neonatal exposure of general anaesthetics significantly increases apoptosis in the brain, resulting in persistent behavioural deficits later in adulthood. Consequently, there is growing concern about the use of general anaesthetics in obstetric and paediatric practice. JM-1232(−) has been developed as a novel intravenous anaesthetic, but the effects of JM-1232(−) on the developing brain are not understood. Here we show that neonatal administration of JM-1232(−) does not lead to detectable behavioural deficits in adulthood, contrarily to other widely-used intravenous anaesthetics. At postnatal day 6 (P6), mice were injected intraperitoneally with a sedative-equivalent dose of JM-1232(−), propofol, or midazolam. Western blot analysis of forebrain extracts using cleaved poly-(adenosine diphosphate-ribose) polymerase antibody showed that JM-1232(−) is accompanied by slight but measurable apoptosis 6 h after administration, but it was relatively small compared to those of propofol and midazolam. Behavioural studies were performed in adulthood, long after the neonatal anaesthesia, to evaluate the long-term effects on cognitive, social, and affective functions. P6 administration to JM-1232(−) was not accompanied by detectable long-term behavioural deficits in adulthood. However, animals receiving propofol or midazolam had impaired social and/or cognitive functions. These data suggest that JM-1232(−) has prospects for use in obstetric and paediatric practice.
Collapse
|
29
|
Jiang T, Xu S, Shen Y, Xu Y, Li Y. Genistein Attenuates Isoflurane-Induced Neuroinflammation by Inhibiting TLR4-Mediated Microglial-Polarization in vivo and in vitro. J Inflamm Res 2021; 14:2587-2600. [PMID: 34168482 PMCID: PMC8216758 DOI: 10.2147/jir.s304336] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background Isoflurane, a widely used anesthetic in surgery, has been found to induce neurotoxicity. In parallel, genistein is thought to attenuate isoflurane-induced neurotoxicity, although underlying molecular mechanisms are still unclear. In this study, we studied the protective effects of genistein on isoflurane-induced neuroinflammation in rats and BV2 cells. Methods Sprague-Dawley rat pups were exposed to 0.75% isoflurane for 6 hours at postnatal day 7 (P7), and genistein (20, 40, or 80 mg/kg/day) or saline administered from P3 to P15. Hippocampal single-cell suspensions were prepared and apoptosis analyzed by flow cytometry. mRNA expression was determined by RT-qPCR, while protein expression was assessed using Western blot, immunochemistry and immunofluorescence. TLR4 was knocked-out in BV2 cells through CRISPR-Cas9. Results Genistein treatment reduced isoflurane-induced apoptosis and inflammation in rat hippocampus. Importantly, genistein promoted M2 and suppressed M1 microglia polarization in rat hippocampus after stimulation with isoflurane. In addition, genistein reduced isoflurane-induced protein expression levels of TLR4, MyD88, TRAF6, p-TAK1, p-p38, p-ERK, p-IκBα and p-NF-κB in rat hippocampus. In BV2 cells exposed to isoflurane, genistein treatment decreased IL-1β, TNF-α, IL-6 and IL-8 mRNA expressions, promoted M2 and suppressed M1 microglia polarization. Similarly, genistein also decreased TLR4 protein levels in isoflurane-induced BV2 cells. However, genistein did not affect CD16, iNOS, CD206 and Arg1 protein levels in TLR4-KO BV2 cells exposed to isoflurane. Conclusion Genistein attenuates isoflurane-induced neurotoxicity by inhibiting TLR4-mediated microglial inflammation in vivo and in vitro.
Collapse
Affiliation(s)
- Tao Jiang
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Shoucai Xu
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yangyang Shen
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yong Xu
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yuwen Li
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| |
Collapse
|
30
|
Dong Y, Liang F, Huang L, Fang F, Yang G, Tanzi RE, Zhang Y, Quan Q, Xie Z. The anesthetic sevoflurane induces tau trafficking from neurons to microglia. Commun Biol 2021; 4:560. [PMID: 33980987 PMCID: PMC8115254 DOI: 10.1038/s42003-021-02047-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/29/2021] [Indexed: 01/08/2023] Open
Abstract
Accumulation and spread of tau in Alzheimer's disease and other tauopathies occur in a prion-like manner. However, the mechanisms and downstream consequences of tau trafficking remain largely unknown. We hypothesized that tau traffics from neurons to microglia via extracellular vesicles (EVs), leading to IL-6 generation and cognitive impairment. We assessed mice and neurons treated with anesthetics sevoflurane and desflurane, and applied nanobeam-sensor technology, an ultrasensitive method, to measure tau/p-tau amounts. Sevoflurane, but not desflurane, increased tau or p-tau amounts in blood, neuron culture medium, or EVs. Sevoflurane increased p-tau amounts in brain interstitial fluid. Microglia from tau knockout mice took up tau and p-tau when treated with sevoflurane-conditioned neuron culture medium, leading to IL-6 generation. Tau phosphorylation inhibitor lithium and EVs generation inhibitor GW4869 attenuated tau trafficking. GW4869 mitigated sevoflurane-induced cognitive impairment in mice. Thus, tau trafficking could occur from neurons to microglia to generate IL-6, leading to cognitive impairment.
Collapse
Affiliation(s)
- Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Feng Liang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Lining Huang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Fang Fang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Qimin Quan
- Rowland Institute at Harvard University, Cambridge, MA, USA
- NanoMosaic, Woburn, MA, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
31
|
Lee JR, Joseph B, Hofacer RD, Upton B, Lee SY, Ewing L, Zhang B, Danzer SC, Loepke AW. Effect of dexmedetomidine on sevoflurane-induced neurodegeneration in neonatal rats. Br J Anaesth 2021; 126:1009-1021. [PMID: 33722372 DOI: 10.1016/j.bja.2021.01.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Structural brain abnormalities in newborn animals after prolonged exposure to all routinely used general anaesthetics have raised substantial concerns for similar effects occurring in millions of children undergoing surgeries annually. Combining a general anaesthetic with non-injurious sedatives may provide a safer anaesthetic technique. We tested dexmedetomidine as a mitigating therapy in a sevoflurane dose-sparing approach. METHODS Neonatal rats were randomised to 6 h of sevoflurane 2.5%, sevoflurane 1% with or without three injections of dexmedetomidine every 2 h (resulting in 2.5, 5, 10, 25, 37.5, or 50 μg kg-1 h-1), or fasting in room air. Heart rate, oxygen saturation, level of hypnosis, and response to pain were measured during exposure. Neuronal cell death was quantified histologically after exposure. RESULTS Sevoflurane at 2.5% was more injurious than at 1% in the hippocampal cornu ammonis (CA)1 and CA2/3 subfields; ventral posterior and lateral dorsal thalamic nuclei; prefrontal, retrosplenial, and somatosensory cortices; and subiculum. Although sevoflurane 1% did not provide complete anaesthesia, supplementation with dexmedetomidine dose dependently increased depth of anaesthesia and diminished responses to pain. The combination of sevoflurane 1% and dexmedetomidine did not reliably reduce neuronal apoptosis relative to an equianaesthetic dose of sevoflurane 2.5%. CONCLUSIONS A sub-anaesthetic dose of sevoflurane combined with dexmedetomidine achieved a level of anaesthesia comparable with that of sevoflurane 2.5%. Similar levels of anaesthesia caused comparable programmed cell death in several developing brain regions. Depth of anaesthesia may be an important factor when comparing the neurotoxic effects of different anaesthetic regimens.
Collapse
Affiliation(s)
- Jeong-Rim Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul
| | - Bernadin Joseph
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Brian Upton
- Medical Scientist Training Program, University of Cincinnati, Cincinnati, OH, USA
| | - Samuel Y Lee
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA
| | - Loren Ewing
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA
| | - Bingqing Zhang
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Steve C Danzer
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA
| | - Andreas W Loepke
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Yu L, Wen G, Zhu S, Hu X, Huang C, Yang Y. Abnormal phosphorylation of tau protein and neuroinflammation induced by laparotomy in an animal model of postoperative delirium. Exp Brain Res 2021; 239:867-880. [PMID: 33409674 DOI: 10.1007/s00221-020-06007-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Postoperative delirium (POD) is an acute neuropsychological disturbance after surgery, whose prevalence is related with advancing age. Neuroinflammation and abnormal tau phosphorylation that commonly presenting in Alzheimer's disease (AD) may contribute to the progression and duration of POD. To study the acute influence of surgery on cognitive function, wild type male C57BL/6 N mice were randomly divided into three groups: Control (CON), Laparotomy at 4 h and 24 h (LAP-4 h, LAP-24 h), then subjected to laparotomy under sevoflurane anaesthesia. The cognitive performance, peripheral and central inflammatory responses and tau phosphorylation levels were evaluated at 4 h and 24 h postoperatively. When LAP4-hrs displayed anxiety behaviors with high mRNA levels of inflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, IL-8, TNF-α and MCP-1 in the liver, and IL-8 in the hippocampus, results at 24 h were different. In the liver, only IL-10 protein was obviously elevated, but in the hippocampus, both pro- and anti-inflammatory cytokines were significantly decreased whilst the elimination of anxiety. The activity of major related kinases and phosphatases was remarkably changed which may contribute to the dephosphorylated tau protein. With tremendous neuropathological changes and significant numbers of activated microglias and astrocytes observed in the sub-regions of hippocampus, the memory impairment existed at both 4 h and 24 h. Since the association of dephosphorylated tau with POD, these findings may supply novel implications for the understanding of tauopathies and as a theoretical basis for preventions from the postoperative cognitive dysfunction (POCD).
Collapse
Affiliation(s)
- Le Yu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Guanghua Wen
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China
| | - Shoufeng Zhu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Xianwen Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Chunxia Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China. .,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China.
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.
| |
Collapse
|
33
|
Thapak P, Khare P, Bishnoi M, Sharma SS. Neuroprotective Effect of 2-Aminoethoxydiphenyl Borate (2-APB) in Amyloid β-Induced Memory Dysfunction: A Mechanistic Study. Cell Mol Neurobiol 2020; 42:1211-1223. [PMID: 33219878 DOI: 10.1007/s10571-020-01012-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
β-Amyloid (Aβ) peptide is a characteristic feature of Alzheimer's disease (AD) and accumulation of Aβ is associated with loss of synaptic plasticity and neuronal cell death. Aggregation of Aβ initiates numerous molecular signalling pathways leading to oxidative stress, mitochondrial dysfunction as well as an imbalance of calcium ion influx homeostasis. Recently, it has been shown that transient receptor potential melastatin 2 (TRPM2), a non-selective calcium-permeable cation channel has been postulated to play a vital role in the neuronal death, indicating the potential of TRPM2 inhibition in CNS disease. In this study, neuroprotective potential of 2-aminoethoxydiphenyl borate (2-APB), a broad-spectrum calcium channels blocker was investigated in Aβ-induced memory deficits in rats. In addition, effect of 2-APB on TRPM2 channels gene and protein expressions and also on calcium and memory related proteins was investigated in the hippocampus. Intracerebroventricular (I.C.V.) administration of Aβ (Aβ25-35, 10 μg) markedly induced cognitive impairment and upregulation of mRNA and protein expression of TRPM2 in the hippocampus. In addition, AChE activity was also increased in the cortex of the Aβ administered animals. Three-week treatment with 2-APB led to the down-regulation of TRPM2 mRNA and protein expression in the hippocampus and also improved the cognitive functions which was evident from the behavioral parameters. Moreover, 2-APB treatment also increased the calcium and memory associated proteins namely p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus and reduced the mRNA level of calcium buffering proteins and calcineurin A (PPP3CA) in the hippocampus. Furthermore, 2-APB treatment significantly reduced the AChE activity in the cortex. Thus, our findings suggest the neuroprotective effect of 2-APB in Aβ-induced cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
34
|
The Protective Effects of Ramelteon Against Isoflurane-Induced Insults and Inflammatory Response in Brain Microvascular Endothelial Cells. Neurotox Res 2020; 39:677-686. [PMID: 33211285 DOI: 10.1007/s12640-020-00309-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022]
Abstract
Anesthetic-induced cognitive impairment has been observed clinically. The mechanism underlying anesthetic-induced cognitive impairment is closely associated with neuronal apoptosis and neuroinflammation. Ramelteon is a potent and highly selective melatonin receptor agonist that has been used for the treatment of insomnia and has been reported to have an anti-inflammatory effect. In this study, we aimed to investigate the protective effects of Ramelteon against the cytotoxicity induced by isoflurane in brain microvascular endothelial cells. Our results show that Ramelteon ameliorated oxidative stress by suppressing the generation of mitochondrial reactive oxygen species (ROS) in human brain microvascular endothelial cells (HBMVECs). In addition, Ramelteon displayed a robust anti-inflammatory capacity against isoflurane-induced insults and inflammation by reducing the generation of interleukin-1β (IL-1β), transforming growth factor-β (TGF-β), monocyte chemotactic protein 1 (MCP-1), stromal cell-derived factor-1 (SDF-1), matrix metalloproteinase-2 (MMP-2), and MMP-9. Furthermore, Ramelteon reduced the expression of cell adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and E-selectin. Importantly, Ramelteon downregulated the activation of the p38MAPK/NF-κB signaling pathway, which is the key transcriptional regulator in the inflammation process. Our findings in the present study provide new evidence for the use of Ramelteon in the prevention of isoflurane-induced insults in brain endothelial cells.
Collapse
|
35
|
Zhao S, Fan Z, Hu J, Zhu Y, Lin C, Shen T, Li Z, Li K, Liu Z, Chen Y, Zhang B. The differential effects of isoflurane and sevoflurane on neonatal mice. Sci Rep 2020; 10:19345. [PMID: 33168900 PMCID: PMC7652873 DOI: 10.1038/s41598-020-76147-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Previous research has shown that exposure to volatile anesthetics can induce acute neuroinflammation and neuroapoptopsis in neonatal rodents and that these events can lead to cognitive dysfunction at later stages. Isoflurane and sevoflurane are two of the most popular anesthetics used in the field of pediatrics. However, the relative impact of these two anesthetics on the developing brain at distinct time points after the induction of anesthesia has not been compared. In the present study, we exposed 7-day-old mice to clinically equivalent doses of isoflurane (1.5%) and sevoflurane (2.5%) for 4 h and then investigated consequential changes in the brains of these mice at six different time points. We analyzed the levels of proteins that are directly related to neuroapoptosis, neuroinflammation, synaptic function, and memory, in the brains of neonatal mice. Exposure of neonatal mice to isoflurane and sevoflurane resulted in acute neuronal apoptosis. Our analysis observed significant levels of neuroinflammation and changes in the expression levels of proteins associated with both synaptic transmission and memory in mice from the isoflurane group but not the sevoflurane group. Our results therefore indicate that isoflurane and sevoflurane induce differential effects in the brains of neonatal mice.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Ziqi Fan
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jing Hu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yueli Zhu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Caixiu Lin
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Ting Shen
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zheyu Li
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Kaicheng Li
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhirong Liu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yanxing Chen
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.
| |
Collapse
|
36
|
Fox AP, Wagner KR, Towle VL, Xie KG, Xie Z. Caffeine reverses the unconsciousness produced by light anesthesia in the continued presence of isoflurane in rats. PLoS One 2020; 15:e0241818. [PMID: 33152041 PMCID: PMC7643991 DOI: 10.1371/journal.pone.0241818] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Currently no drugs are employed clinically to reverse the unconsciousness induced by general anesthetics. Our previous studies showed that caffeine, when given near the end of an anesthesia session, accelerated emergence from isoflurane anesthesia, likely caused by caffeine’s ability to elevate intracellular cAMP levels and to block adenosine receptors. These earlier studies showed that caffeine did not rouse either rats or humans from deep anesthesia (≥ 1 minimum alveolar concentration, MAC). In this current crossover study, we examined whether caffeine reversed the unconsciousness produced by light anesthesia (< 1 MAC) in the continued presence of isoflurane. The primary endpoint of this study was to measure isoflurane levels at the time of recovery of righting reflex, which was a proxy for consciousness. Rats were deeply anesthetized with 2% isoflurane (~1.5 MAC) for 20 minutes. Subsequently, isoflurane was reduced to 1.2% for 10 minutes, then by 0.2% every 10 min; animals were monitored until the recovery of righting reflex occurred, in the continued presence of isoflurane. Respiration rate, heart rate and electroencephalogram (EEG) were monitored. Our results show that caffeine-treated rats recovered their righting reflex at a significantly higher inspired isoflurane concentration, corresponding to light anesthesia, than the same rats treated with saline (control). Respiration rate and heart rate increased initially after caffeine injection but were then unchanged for the rest of the anesthesia session. Deep anesthesia is correlated with burst suppression in EEG recordings. Our data showed that caffeine transiently reduced the burst suppression time produced by deep anesthesia, suggesting that caffeine altered neuronal circuit function but not to a point where it caused arousal. In contrast, under light anesthesia, caffeine shifted the EEG power to high frequency beta and gamma bands. These data suggest that caffeine may represent a clinically viable drug to reverse the unconsciousness produced by light anesthesia.
Collapse
Affiliation(s)
- Aaron P. Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Kyle R. Wagner
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States of America
| | - Vernon L. Towle
- Department of Neurology, The University of Chicago, Chicago, Illinois, United States of America
| | - Kelvin G. Xie
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
37
|
Nie Y, Li S, Yan T, Ma Y, Ni C, Wang H, Zheng H. Propofol Attenuates Isoflurane-Induced Neurotoxicity and Cognitive Impairment in Fetal and Offspring Mice. Anesth Analg 2020; 131:1616-1625. [PMID: 33079886 DOI: 10.1213/ane.0000000000004955] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Anesthesia in pregnant rodents causes neurotoxicity in fetal and offspring rodents. However, the underlying mechanisms and targeted treatments remain largely to be determined. Isoflurane and propofol are among commonly used anesthetics. Thus, we set out to investigate whether propofol can mitigate the isoflurane-induced neurotoxicity in mice. METHODS Pregnant C57BL/6 mice at gestational day 15 (G15) were randomly assigned to 4 groups: control, isoflurane, propofol, and isoflurane plus propofol. Levels of interleukin (IL)-6 and poly-ADP ribose polymerase (PARP) fragment were measured in the brains of G15 embryos, and levels of postsynaptic density (PSD)-95 and synaptophysin were determined in the hippocampal tissues of postnatal day 31 (P31) offspring using Western blotting and immunohistochemical staining. Learning and memory functions in P31 offspring were determined using a Morris water maze test. RESULTS Isoflurane anesthesia in pregnant mice at G15 significantly increased brain IL-6 (222.6% ± 36.45% vs 100.5% ± 3.43%, P < .0001) and PARP fragment (384.2% ± 50.87% vs 99.59% ± 3.25%, P < .0001) levels in fetal mice and reduced brain PSD-95 (30.76% ± 2.03% vs 100.8% ± 2.25%, P < .0001) and synaptophysin levels in cornu ammonis (CA) 1 region (57.08% ± 4.90% vs 100.6% ± 2.20%, P < .0001) and dentate gyrus (DG; 56.47% ± 3.76% vs 99.76% ± 1.09%, P < .0001) in P31 offspring. Isoflurane anesthesia also impaired cognitive function in offspring at P31. Propofol significantly mitigated isoflurane-induced increases in brain IL-6 (117.5% ± 10.37% vs 222.6% ± 36.45%, P < .0001) and PARP fragment (205.1% ± 35.99% vs 384.2% ± 50.87%, P < .0001) levels in fetal mice, as well as reductions in PSD-95 (49.79% ± 3.43% vs 30.76% ± 2.03%, P < .0001) and synaptophysin levels in CA1 region (85.57% ± 2.97% vs 57.08% ± 4.90%, P < .0001) and DG (85.05% ± 1.87% vs 56.47% ± 3.76%, P < .0001) in hippocampus of P31 offspring. Finally, propofol attenuated isoflurane-induced cognitive impairment in offspring. CONCLUSIONS These findings suggest that gestational isoflurane exposure in mice induces neuroinflammation and apoptosis in embryos and causes cognitive impairment in offspring. Propofol can attenuate these isoflurane-induced detrimental effects.
Collapse
Affiliation(s)
- Yangyang Nie
- From the Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Shuai Li
- From the Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Yan
- From the Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Ni
- From the Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zheng
- From the Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Thapak P, Bishnoi M, Sharma SS. Pharmacological Inhibition of Transient Receptor Potential Melastatin 2 (TRPM2) Channels Attenuates Diabetes-induced Cognitive Deficits in Rats: A Mechanistic Study. Curr Neurovasc Res 2020; 17:249-258. [DOI: 10.2174/1567202617666200415142211] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 02/08/2023]
Abstract
Background:
Diabetes is a chronic metabolic disorder affecting the central nervous system.
A growing body of evidence has depicted that high glucose level leads to the activation of the
transient receptor potential melastatin 2 (TRPM2) channels. However, there are no studies targeting
TRPM2 channels in diabetes-induced cognitive decline using a pharmacological approach.
Objective:
The present study intended to investigate the effects of 2-aminoethoxydiphenyl borate
(2-APB), a TRPM2 inhibitor, in diabetes-induced cognitive impairment.
Methods:
Streptozotocin (STZ, 50 mg/kg, i.p.) was used to induce diabetes in rats. Animals were
randomly divided into the treatment group, model group and age-matched control and pre se
group. 2-APB treatment was given for three weeks to the animals. After 10 days of behavioural
treatment, parameters were performed. Animals were sacrificed at 10th week of diabetic induction
and the hippocampus and cortex were isolated. After that, protein and mRNA expression study
was performed in the hippocampus. Acetylcholinesterase (AchE) activity was done in the cortex.
Results: :
Our study showed the 10th week diabetic animals developed cognitive impairment, which
was evident from the behavioural parameters. Diabetic animals depicted an increase in the TRPM2
mRNA and protein expression in the hippocampus as well as increased AchE activity in the cortex.
However, memory associated proteins were down-regulated, namely Ca2+/calmodulin-dependent
protein kinase II (CaMKII-Thr286), glycogen synthase kinase 3 beta (GSK-3β-Ser9), cAMP
response element-binding protein (CREB-Ser133), and postsynaptic density protein 95 (PSD-95).
Gene expression of parvalbumin, calsequestrin and brain-derived neurotrophic factor (BDNF)
were down-regulated while mRNA level of calcineurin A/ protein phosphatase 3 catalytic subunit
alpha (PPP3CA) was upregulated in the hippocampus of diabetic animals. A three-week treatment
with 2-APB significantly ameliorated the alteration in behavioural cognitive parameters in diabetic
rats. Moreover, 2-APB also down-regulated the expression of TRPM2 mRNA and protein in the
hippocampus as well as AchE activity in the cortex of diabetic animals as compared to diabetic
animals. Moreover, the 2-APB treatment also upregulated the CaMKII (Thr-286), GSK-3β (Ser9),
CREB (Ser133), and PSD-95 expression and mRNA levels of parvalbumin, calsequestrin, and
BDNF while mRNA level of calcineurin A was down-regulated in the hippocampus of diabetic
animals.
Conclusion: :
This study confirms the ameliorative effect of TRPM2 channel inhibitor in the diabetes-
induced cognitive deficits. Inhibition of TRPM2 channels reduced the calcium associated
downstream signaling and showed a neuroprotective effect of TRPM2 channels in diabetesinduced
cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Punjab, India
| | - Shyam S. Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| |
Collapse
|
39
|
Jin X, Ji L, Chen Q, Sheng R, Ji F, Yang J. Anesthesia plus surgery in neonatal period impairs preference for social novelty in mice at the juvenile age. Biochem Biophys Res Commun 2020; 530:603-608. [PMID: 32747091 DOI: 10.1016/j.bbrc.2020.07.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
Anesthetic sevoflurane could induce neurotoxicity in developing brain and cause adverse neurobehavioral outcomes in mice, including inattention, social interaction deficit, and learning and memory impairment. However, there is less data on the effect of anesthesia plus surgery on social interaction behavior. Therefore, we investigated whether the combination of anesthesia and surgical stimulation could induce behavioral and biochemical changes in mice. Firstly, the six-day-old mice were received either 3% sevoflurane anesthesia or abdominal surgery under sevoflurane anesthesia. Then, these mice were scheduled to social interaction test in three-chambered social paradigm at one-month-old. In addition, the brain tissues of neonatal mice were harvested at 24 h after treatment, for measuring the levels of OXTR and NMDAR1 in Western blot analysis. We found that neonatal anesthesia with sevoflurane in a clinically-relevant dosage could not induce social interaction deficit. Nevertheless, anesthesia plus surgery was able to impair preference for social novelty in mice. Moreover, anesthesia plus surgery decreased the levels of OXTR in hippocampus and cortex of mice, as well as NMDAR1 in hippocampus. Collectively, these results suggested that anesthesia plus surgery could impair social novelty preference, but not sociability in mice, and that social memory might be more vulnerable than social affiliation in biological property. Furthermore, reduction in the levels of cortex OXTR and hippocampus NMDAR1 could be associated with social recognition memory in mice.
Collapse
Affiliation(s)
- Xin Jin
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Lei Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qingcai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Rui Sheng
- Department of Pharmacology, Soochow University School of Pharmaceutical Science, Suzhou, 215123, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianping Yang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
40
|
Kapoor MC. Depth of anesthesia monitoring in cardiac surgery-Standard of care soon? Ann Card Anaesth 2020; 23:260-261. [PMID: 32687079 PMCID: PMC7559963 DOI: 10.4103/aca.aca_141_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/29/2019] [Indexed: 11/04/2022] Open
Affiliation(s)
- Mukul Chandra Kapoor
- Department of Anesthesiology, Max Smart Super Specialty Hospital, Saket, New Delhi, India
| |
Collapse
|
41
|
Zhang Y, Zhao Y, Ran Y, Guo J, Cui H, Liu S. Notoginsenoside R1 attenuates sevoflurane-induced neurotoxicity. Transl Neurosci 2020; 11:215-226. [PMID: 33335762 PMCID: PMC7711878 DOI: 10.1515/tnsci-2020-0118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Sevoflurane, a volatile anesthetic, is known to induce widespread neuronal degeneration and apoptosis. Recently, the stress-inducible protein sestrin 2 and adenosine monophosphate-activated protein kinase (AMPK) have been found to regulate the levels of intracellular reactive oxygen species (ROS) and suppress oxidative stress. Notoginsenoside R1 (NGR1), a saponin isolated from Panax notoginseng, has been shown to exert neuroprotective effects. The effects of NGR1 against neurotoxicity induced by sevoflurane were assessed. Methods Sprague-Dawley rat pups on postnatal day 7 (PD7) were exposed to sevoflurane (3%) anesthesia for 6 h. NGR1 at doses of 12.5, 25, or 50 mg/kg body weight was orally administered to pups from PD2 to PD7. Results Pretreatment with NGR1 attenuated sevoflurane-induced generation of ROS and reduced apoptotic cell counts. Western blotting revealed decreased cleaved caspase 3 and Bad and Bax pro-apoptotic protein expression. NGR1 substantially upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) expression along with increased heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 levels, suggesting Nrf2 signaling activation. Enhanced sestrin-2 and phosphorylated AMPK expression were noticed following NGR1 pretreatment. Conclusion This study revealed the neuroprotective effects of NGR1 through effective suppression of apoptosis and ROS via regulation of apoptotic proteins and activation of Nrf2/HO-1 and sestrin 2/AMPK signaling cascades.
Collapse
Affiliation(s)
- Yibing Zhang
- Comprehensive Teaching and Research Office of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Yong Zhao
- GLP Laboratory, Institute of Chinese Materia Medica, China Academy of Traditional Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Yongwang Ran
- Department of Radiology, Qianjiang Central Hospital of Chongqing, Chongqing, 409099, People's Republic of China
| | - Jianyou Guo
- Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Haifeng Cui
- GLP Laboratory, Institute of Chinese Materia Medica, China Academy of Traditional Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Sha Liu
- Comprehensive Teaching and Research Office of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401331, People's Republic of China
| |
Collapse
|
42
|
Walker KA, Gottesman RF, Coresh J, Sharrett AR, Knopman DS, Mosley TH, Alonso A, Zhou Y, Wong DF, Brown CH. Association of Surgical Hospitalization with Brain Amyloid Deposition: The Atherosclerosis Risk in Communities-Positron Emission Tomography (ARIC-PET) Study. Anesthesiology 2020; 132:1407-1418. [PMID: 32412719 PMCID: PMC7540736 DOI: 10.1097/aln.0000000000003255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND As more older adults undergo surgery, it is critical to understand the long-term effects of surgery on brain health, particularly in relation to the development of Alzheimer's disease. This study examined the association of surgical hospitalization with subsequent brain β-amyloid deposition in nondemented older adults. METHODS The Atherosclerosis Risk in Communities-Positron Emission Tomography (ARIC-PET) study is a prospective cohort study of 346 participants without dementia who underwent florbetapir PET imaging. Active surveillance of local hospitals and annual participant contact were used to gather hospitalization and surgical information (International Classification of Disease, Ninth Revision, Clinical Modification codes) over the preceding 24-yr period. Brain amyloid measured using florbetapir PET imaging was the primary outcome. Elevated amyloid was defined as a standardized uptake value ratio of more than 1.2. RESULTS Of the 313 participants included in this analysis (age at PET: 76.0 [SD 5.4]; 56% female), 72% had a prior hospitalization, and 50% had a prior surgical hospitalization. Elevated amyloid occurred in 87 of 156 (56%) participants with previous surgical hospitalization, compared with 45 of 87 (52%) participants who had no previous hospitalization. Participants with previous surgical hospitalizations did not show an increased odds of elevated brain amyloid (odds ratio, 1.32; 95% CI, 0.72 to 2.40; P = 0.370) after adjusting for confounders (primary analysis). Results were similar using the reference group of all participants without previous surgery (hospitalized and nonhospitalized; odds ratio, 1.58; 95% CI, 0.96 to 2.58; P = 0.070). In a prespecified secondary analysis, participants with previous surgical hospitalization did demonstrate increased odds of elevated amyloid when compared with participants hospitalized without surgery (odds ratio, 2.10; 95% CI, 1.09 to 4.05; P = 0.026). However, these results were attenuated and nonsignificant when alternative thresholds for amyloid-positive status were used. CONCLUSIONS The results do not support an association between surgical hospitalization and elevated brain amyloid.
Collapse
Affiliation(s)
- Keenan A Walker
- From the Departments of Neurology (K.A.W., R.F.G.) Radiology (D.F.W.) Anesthesiology (C.H.B.), Johns Hopkins School of Medicine, Baltimore, Maryland the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (R.F.G., J.C., A.R.S.) the Department of Neurology, Mayo Clinic, Rochester, Minnesota (D.S.K.) Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, Mississippi (T.H.M.) the Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia (A.A.) the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri (Y.Z.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Liu M, Li M, Zhou Y, Zhou Q, Jiang Y. HSP90 inhibitor 17AAG attenuates sevoflurane-induced neurotoxicity in rats and human neuroglioma cells via induction of HSP70. J Transl Med 2020; 18:166. [PMID: 32293462 PMCID: PMC7158111 DOI: 10.1186/s12967-020-02332-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/06/2020] [Indexed: 12/23/2022] Open
Abstract
Background 17AAG has been extensively studied for its antitumor effects that protect cells from lethal stress by maintaining protein stability. The role of 17AAG in sevoflurane-induced neuronal injury has never been studied. We aim to investigate the effect of 17AAG on sevoflurane-induced neurotoxicity in vivo and in vitro. Methods Sevoflurane-induced hippocampal neuron injury model was established in aged Sprague–Dawley rats. Pretreatment of vehicle or 17AAG was administered prior to sevoflurane inhalation. H4 neuroglioma cells were pretreated with vehicle or 17AAG and exposed to sevoflurane. Apoptosis, oxidative stress, expression of interleukin-6 (IL-6), and activation of the nuclear factor-κB (NF-κB) signaling pathway in H4 cells were examined by Hoechst assay, flow cytometry, Western blot, and immunofluorescent staining. RNA interference against HSPA1A was performed to test the function of HSP70 in neuroprotection. Results Exogenous 17AAG reduced sevoflurane-induced apoptosis and oxidative stress in rat hippocampal neurons and in H4 cells. In H4 cells, 17AAG suppressed sevoflurane-induced upregulation of IL-6 and activation of NF-κB signaling. 17AAG enhanced sevoflurane-induced upregulation of HSP70 in rat hippocampal neurons and in H4 cells. Conversely, silencing of HSPA1A in H4 cells blocked the cytoprotective effect of 17AAG against sevoflurane-induced apoptosis and oxidative stress, and prevented upregulation of IL-6 and activation of NF-κB signaling. Conclusions 17AAG protects against sevoflurane-induced neurotoxicity in vivo and in vitro via HSP70-dependent inhibition of apoptosis, oxidative stress, and pro-inflammatory signaling pathway.
Collapse
Affiliation(s)
- Min Liu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139, Changsha City, 410000, Hunan Province, People's Republic of China
| | - Moyun Li
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha City, 410000, Hunan Province, People's Republic of China
| | - Yu Zhou
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139, Changsha City, 410000, Hunan Province, People's Republic of China
| | - Qian Zhou
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139, Changsha City, 410000, Hunan Province, People's Republic of China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139, Changsha City, 410000, Hunan Province, People's Republic of China.
| |
Collapse
|
44
|
Jia J, Zhu J, Yang Q, Wang Y, Zhang Z, Chen C. The Role of Histone Acetylation in the Sevoflurane-induced Inhibition of Neurogenesis in the Hippocampi of Young Mice. Neuroscience 2020; 432:73-83. [DOI: 10.1016/j.neuroscience.2020.02.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022]
|
45
|
Hypoxia, hypercarbia, and mortality reporting in studies of anaesthesia-related neonatal neurodevelopmental delay in rodent models. Eur J Anaesthesiol 2020; 37:70-84. [DOI: 10.1097/eja.0000000000001105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
46
|
Zhu J, Zhang Z, Jia J, Wang L, Yang Q, Wang Y, Chen C. Sevoflurane Induces Learning and Memory Impairment in Young Mice Through a Reduction in Neuronal Glucose Transporter 3. Cell Mol Neurobiol 2019; 40:879-895. [PMID: 31884568 PMCID: PMC7295720 DOI: 10.1007/s10571-019-00779-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Sevoflurane, which is widely used in paediatric anaesthesia, induces neural apoptosis in the developing brain and cognitive impairment in young mammals. Glucose hypometabolism is the key pathophysiological modulator of cognitive dysfunction. However, the effects and mechanism of sevoflurane on cerebral glucose metabolism after its use as an anaesthetic and its complete elimination are still unknown. We therefore investigated the influence of sevoflurane on neuronal glucose transporter isoform 3 (GLUT3) expression, glucose metabolism and apoptosis in vivo and in vitro and on neurocognitive function in young mice 24 h after the third exposure to sevoflurane. Postnatal day 14 (P14) mice and neural cells were exposed to 3% sevoflurane 2 h daily for three days. We found that sevoflurane anaesthesia decreased GLUT3 gene and protein expression in the hippocampus and temporal lobe, consistent with a decrease in glucose metabolism in the hippocampus and temporal lobe observed by [18F] fluorodeoxyglucose positron emission tomography (18F-FDG PET). Moreover, sevoflurane anaesthesia increased the number of TUNEL-positive cells and the levels of Bax, cleaved caspase 3 and cleaved PARP and reduced Bcl-2 levels in the hippocampus and temporal lobe. Young mice exposed to sevoflurane multiple times also showed learning and memory impairment. In addition, sevoflurane inhibited GLUT3 expression in primary hippocampal neurons and PC12 cells. GLUT3 overexpression in cultured neurons ameliorated the sevoflurane-induced decrease in glucose utilization and increase in the apoptosis rate. These data indicate that GLUT3 deficiency may contribute to sevoflurane-induced learning and memory deficits in young mice.
Collapse
Affiliation(s)
- Jinpiao Zhu
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Zongze Zhang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Junke Jia
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Lirong Wang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Qiuyue Yang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yanlin Wang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Chang Chen
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
47
|
Song SY, Meng XW, Xia Z, Liu H, Zhang J, Chen QC, Liu HY, Ji FH, Peng K. Cognitive impairment and transcriptomic profile in hippocampus of young mice after multiple neonatal exposures to sevoflurane. Aging (Albany NY) 2019; 11:8386-8417. [PMID: 31582589 PMCID: PMC6814607 DOI: 10.18632/aging.102326] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023]
Abstract
Children with repeated inhalational anesthesia may develop cognitive disorders. This study aimed to investigate the transcriptome-wide response of hippocampus in young mice that had been exposed to multiple sevoflurane in the neonatal period. Mice received 3% sevoflurane for 2 h on postnatal day (PND) 6, 8, and 10, followed by arterial blood gas test on PND 10, behavioral experiments on PND 31–36, and RNA sequencing (RNA-seq) of hippocampus on PND 37. Functional annotation and protein-protein interaction analyses of differentially expressed genes (DEGs) and quantitative reverse transcription polymerase chain reaction (qPCR) were performed. Neonatal sevoflurane exposures induced cognitive and social behavior disorders in young mice. RNA-seq identified a total of 314 DEGs. Several enriched biological processes (ion channels, brain development, learning, and memory) and signaling pathways (oxytocin signaling pathway and glutamatergic, cholinergic, and GABAergic synapses) were highlighted. As hub-proteins, Pten was involved in nervous system development, synapse assembly, learning, memory, and behaviors, Nos3 and Pik3cd in oxytocin signaling pathway, and Cdk16 in exocytosis and phosphorylation. Some top DEGs were validated by qPCR. This study revealed a transcriptome-wide profile in mice hippocampus after multiple neonatal exposures to sevoflurane, promoting better understanding of underlying mechanisms and investigation of preventive strategies.
Collapse
Affiliation(s)
- Shao-Yong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - ZhengYuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA 95817, USA
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA 95817, USA
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qing-Cai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
48
|
Liang R, Ou S, Han Y, Xu J, Zhou S. Plasma amyloid beta level changes in aged mice with cognitive dysfunction following sevoflurane exposure. Exp Gerontol 2019; 129:110737. [PMID: 31521721 DOI: 10.1016/j.exger.2019.110737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/05/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Previous studies have stated that cognitive impairment induced by anesthetics was associated with amyloid beta (Aβ). However, few researchers have investigated the transport of Aβ inside and outside of the brain. AIM We attempted to probe the effects of sevoflurane on cognitive functions, the plasma Aβ, and transporters of Aβ in aged mice. The receptor for advanced glycation end-products (RAGE) is an Aβ influx protein, and Low-density lipoprotein receptor-related protein-1 (LRP-1) is an Aβ efflux protein. METHODS Aged mice were divided into the control group and the sevoflurane group. The mice were exposed to 100% oxygen or 2.5% sevoflurane for 2 h. The abilities of spatial learning and memory in mice were tested using the Morris water maze. Aβ concentrations of plasma were measured with enzyme-linked immunosorbent assay kits. The RAGE and LRP-1 gene levels in the brain were assessed with quantitative polymerase chain reaction, and the protein levels were determined by western blot analysis. The locations of RAGE in the brain were confirmed via immunofluorescence. RESULTS In the sevoflurane group mice, the escape latency was increased on the 5th day of training, and the time spent in the target quadrant was decreased on the 7th day after anesthesia. Sevoflurane reduced the concentration of plasma Aβ1-40. In addition, sevoflurane increased both gene and protein levels of RAGE in the brain, and increased RAGE proteins co-localized with the hippocampal vascular endothelial cells. CONCLUSION RAGE over-expression in the hippocampal vascular endothelial cells possibly resulted in the excessive transport of the plasma Aβ1-40 into the brain after treatment with sevoflurane, which was associated with sevoflurane-induced cognitive dysfunction in aged mice.
Collapse
Affiliation(s)
- Rui Liang
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Shanshan Ou
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Yuxiang Han
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Shaopeng Zhou
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China.
| |
Collapse
|
49
|
Iqbal F, Thompson AJ, Riaz S, Pehar M, Rice T, Syed NI. Anesthetics: from modes of action to unconsciousness and neurotoxicity. J Neurophysiol 2019; 122:760-787. [PMID: 31242059 DOI: 10.1152/jn.00210.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern anesthetic compounds and advanced monitoring tools have revolutionized the field of medicine, allowing for complex surgical procedures to occur safely and effectively. Faster induction times and quicker recovery periods of current anesthetic agents have also helped reduce health care costs significantly. Moreover, extensive research has allowed for a better understanding of anesthetic modes of action, thus facilitating the development of more effective and safer compounds. Notwithstanding the realization that anesthetics are a prerequisite to all surgical procedures, evidence is emerging to support the notion that exposure of the developing brain to certain anesthetics may impact future brain development and function. Whereas the data in support of this postulate from human studies is equivocal, the vast majority of animal research strongly suggests that anesthetics are indeed cytotoxic at multiple brain structure and function levels. In this review, we first highlight various modes of anesthetic action and then debate the evidence of harm from both basic science and clinical studies perspectives. We present evidence from animal and human studies vis-à-vis the possible detrimental effects of anesthetic agents on both the young developing and the elderly aging brain while discussing potential ways to mitigate these effects. We hope that this review will, on the one hand, invoke debate vis-à-vis the evidence of anesthetic harm in young children and the elderly, and on the other hand, incentivize the search for better and less toxic anesthetic compounds.
Collapse
Affiliation(s)
- Fahad Iqbal
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Thompson
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Saba Riaz
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Pehar
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Rice
- Department of Anesthesiology, Perioperative and Pain Medicine, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
50
|
Ton HT, Yang L, Xie Z. Sevoflurane increases locomotion activity in mice. PLoS One 2019; 14:e0206649. [PMID: 31112538 PMCID: PMC6528997 DOI: 10.1371/journal.pone.0206649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Clinical observations show emergence of agitation and hyperactivity during the anesthesia induction and/or recovery period post-anesthesia. However, an animal model to illustrate this clinical phenomenon has not yet been established. We therefore set out to investigate whether sevoflurane, a commonly used anesthetic, could alter locomotion in mice during the anesthesia induction and recovery period post-anesthesia. The activity of the mice was recorded 5 minutes before, during (for 30 minutes), and 40 minutes after the administration of the anesthetic sevoflurane [1-, 1.5- and 2-fold minimum alveolar concentration] at 370 C. The total walking distance and velocity of movement were measured and quantified as the indexes of locomotion. We found that the anesthetic sevoflurane increased the locomotion of the mice during the induction period of the anesthesia. During the recovery phase after anesthesia, the mice exhibited increased locomotion for a short period of time (about 5 minutes) and then displayed a sharp decrease in mobility for up to 60 minutes following the end of anesthesia administration. The anesthetic sevoflurane did not significantly alter the food intake and body weight of the mice. Furthermore, we found that Alzheimer’s disease transgenic mice exhibited a greater degree of sevoflurane-induced hyperactivity than the wild-type mice did. Our results showed that inhalation of the anesthetic sevoflurane induced an acute hyperactivity in mice, particularly among Alzheimer’s disease transgenic mice. These findings from the pilot studies have established an animal model to promote further studies into postoperative emergence agitation, hyperactivity and the underlying mechanisms into these conditions.
Collapse
Affiliation(s)
- Hoai T. Ton
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States of America
- Department of Biology, Vinh University, Vinh City, Nghe An, Vietnam
| | - Lei Yang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States of America
- Department of Anesthesia, Ruijing Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States of America
- * E-mail:
| |
Collapse
|