1
|
Du W, Zhang X, Li S, Xie X. Novel Perspective on Sevoflurane-Induced Cognitive Dysfunction: Implications of Neuronal SIRPα and Microglial Synaptic Remodeling. ACS Chem Neurosci 2024; 15:4500-4516. [PMID: 39644326 DOI: 10.1021/acschemneuro.4c00485] [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: 12/09/2024] Open
Abstract
This study aims to investigate the role of neuronal SIRPα and microglial synaptic remodeling in sevoflurane-induced cognitive dysfunction in newborn mice. Newborn mice were exposed to sevoflurane, followed by behavioral assessments and single-cell transcriptome sequencing of cortical cells. Lentivirus-mediated overexpression of neuronal SIRPα and assessment of the microglial morphology and synaptic function were conducted. Sevoflurane exposure resulted in social cognitive impairments without affecting motor coordination. Transcriptomic analysis revealed no significant changes in cortical microglial cells or neurons. However, sevoflurane inhibited nonsynaptic synapse modification by microglia. Overexpression of neuronal SIRPα enhanced microglial function, promoted neuron development, and ameliorated cognitive impairments. SCENIC analysis identified a correlation between IRF8 and SIRPα expression. This study sheds light on the involvement of neuronal SIRPα and microglial synaptic remodeling in sevoflurane-induced cognitive dysfunction. Understanding these mechanisms offers new avenues for exploring cognitive impairment pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Wei Du
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Xiaomin Zhang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Songze Li
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Xin Xie
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| |
Collapse
|
2
|
Han S, Bian R, Chen Y, Liang J, Zhao P, Gu Y, Zhang D. Dysregulation of the Gut Microbiota Contributes to Sevoflurane-Induced Cognitive Dysfunction in Aged Mice by Activating the NLRP3 Inflammasome. Mol Neurobiol 2024; 61:10500-10516. [PMID: 38740706 DOI: 10.1007/s12035-024-04229-x] [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: 01/05/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Postoperative cognitive dysfunction (POCD), a common complication in elderly patients after surgery, seriously affects patients' quality of life. Long-term or repeated inhalation of sevoflurane can cause neuroinflammation, which is a risk factor for POCD. However, the underlying mechanism needs to be further explored. Recent research had revealed a correlation between neurological disorders and changes in the gut microbiota. Dysfunction of the gut microbiota is involved in the occurrence and development of central nervous system diseases. Here, we found that cognitive dysfunction in aged mice induced by sevoflurane exposure (3%, 2 hours daily, for 3 days) was related to gut microbiota dysbiosis, while probiotics improved cognitive function by alleviating dysbiosis. Sevoflurane caused a significant decrease in the abundance of Akkermansia (P<0.05), while probiotics restored the abundance of Akkermansia. Compared to those in the control group, sevoflurane significantly increased the expression of NLRP3 inflammasome-associated proteins in the gut and brain in the sevoflurane-exposed group, thus causing neuroinflammation and synaptic damage, which probiotics can mitigate (con vs. sev, P < 0.01; p+sev vs. sev, P < 0.05). In conclusion, for the first time, our study revealed that dysbiosis of the gut microbiota caused by sevoflurane anesthesia contributes to the NLRP3 inflammasome-mediated neuroinflammation and cognitive dysfunction from the perspective of the gut-brain axis. Perhaps postoperative cognitive impairment in elderly patients can be alleviated or even prevented by regulating the gut microbiota. This study provides new insights and methods for the prevention and treatment of cognitive impairment induced by sevoflurane.
Collapse
Affiliation(s)
- Shanshan Han
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi, 214002, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Ruxi Bian
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yuxuan Chen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Junjie Liang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yanfang Gu
- Department of Gynecology, Jiangnan University Affiliated Hospital, Wuxi, 214002, China.
| | - Dengxin Zhang
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi, 214002, China.
| |
Collapse
|
3
|
Zhang Q, Li Y, Zhang J, Cui Y, Sun S, Chen W, Shi L, Zhang Y, Hou Z. IL-17A is a key regulator of neuroinflammation and neurodevelopment in cognitive impairment induced by sevoflurane. Free Radic Biol Med 2024; 227:12-26. [PMID: 39581388 DOI: 10.1016/j.freeradbiomed.2024.11.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Increasing numbers of animal studies have shown that repeat sevoflurane exposure during developmental stage may lead to long-term cognitive impairment. Nevertheless, the exact pathogenesis remains unclear. Interleukin 17A (IL-17A) has been associated with cognitive decline in various neurological disorders. Here we found that the expression of IL-17A was up-regulated in hippocampus of sevoflurane exposed neonatal mice. Genetic deletion of IL-17A or inhibition of IL-17A improved behavioral function and down-regulated neuroinflammation related genes, interleukin 1β (IL-1β), interleukin 6 (IL-6), Nicotinamide adenine dinucleotide phosphate(NADPH) oxidase 2 (NOX2) and NADPH oxidase 4 (NOX4) in hippocampus of sevoflurane exposed neonatal mice. Moreover, negative regulation of IL-17A/Interleukin 17A receptor(IL-17RA) promoted the extracellular signal-regulated protein kinase (ERK) signaling pathway and nucleation of cyclic adenosine monophosphate (cAMP) response element-binding (CREB) in neurons of cognitive impaired mice. Knockdown of IL-17A in vivo identified neurons-localized IL-17A as a major factor in neuroinflammation and neurodevelopment. Collectively, our results suggested that IL-17A was required for the pathogenesis of neuroinflammatory response and identify IL-17A as a potential therapeutic target for cognitive impairment exposed by general anesthetics during infancy.
Collapse
Affiliation(s)
- Qi Zhang
- Postdoctoral Mobile Station of the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, PR China; Department of Anesthesiology, Hebei Children's Hospital Affiliated to Hebei Medical University, Hebei, 050031, PR China; Key Laboratory of Pediatric Epilepsy and Neurological Disorders of Hebei Province, PR China
| | - Yanan Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Hebei, PR China
| | - Jiajie Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunyi Cui
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Suzhen Sun
- Key Laboratory of Pediatric Epilepsy and Neurological Disorders of Hebei Province, PR China; Department of Neurology, Hebei Children's Hospital Affiliated to Hebei Medical University, Hebei, 050031, PR China
| | - Wei Chen
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Lei Shi
- Department of Anesthesiology, Hebei Children's Hospital Affiliated to Hebei Medical University, Hebei, 050031, PR China.
| | - Yingze Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China; Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, PR China; NHC Key Laboratory of Intelligent Orthopaedic Equipment (the Third Hospital of Hebei Medical University), Hebei, PR China.
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China; Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, PR China; NHC Key Laboratory of Intelligent Orthopaedic Equipment (the Third Hospital of Hebei Medical University), Hebei, PR China.
| |
Collapse
|
4
|
Zhang Z, Yang W, Wang L, Zhu C, Cui S, Wang T, Gu X, Liu Y, Qiu P. Unraveling the role and mechanism of mitochondria in postoperative cognitive dysfunction: a narrative review. J Neuroinflammation 2024; 21:293. [PMID: 39533332 PMCID: PMC11559051 DOI: 10.1186/s12974-024-03285-3] [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: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a frequent neurological complication encountered during the perioperative period with unclear mechanisms and no effective treatments. Recent research into the pathogenesis of POCD has primarily focused on neuroinflammation, oxidative stress, changes in neural synaptic plasticity and neurotransmitter imbalances. Given the high-energy metabolism of neurons and their critical dependency on mitochondria, mitochondrial dysfunction directly affects neuronal function. Additionally, as the primary organelles generating reactive oxygen species, mitochondria are closely linked to the pathological processes of neuroinflammation. Surgery and anesthesia can induce mitochondrial dysfunction, increase mitochondrial oxidative stress, and disrupt mitochondrial quality-control mechanisms via various pathways, hence serving as key initiators of the POCD pathological process. We conducted a review on the role and potential mechanisms of mitochondria in postoperative cognitive dysfunction by consulting relevant literature from the PubMed and EMBASE databases spanning the past 25 years. Our findings indicate that surgery and anesthesia can inhibit mitochondrial respiration, thereby reducing ATP production, decreasing mitochondrial membrane potential, promoting mitochondrial fission, inducing mitochondrial calcium buffering abnormalities and iron accumulation, inhibiting mitophagy, and increasing mitochondrial oxidative stress. Mitochondrial dysfunction and damage can ultimately lead to impaired neuronal function, abnormal synaptic transmission, impaired synthesis and release of neurotransmitters, and even neuronal death, resulting in cognitive dysfunction. Targeted mitochondrial therapies have shown positive outcomes, holding promise as a novel treatment for POCD.
Collapse
Affiliation(s)
- Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Wei Yang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Lanbo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chengyao Zhu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Shuyan Cui
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Tian Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
5
|
Meng X, Wang Y, Zhao W, Chen Y, Li W, Peng K, Xu H, Yang Y, Shan X, Huo W, Liu H, Ji F. Identification of differential m6A RNA methylomes and ALKBH5 as a potential prevention target in the developmental neurotoxicity induced by multiple sevoflurane exposures. FASEB J 2024; 38:e23793. [PMID: 39003634 DOI: 10.1096/fj.202400664r] [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: 03/26/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Sevoflurane, as a commonly used inhaled anesthetic for pediatric patients, has been reported that multiple sevoflurane exposures are associated with a greater risk of developing neurocognitive disorder. N6-Methyladenosine (m6A), as the most common mRNA modification in eukaryotes, has emerged as a crucial regulator of brain function in processes involving synaptic plasticity, learning and memory, and neurodevelopment. Nevertheless, the relevance of m6A RNA methylation in the multiple sevoflurane exposure-induced developmental neurotoxicity remains mostly elusive. Herein, we evaluated the genome-wide m6A RNA modification and gene expression in hippocampus of mice that received with multiple sevoflurane exposures using m6A-sequencing (m6A-seq) and RNA-sequencing (RNA-seq). We discovered 19 genes with differences in the m6A methylated modification and differential expression in the hippocampus. Among these genes, we determined that a total of nine differential expressed genes may be closely associated with the occurrence of developmental neurotoxicity induced by multiple sevoflurane exposures. We further found that the alkB homolog 5 (ALKBH5), but not methyltransferase-like 3 (METTL3) and Wilms tumor 1-associated protein (WTAP), were increased in the hippocampus of mice that received with multiple sevoflurane exposures. And the IOX1, as an inhibitor of ALKBH5, significantly improved the learning and memory defects and reduced neuronal damage in the hippocampus of mice induced by multiple sevoflurane exposures. The current study revealed the role of m6A methylated modification and m6A-related regulators in sevoflurane-induced cognitive impairment, which might provide a novel insight into identifying biomarkers and therapeutic strategies for inhaled anesthetic-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Xiaowen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Yichan Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Weiming Zhao
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Ying Chen
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wenting Li
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Hanbing Xu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Yufan Yang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Xisheng Shan
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Wenwen Huo
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Huayue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
- Ambulatory Surgery Center, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
6
|
Xie A, Zhang X, Ju F, Zhou Y, Wu D, Han J. Sevoflurane impedes neuropathic pain by maintaining endoplasmic reticulum stress and oxidative stress homeostasis through inhibiting the activation of the PLCγ/CaMKII/IP3R signaling pathway. Aging (Albany NY) 2024; 16:11062-11071. [PMID: 38975935 PMCID: PMC11272110 DOI: 10.18632/aging.206001] [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: 06/13/2023] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE To investigate the effect of sevoflurane on neuropathic pain induced by chronic constriction injury (CCI) of sciatic nerve in mice, and to elucidate its mechanism by animal experiments. METHODS AND RESULTS Thirty-two C57BL/6 mice were randomly divided into four groups: Sham group, Model group, Control group and Sevoflurane group. First, a mouse model of neuropathic pain was established. Then, the mice in each group were killed on Day 14 after operation to harvest the enlarged lumbosacral spinal cord. In contrast with the Model group, the Sevoflurane group displayed a significantly increased paw withdrawal mechanical threshold (PWMT) and significantly prolonged paw withdrawal thermal latency (PWTL) from Day 5 after operation. The morphological changes of lumbosacral spinal cord were observed by hematoxylin-eosin (HE) staining and transmission electron microscopy. Pathological results showed that sevoflurane reduced nuclear pyknosis in lumbosacral spinal cord tissue, with a large number of mitochondrial crista disappearance and mitochondrial swelling. The results of Western blotting showed that sevoflurane significantly decreased the protein expressions of phosphorylated phospholipase Cγ (p-PLCγ), phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) and phosphorylated inositol 1,4,5-triphosphate receptor (p-IP3R), and reduced the protein expressions of endoplasmic reticulum (ER) stress proteins glucose-regulated protein 78 (GRP78) and GRP94, oxidative stress-related proteins P22 and P47 and inflammatory factors nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), interleukin-1 β (IL-1β), and tumor necrosis factor-α (TNF-α). CONCLUSIONS Sevoflurane inhibits neuropathic pain by maintaining ER stress and oxidative stress homeostasis through inhibiting the activation of the PLCγ/CaMKII/IP3R signaling pathway.
Collapse
Affiliation(s)
- An Xie
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Xianjie Zhang
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Yukai Zhou
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Dan Wu
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Jia Han
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| |
Collapse
|
7
|
Zhu R, Liu L, Mao T, Wang X, Li Y, Li T, Lv S, Zeng S, Fu N, Li N, Wang Y, Sun M, Zhang J. Mfn2 regulates mitochondria and mitochondria-associated endoplasmic reticulum membrane function in neurodegeneration induced by repeated sevoflurane exposure. Exp Neurol 2024; 377:114807. [PMID: 38704082 DOI: 10.1016/j.expneurol.2024.114807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Repeated sevoflurane exposure in neonatal mice can leads to neuronal apoptosis and mitochondrial dysfunction. The mitochondria are responsible for energy production to maintain homeostasis in the central nervous system. The mitochondria-associated endoplasmic reticulum membrane (MAM) is located between the mitochondria and endoplasmic reticulum (ER), and it is critical for mitochondrial function and cell survival. MAM malfunction contributes to neurodegeneration, however, whether it is involved in sevoflurane-induced neurotoxicity remains unknown. Our study demonstrated that repeated sevoflurane exposure induced mitochondrial dysfunction and dampened the MAM structure. The upregulated ER-mitochondria tethering enhanced Ca2+ transition from the cytosol to the mitochondria. Overload of mitochondrial Ca2+ contributed to opening of the mitochondrial permeability transition pore (mPTP), which caused neuronal apoptosis. Mitofusin 2(Mfn2), a key regulator of ER-mitochondria contacts, was found to be suppressed after repeated sevoflurane exposure, while restoration of Mfn2 expression alleviated cognitive dysfunction due to repeated sevoflurane exposure in the adult mice. These evidences suggest that sevoflurane-induced MAM malfunction is vulnerable to Mfn2 suppression, and the enhanced ER-mitochondria contacts promotes mitochondrial Ca2+ overload, contributing to mPTP opening and neuronal apoptosis. This paper sheds light on a novel mechanism of sevoflurane-induced neurotoxicity. Furthermore, targeting Mfn2-mediated regulation of the MAM structure and mitochondrial function may provide a therapeutic advantage in sevoflurane-induced neurodegeneration.
Collapse
Affiliation(s)
- Ruilou Zhu
- 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, PR China 450003
| | - Lu Liu
- 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, PR China 450003; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, PR China 450001
| | - Tian Mao
- 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, PR China 450003; School of Clinical Medicine, Henan University, Kaifeng, Henan, PR China, 475004
| | - Xiaoling Wang
- 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, PR China 450003; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, PR China 450001
| | - Yubao Li
- 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, PR China 450003; Department of Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, PR China, 453003
| | - Ting Li
- 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, PR China 450003; Department of Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, PR China, 453003
| | - Shuang Lv
- 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, PR China 450003
| | - Shuang Zeng
- 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, PR China 450003
| | - Ningning Fu
- 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, PR China 450003
| | - Ningning Li
- 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, PR China 450003
| | - Yangyang Wang
- 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, PR China 450003
| | - Mingyang Sun
- 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, PR China 450003
| | - 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, PR China 450003.
| |
Collapse
|
8
|
Yang J, Zhi W, Wang L. Role of Tau Protein in Neurodegenerative Diseases and Development of Its Targeted Drugs: A Literature Review. Molecules 2024; 29:2812. [PMID: 38930877 PMCID: PMC11206543 DOI: 10.3390/molecules29122812] [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: 04/26/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Tau protein is a microtubule-associated protein that is widely distributed in the central nervous system and maintains and regulates neuronal morphology and function. Tau protein aggregates abnormally and forms neurofibrillary tangles in neurodegenerative diseases, disrupting the structure and function of neurons and leading to neuronal death, which triggers the initiation and progression of neurological disorders. The aggregation of tau protein in neurodegenerative diseases is associated with post-translational modifications, which may affect the hydrophilicity, spatial conformation, and stability of tau protein, promoting tau protein aggregation and the formation of neurofibrillary tangles. Therefore, studying the role of tau protein in neurodegenerative diseases and the mechanism of aberrant aggregation is important for understanding the mechanism of neurodegenerative diseases and finding therapeutic approaches. This review describes the possible mechanisms by which tau protein promotes neurodegenerative diseases, the post-translational modifications of tau protein and associated influencing factors, and the current status of drug discovery and development related to tau protein, which may contribute to the development of new therapeutic approaches to alleviate or treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiakai Yang
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Weijia Zhi
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Lifeng Wang
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
9
|
Cai J, Lin Y, Zhou B, Xiao F, Xu G, Lu J. SHARPIN contributes to sevoflurane-induced neonatal neurotoxicity through up-regulating HMGB1 to repress M2 like-macrophage polarization. Metab Brain Dis 2024; 39:841-853. [PMID: 38805141 DOI: 10.1007/s11011-024-01355-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Sevoflurane exposure can result in neurotoxicity especially among children, which remains an important complication after surgery. However, its related mechanisms remain unclear. Here, we investigated the biological roles of SHARPIN in sevoflurane-induced neurotoxicity. As detected by qPCR, Western blotting and immunohistochemical staining, SHARPIN and HMGB1 expression was elevated in sevoflurane-stimulated mice as compared with the control mice. SHARPIN depletion attenuated hippocampus injury, repressed the expression of HMGB1 and M1-like macrophage markers (iNOS, TNF-α, IL-1β, IL-6), but enhanced the expression of M2-like macrophage markers (ARG-1, IL-10). GST pull-down and Co-IP assays demonstrated that SHARPIN directly interacted with HMGB1 to enhance HMGB1 expression in SH-SY5Y cells. The inhibitory effects of SHARPIN silencing on inflammatory reaction and M1-like macrophages were counteracted by HMGB1 overexpression. Finally, SHARPIN-HMGB1 pathway affected neuroinflammation triggered by sevoflurane via modulating macrophage polarization. Collectively, our data suggested that SHARPIN stimulated sevoflurane-induced neurotoxicity via converting M2-like macrophages to M1-like macrophages by enhancing HMGB1 expression. SHARPIN intervention may be a promising therapeutic method to relieve sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Junying Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jun Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
10
|
Gao J, Zhang H, Zhou L, Liu J, Zhuo E, Shen Y, Liu X, Shen Q. MANF Alleviates Sevoflurane-Induced Cognitive Impairment in Neonatal Mice by Modulating Microglial Activation and Polarization. Mol Neurobiol 2024; 61:3357-3368. [PMID: 37989984 DOI: 10.1007/s12035-023-03792-z] [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: 08/11/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
The precise mechanism underlying sevoflurane-induced neurotoxicity and cognitive impairment remains largely unknown. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a neuroprotective factor that has shown promise in various neurological disorders. However, its impact on sevoflurane-induced alterations has not been investigated. Thus, the objective of this study was to examine the effect of MANF in mitigating sevoflurane-induced neurotoxicity in young mice. Anesthesia with 3% sevoflurane 2 h daily was administered to young mice on postnatal day (P) 3, 6 and 9. We also constructed mono-macrophage-specific MANF knockout (MKO) mice in the mechanistic studies. Finally, the recombinant human MANF (rhMANF, 20 μg) protein was intraperitoneally administrated to neonatal mice before the sevoflurane anesthesia and the cognitive function, levels of pro-inflammatory cytokine and synapse-associated protein PSD95, the status of neural apoptosis, microglia activation and oxidative stress in hippocampus of the mice were investigated. The sevoflurane anesthesia increased the expression of endogenous MANF in the hippocampus, especially in microglia. MKO upregulated the expression of tumor necrosis factor-α (TNF-α), accelerated the neural apoptosis and the activation of microglia in hippocampus in young mice. MANF reversed the sevoflurane-induced cognitive impairment and inhibited the upregulation of TNF-α, the neural apoptosis and the reduction of the postsynaptic density protein-95 (PSD95) induced by sevoflurane anesthesia. Also, pretreatment with MANF alleviated the sevoflurane-induced activation of microglia and oxidative stress. Our current results demonstrated that MANF ameliorated neurotoxicity induced by the sevoflurane anesthesia in young mice, and such protective effect was associated with inhibition of microglia activation and neuroinflammation.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China
| | - Huiping Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China
- Anhui Public Health Clinical Center, Hefei, 230032, China
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, 225009, Jiangsu, China
| | - Leiying Zhou
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China
| | - Jiaqi Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China
| | - Enba Zhuo
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China
| | - Yujun Shen
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Xuesheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China.
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China.
| | - Qiying Shen
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road, Hefei, 230000, China.
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
11
|
Liang LR, Liu B, Cao SH, Zhao YY, Zeng T, Zhai MT, Fan Z, He DY, Ma SX, Shi XT, Zhang Y, Zhang H. Integrated ribosome and proteome analyses reveal insights into sevoflurane-induced long-term social behavior and cognitive dysfunctions through ADNP inhibition in neonatal mice. Zool Res 2024; 45:663-678. [PMID: 38766748 PMCID: PMC11188609 DOI: 10.24272/j.issn.2095-8137.2023.315] [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: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 05/22/2024] Open
Abstract
A growing number of studies have demonstrated that repeated exposure to sevoflurane during development results in persistent social abnormalities and cognitive impairment. Davunetide, an active fragment of the activity-dependent neuroprotective protein (ADNP), has been implicated in social and cognitive protection. However, the potential of davunetide to attenuate social deficits following sevoflurane exposure and the underlying developmental mechanisms remain poorly understood. In this study, ribosome and proteome profiles were analyzed to investigate the molecular basis of sevoflurane-induced social deficits in neonatal mice. The neuropathological basis was also explored using Golgi staining, morphological analysis, western blotting, electrophysiological analysis, and behavioral analysis. Results indicated that ADNP was significantly down-regulated following developmental exposure to sevoflurane. In adulthood, anterior cingulate cortex (ACC) neurons exposed to sevoflurane exhibited a decrease in dendrite number, total dendrite length, and spine density. Furthermore, the expression levels of Homer, PSD95, synaptophysin, and vglut2 were significantly reduced in the sevoflurane group. Patch-clamp recordings indicated reductions in both the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs). Notably, davunetide significantly ameliorated the synaptic defects, social behavior deficits, and cognitive impairments induced by sevoflurane. Mechanistic analysis revealed that loss of ADNP led to dysregulation of Ca 2+ activity via the Wnt/β-catenin signaling, resulting in decreased expression of synaptic proteins. Suppression of Wnt signaling was restored in the davunetide-treated group. Thus, ADNP was identified as a promising therapeutic target for the prevention and treatment of neurodevelopmental toxicity caused by general anesthetics. This study provides important insights into the mechanisms underlying social and cognitive disturbances caused by sevoflurane exposure in neonatal mice and elucidates the regulatory pathways involved.
Collapse
Affiliation(s)
- Li-Rong Liang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Shu-Hui Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - You-Yi Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Tian Zeng
- Department of Anesthesiology, 986th Air Force Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Mei-Ting Zhai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Ze Fan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Dan-Yi He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - San-Xin Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao-Tong Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yao Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Hui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032, China. E-mail:
| |
Collapse
|
12
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [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: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
13
|
Yan J, Ton H, Yan J, Dong Y, Xie Z, Jiang H. Anesthetic Sevoflurane Induces Enlargement of Dendritic Spine Heads in Mouse Neurons via Tau-Dependent Mechanisms. Anesth Analg 2024:00000539-990000000-00796. [PMID: 38507523 DOI: 10.1213/ane.0000000000006941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
BACKGROUND Sevoflurane induces neuronal dysfunction and cognitive impairment. However, the underlying mechanism remains largely to be determined. Tau, cyclophilin D, and dendritic spine contribute to cognitive function. But whether changes in dendritic spines are involved in the effects of sevoflurane and the potential association with tau and cyclophilin D is not clear. METHODS We harvested hippocampal neurons from wild-type mice, tau knockout mice, and cyclophilin D knockout mice. We treated these neurons with sevoflurane at day in vitro 7 and measured the diameter of dendritic spine head and the number of dendritic spines. Moreover, we determined the effects of sevoflurane on the expression of excitatory amino acid transporter 3 (EAAT3), extracellular glutamate levels, and miniature excitatory postsynaptic currents (mEPSCs). Finally, we used lithium, cyclosporine A, and overexpression of EAAT3 in the interaction studies. RESULTS Sevoflurane-induced tau phosphgorylation increased the diameter of dendritic spine head and decreased the number of dendritic spines in neurons harvested from wild-type and cyclophilin D knockout mice, but not tau knockout mice. Sevoflurane decreased the expression of EAAT3, increased extracellular glutamate levels, and decreased the frequency of mEPSCs in the neurons. Overexpression of EAAT3 mitigated the effects of sevoflurane on dendritic spines. Lithium, but not cyclosporine A, attenuated the effects of sevoflurane on dendritic spines. Lithium also inhibited the effects of sevoflurane on EAAT3 expression and mEPSCs. CONCLUSIONS These data suggest that sevoflurane induces a tau phosphorylation-dependent demtrimental effect on dendritic spine via decreasing EAAT3 expression and increasing extracellular glutamate levels, leading to neuronal dysfunction.
Collapse
Affiliation(s)
- Jia Yan
- From the Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Hoai Ton
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Jing Yan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Hong Jiang
- From the Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Zhang C, Chen X, Liu R, Zhao G. HSP90 Inhibition Attenuated Isoflurane-Induced Neurotoxicity in Mice and Human Neuroglioma Cells. Neurochem Res 2024; 49:706-717. [PMID: 38055149 DOI: 10.1007/s11064-023-04060-1] [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: 05/09/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023]
Abstract
Isoflurane, a widely used inhalation anesthetic in clinical practice, is associated with an increased risk of neuronal injury. Heat shock protein 90 (HSP90) plays a crucial role in maintaining neuronal homeostasis under stress conditions; however, its role during isoflurane exposure remains poorly understood. In this study, we aimed to investigate the protective effects of HSP90 inhibition and explore the regulatory mechanisms underlying these effects during isoflurane exposure. We found that the HSP90 inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17 AAG) has great protective effects in mitigating isoflurane-induced ferroptosis of mouse hippocampus and cultured neuronal cells. We focused on the activity of the crucial protein GPX4 in ferroptosis and found that 17 AAG exerted protective effects, preserving the physiological GPX4 activity under isoflurane exposure; further, 17 AAG restored the protein level of GPX4. Further, we observed that the chaperone-mediated autophagy (CMA) pathway was activated; 17 AAG also mediated GPX4 degradation under isoflurane exposure. Additionally, it interfered with the formation of complexes between HSP90 and Lamp-2a, inhibiting CMA activity, followed by the blockade of GPX4 degradation, further affecting the isoflurane-induced ferroptosis. Based on these findings, we proposed HSP90 inhibition as a protective mechanism against isoflurane-induced ferroptosis in neurons.
Collapse
Affiliation(s)
- Chunlu Zhang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xi Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ruizhu Liu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Guoqing Zhao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin University, Changchun, China.
| |
Collapse
|
15
|
Tsolaki M, Sia E, Giannouli V. Anesthesia and dementia: An up-to-date review of the existing literature. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:181-190. [PMID: 35981552 DOI: 10.1080/23279095.2022.2110871] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Concerns around the impact of anesthesia on cognitive decline and dementia, including Alzheimer's Disease (AD), have been increasing and recently attracting considerable attention in the research community. One unanswered question is whether anesthesia is a risk factor of dementia, specifically AD type dementia. A large body of evidence, coming from in vivo and in vitro models, suggests that exposure to anesthetic agents may increase the risk of AD through mechanisms of action similar to AD's neuropathology. In terms of clinical studies, our knowledge of the relationship between anesthesia and dementia is based on limited data, with most studies suggesting that there is no association. The aim of this paper was therefore to outline recent clinical studies exploring this controversial relationship and discuss future directions in terms of study design and potential areas of study. As the aging population and the prevalence of dementia and AD increases, we need a better understanding of anesthesia as a risk factor for neurodegeneration through well-designed studies. Despite the controversy, there seems to be little evidence to support that anesthesia itself or other surgical and patient factors can cause or accelerate AD.
Collapse
Affiliation(s)
- Magda Tsolaki
- 1st Department of Neurology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Greek Association of Alzheimer's Disease and Related Disorders, Thessaloniki, Greece
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI - AUTh), Thessaloniki, Greece
| | - Eleni Sia
- Greek Association of Alzheimer's Disease and Related Disorders, Thessaloniki, Greece
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI - AUTh), Thessaloniki, Greece
| | - Vaitsa Giannouli
- 1st Department of Neurology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Greek Association of Alzheimer's Disease and Related Disorders, Thessaloniki, Greece
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI - AUTh), Thessaloniki, Greece
| |
Collapse
|
16
|
Zuo Y, Xie J, Zhang X, Thirupathi A, Liu X, Zhang D, Zhang J, Shi Z. Sevoflurane causes cognitive impairment by inducing iron deficiency and inhibiting the proliferation of neural precursor cells in infant mice. CNS Neurosci Ther 2024; 30:e14612. [PMID: 38334030 PMCID: PMC10853893 DOI: 10.1111/cns.14612] [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: 11/08/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
AIMS Numerous studies on animals have shown that exposure to general anesthetics in infant stage may cause neurocognitive impairment. However, the exact mechanism is not clear. The dysfunction of iron metabolism can cause neurodevelopmental disorders. Therefore, we investigated the effect of iron metabolism disorder induced by sevoflurane (Sev) on cognitive function and the proliferation of neural precursor cells (NPCs) and neural stem cells (NSCs) in infant mice. METHODS C57BL/6 mice of postnatal day 14 and neural stem cells NE4C were treated with 2% Sev for 6 h. We used the Morris water maze (MWM) to test the cognitive function of infant mice. The proliferation of NPCs was measured using bromodeoxyuridine (BrdU) label and their markers Ki67 and Pax6 in infant brain tissues 12 h after anesthesia. Meanwhile, we used immunohistochemical stain, immunofluorescence assay, western blot, and flow cytometer to evaluate the myelinogenesis, iron levels, and cell proliferation in cortex and hippocampus or in NE4C cells. RESULTS The results showed that Sev significantly caused cognitive deficiency in infant mice. Further, we found that Sev inhibited oligodendrocytes proliferation and myelinogenesis by decreasing MBP and CC-1 expression and iron levels. Meanwhile, Sev also induced the iron deficiency in neurons and NSCs by downregulating FtH and FtL expression and upregulating the TfR1 expression in the cortex and hippocampus, which dramatically suppressed the proliferation of NSCs and NPCs as indicated by decreasing the colocalization of Pax6+ and BrdU+ cells, and caused the decrease in the number of neurons. Interestingly, iron supplementation before anesthesia significantly improved iron deficiency in cortex and hippocampus and cognitive deficiency induced by Sev in infant mice. Iron therapy inhibited the decrease of MBP expression, iron levels in neurons and oligodendrocytes, and DNA synthesis of Pax6+ cells in hippocampus induced by Sev. Meanwhile, the number of neurons was partially recovered in hippocampus. CONCLUSION The results from the present study demonstrated that Sev-induced iron deficiency might be a new mechanism of cognitive impairment caused by inhaled anesthetics in infant mice. Iron supplementation before anesthesia is an effective strategy to prevent cognitive impairment caused by Sev in infants.
Collapse
Affiliation(s)
- Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jinhong Xie
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Xue Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | | | - Xiaopeng Liu
- The Second Affiliated Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Di Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jianhua Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| |
Collapse
|
17
|
Zhang K, Chang Q, Li F, Li Y, Ding R, Yu Y. The locus coeruleus-dorsal hippocampal CA1 pathway is involved in depression-induced perioperative neurocognitive disorders in adult mice. CNS Neurosci Ther 2024; 30:e14406. [PMID: 37577850 PMCID: PMC10848051 DOI: 10.1111/cns.14406] [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: 06/09/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND Patients undergoing surgical anesthesia increasingly suffer from preoperative depression. Clinical studies have shown that depression is a risk factor for perioperative neurocognitive disorders (PNDs) in elder patients. However, the underlying mechanism, especially at the neural circuit level, remains poorly understood. METHODS Right carotid artery separation under sevoflurane and chronic social defeat stress (CSDS) in adult mice were used to establish surgical anesthesia and chronic depression models. Cognitive function was assessed by the Y maze and novel object recognition tests. A chemogenetic approach was used to modulate the locus coeruleus-dorsal hippocampal CA1 (LC-dCA1) circuit. Hippocampal synaptic alterations were evaluated by Golgi staining and whole-cell patch clamp recording. RESULTS We found that CSDS induced synaptic impairments in dorsal hippocampal CA1 pyramidal neurons and cognitive deficits in adult mice after surgery under sevoflurane. Chemogenetic activation of the LC-dCA1 pathway significantly alleviated the CSDS-induced synaptic impairments and cognitive dysfunction. On the contrary, inhibition of this pathway could mimic CSDS-induced deficits. Furthermore, we showed that dopamine played an important role in CSDS-induced PNDs in adult mice after surgery/sevoflurane. CONCLUSION Overall, our results have demonstrated a vital role for the LC-dCA1 pathway in CSDS-induced PNDs in adult mice undergoing surgery with sevoflurane anesthesia.
Collapse
Affiliation(s)
- Kai Zhang
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnesthesiologyTianjinChina
| | - Qianqian Chang
- School of PharmacyTianjin Medical UniversityTianjinChina
| | - Feixiang Li
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnesthesiologyTianjinChina
| | - Yun Li
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnesthesiologyTianjinChina
| | - Ran Ding
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
| | - Yonghao Yu
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnesthesiologyTianjinChina
| |
Collapse
|
18
|
Zhao M, Gu H, Pan W, Liu P, Zhu T, Shang H, Jia M, Yang J. SynCAM1 deficiency in the hippocampal parvalbumin interneurons contributes to sevoflurane-induced cognitive impairment in neonatal rats. CNS Neurosci Ther 2024; 30:e14554. [PMID: 38105652 PMCID: PMC10805405 DOI: 10.1111/cns.14554] [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: 08/22/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
AIMS Sevoflurane is widely used for general anesthesia in children. Previous studies reported that multiple neonatal exposures to sevoflurane can induce long-term cognitive impairment in adolescent rats, but the underlying mechanisms were not defined. METHODS Postnatal day 6 (P6) to P8 rat pups were exposed to 30% oxygen with or without 3% sevoflurane balanced with air. The Y maze test (YMT) and Morris water maze (MWM) tests were performed in some cohorts from age P35 to assess cognitive functions, and their brain samples were harvested at age P14, 21, 28, 35, and 42 for measurements of various molecular entities and in vivo electrophysiology experiments at age P35. RESULTS Sevoflurane exposure resulted in cognitive impairment that was associated with decreased synCAM1 expression in parvalbumin (PV) interneurons, a reduction of PV phenotype, disturbed gamma oscillations, and dendritic spine loss in the hippocampal CA3 region. Enriched environment (EE) increased synCAM1 expression in the PV interneurons and attenuated sevoflurane-induced cognitive impairment. The synCAM1 overexpression by the adeno-associated virus vector in the hippocampal CA3 region restored sevoflurane-induced cognitive impairment, PV phenotype loss, gamma oscillations decrease, and dendritic spine loss. CONCLUSION Our data suggested that neonatal sevoflurane exposure results in cognitive impairment through decreased synCAM1 expression in PV interneurons in the hippocampus.
Collapse
Affiliation(s)
- Ming‐ming Zhao
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Han‐wen Gu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Wei‐tong Pan
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Pan‐miao Liu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Ting‐ting Zhu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Hui‐jie Shang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jian‐jun Yang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
19
|
Chen K, Gupta R, Martín‐Ávila A, Cui M, Xie Z, Yang G. Anesthesia-induced hippocampal-cortical hyperactivity and tau hyperphosphorylation impair remote memory retrieval in Alzheimer's disease. Alzheimers Dement 2024; 20:494-510. [PMID: 37695022 PMCID: PMC10843666 DOI: 10.1002/alz.13464] [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: 03/23/2023] [Revised: 07/25/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Anesthesia often exacerbates memory recall difficulties in individuals with Alzheimer's disease (AD), but the underlying mechanisms remain unclear. METHODS We used in vivo Ca2+ imaging, viral-based circuit tracing, and chemogenetic approaches to investigate anesthesia-induced remote memory impairment in mouse models of presymptomatic AD. RESULTS Our study identified pyramidal neuron hyperactivity in the anterior cingulate cortex (ACC) as a significant contributor to anesthesia-induced remote memory impairment. This ACC hyperactivation arises from the disinhibition of local inhibitory circuits and increased excitatory inputs from the hippocampal CA1 region. Inhibiting hyperactivity in the CA1-ACC circuit improved memory recall after anesthesia. Moreover, anesthesia led to increased tau phosphorylation in the hippocampus, and inhibiting this hyperphosphorylation prevented ACC hyperactivity and subsequent memory impairment. DISCUSSION Hippocampal-cortical hyperactivity plays a role in anesthesia-induced remote memory impairment. Targeting tau hyperphosphorylation shows promise as a therapeutic strategy to mitigate anesthesia-induced neural network dysfunction and retrograde amnesia in AD.
Collapse
Affiliation(s)
- Kai Chen
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Riya Gupta
- Barnard College of Columbia UniversityNew YorkNew YorkUSA
| | | | - Meng Cui
- Department of BiologyPurdue UniversityWest LafayetteIndianaUSA
| | - Zhongcong Xie
- Geriatric Anesthesia Research UnitDepartment of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Guang Yang
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
20
|
Xu M, Xia L, Li J, Du Y, Dong Z. 7,8-Dihydroxyflavone ameliorates cognitive impairment induced by repeated neonatal sevoflurane exposures in mice through increasing tau O-GlcNAcylation. Neurosci Lett 2024; 818:137559. [PMID: 37984484 DOI: 10.1016/j.neulet.2023.137559] [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: 06/06/2023] [Revised: 10/07/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Sevoflurane, one of the most commonly used general anesthetics for pediatric anesthesia, has recently gained significant attention in both preclinical and clinical settings due to its potential neurotoxicity in the developing brain. Tau phosphorylation, induced by sevoflurane, is recognized as one of the major causes of neurotoxicity. 7,8-dihydroxyflavone (DHF), a TrkB receptor agonist, has been reported to exhibit potential neuroprotective effects against tauopathies. In this study, our objective was to investigate whether DHF could provide neuroprotective effects against sevoflurane-induced neurotoxicity and explore the underlying molecular mechanisms. METHODS Six-day-old mice were subjected to 2 h of anesthesia with 3 % sevoflurane, with or without pretreatment of DHF (5 mg/kg/day, i.p.) for 3 consecutive days. Autonomic motor ability was assessed by open-field test, while learning and memory abilities were evaluated by the fear conditioning test. Western blotting was conducted to measure the levels of t-TrkB, p-TrkB, tau, and phosphorylated tau. Additionally, a co-immunoprecipitation assay was performed to investigate the interaction between O-GlcNAcylation and tau. RESULTS Repeated neonatal sevoflurane exposures resulted in reduced freezing time during the context and cued fear conditioning tests in adulthood. However, pretreatment with DHF restored the freezing time to the level of the control group, indicating that DHF effectively alleviated cognitive impairments induced by neonatal sevoflurane exposure. We also observed that repeated neonatal sevoflurane exposures increased tau phosphorylation while decreasing tau O-GlcNAcylation. However, DHF pretreatment rebalanced the tau O-GlcNAcylation/phosphorylation ratio by enhancing the interaction between tau and O-GlcNAcylation. CONCLUSION Our findings demonstrate that DHF effectively ameliorates sevoflurane-induced cognitive impairment in developing mice by restoring the balance between tau O-GlcNAcylation and phosphorylation. Therefore, this study suggests that DHF has the potential to be a therapeutic agent for treating cognitive impairment associated with anesthetics, such as sevoflurane.
Collapse
Affiliation(s)
- Mingliang Xu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lei Xia
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Junjie Li
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yehong Du
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
21
|
Diachenko AI, Rodin IA, Krasnova TN, Klychnikov OI, Nefedova LN. The Role of Vitamin K in the Development of Neurodegenerative Diseases. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S57-S70. [PMID: 38621744 DOI: 10.1134/s0006297924140049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 04/17/2024]
Abstract
Neurodegenerative diseases are a growing global health problem with enormous consequences for individuals and society. The most common neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases, can be caused by both genetic factors (mutations) and epigenetic changes caused by the environment, in particular, oxidative stress. One of the factors contributing to the development of oxidative stress that has an important effect on the nervous system is vitamin K, which is involved in redox processes. However, its role in cells is ambiguous: accumulation of high concentrations of vitamin K increases the content of reactive oxygen species increases, while small amounts of vitamin K have a protective effect and activate the antioxidant defense systems. The main function of vitamin K is its involvement in the gamma carboxylation of the so-called Gla proteins. Some Gla proteins are expressed in the nervous system and participate in its development. Vitamin K deficiency can lead to a decrease or loss of function of Gla proteins in the nervous system. It is assumed that the level of vitamin K in the body is associated with specific changes involved in the development of dementia and cognitive abilities. Vitamin K also influences the sphingolipid profile in the brain, which also affects cognitive function. The role of vitamin K in the regulation of biochemical processes at the cellular and whole-organism levels has been studied insufficiently. Further research can lead to the discovery of new targets for vitamin K and development of personalized diets and therapies.
Collapse
Affiliation(s)
- Anna I Diachenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Igor A Rodin
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Tatiana N Krasnova
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Oleg I Klychnikov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Lidia N Nefedova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
22
|
Ocmen E, Erdost HA, Hanci V. The bibliometric analysis of most cited 100 papers in anesthesia-induced neurotoxicity. Medicine (Baltimore) 2023; 102:e36508. [PMID: 38050226 PMCID: PMC10695551 DOI: 10.1097/md.0000000000036508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/16/2023] [Indexed: 12/06/2023] Open
Abstract
Anesthesia-induced neurotoxicity is a major concern for anesthetists for more than 20 years. Many experimental and clinical studies have been conducted on this topic since late 1990s. However, bibliometric analysis of these papers has not been reported. In this study, we aimed to analyze the 100 most cited articles on anesthesia-induced neurotoxicity. It was planned as cross-sectional study. On January 30, 2023, we searched the "Web of Science (WOS)" database for anesthesia-induced neurotoxicity and most cited 100 papers about this topic were obtained. Data such as authors' names, year of publication, name of the journal, type of paper, and citation numbers were analyzed. The most cited 100 papers were read by the investigators, and the anesthetic, animal type in experimental studies, any protective agent and the method for detecting neurotoxicity used in the studies were also noted. There were 75 articles and 22 reviews in the 100 most cited articles. We found that most of the papers in most cited 100 list were published between 2010 to 1024. Most of the papers (11%) were from Harvard University and almost half of the papers (49%) were published in Anesthesiology. A great number of studies were performed in newborns or early childhood (85.5%) and inhalational anesthetics (54.7%) were the most studied anesthetic type. Most of the most cited 100 papers were published in Q1 journals (P = .012) and the continent of the most journals in this list was America (P = .014). The median total and annual citation numbers of funded papers were statistically significantly higher (P < .001 and P < .001 respectively). Anesthesia-induced neurotoxicity is very important, especially for pediatric anesthetists. This study is the first to conduct a bibliometric analysis of the most cited 100 publications on this field. Although there was a gap in the publications about this topic during COVID-19 pandemic, we believe that there will be many more publications on anesthesia-induced neurotoxicity since the mechanism, outcome and possible protection are still unknown.
Collapse
Affiliation(s)
- Elvan Ocmen
- Dokuz Eylul University, Department of Anesthesiology and Reanimation, Balçova/Izmir, Turkey
| | - Hale Aksu Erdost
- Dokuz Eylul University, Department of Anesthesiology and Reanimation, Balçova/Izmir, Turkey
| | - Volkan Hanci
- Dokuz Eylul University, Department of Anesthesiology and Reanimation, Balçova/Izmir, Turkey
| |
Collapse
|
23
|
Gong X, Li Q, Liu Y. Sevoflurane suppresses ALG13 transcription in a CREBBP-dependent manner to induce hippocampal damage and cognitive impairment. Neurosci Lett 2023; 818:137543. [PMID: 39492504 DOI: 10.1016/j.neulet.2023.137543] [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: 08/29/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND Sevoflurane (Sev) is a common clinical anesthetic but may lead to cognitive impairment. This study aims to deconstruct the underpinning molecular mechanism involved in Sev-induced neurological damage. METHODS Bioinformatics analyses was conducted to investigate candidate cognitive impairment-related physiological substrates of Sev. C57BL/6 mice and SH-SY5Y cells were exposed to Sev to generate animal and cellular models, respectively. Neurological impairment in mice was evaluated by Morris water maze test, modified Longa scoring, and pathological changes and cell apoptosis in the hippocampal tissues. In vitro, viability, apoptosis, and inflammatory cytokine concentration in SH-SY5Y cells were measured. Gain- or loss-of-function studies of CREB binding protein (CREBBP) and its predicted target asparagine-linked glycosylation 13 (ALG13) were performed in mice and in SH-SY5Y cells to investigate their roles in neural damage. RESULTS Sev treatment induced neurological deficit in mice and damage on SH-SY5Y cells, and reduced protein level of CREBBP protein in both models. CREBBP overexpression restored learning and memory ability of mice, reduced neurological deficit score, and reduced cell apoptosis while enhancing neuronal viability in the hippocampus. In vitro, the CREBBP overexpression increased viability while suppressing apoptosis and inflammation in SH-SY5Y cells. CREBBP bound to the ALG13 promoter to increase its transcription. Further knockdown of ALG13 negated the neuro-protective functions of CREBBP in vivo and in vitro. CONCLUSION This study demonstrates that Sev targets CREBBP to inhibit ALG13 transcription to induce hippocampal damage and cognitive impairment.
Collapse
Affiliation(s)
- Xiuping Gong
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Qi Li
- Department of Anesthesiology, South China Hospital Affiliated to Shenzhen University, Shenzhen 518000, Guangdong, PR China
| | - Yang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China.
| |
Collapse
|
24
|
Zeng S, Zhang J. Editorial: Anesthetic-induced neurotoxicity and neurocognitive impairment of vulnerable brains. Front Aging Neurosci 2023; 15:1293491. [PMID: 37885899 PMCID: PMC10598338 DOI: 10.3389/fnagi.2023.1293491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Affiliation(s)
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Chen M, Yan R, Ding L, Luo J, Ning J, Zhou R. Research Advances of Mitochondrial Dysfunction in Perioperative Neurocognitive Disorders. Neurochem Res 2023; 48:2983-2995. [PMID: 37294392 DOI: 10.1007/s11064-023-03962-4] [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: 12/21/2022] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/10/2023]
Abstract
Perioperative neurocognitive disorders (PND) increases postoperative dementia and mortality in patients and has no effective treatment. Although the detailed pathogenesis of PND is still elusive, a large amount of evidence suggests that damaged mitochondria may play an important role in the pathogenesis of PND. A healthy mitochondrial pool not only provides energy for neuronal metabolism but also maintains neuronal activity through other mitochondrial functions. Therefore, exploring the abnormal mitochondrial function in PND is beneficial for finding promising therapeutic targets for this disease. This article summarizes the research advances of mitochondrial energy metabolism disorder, inflammatory response and oxidative stress, mitochondrial quality control, mitochondria-associated endoplasmic reticulum membranes, and cell death in the pathogenesis of PND, and briefly describes the application of mitochondria-targeted therapies in PND.
Collapse
Affiliation(s)
- Mengjie Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Ruyu Yan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Lingling Ding
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Jiansheng Luo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Jiaqi Ning
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Ruiling Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| |
Collapse
|
26
|
Liang F, Li M, Xu M, Zhang Y, Dong Y, Soriano SG, McCann ME, Yang G, Xie Z. Sevoflurane anaesthesia induces cognitive impairment in young mice through sequential tau phosphorylation. Br J Anaesth 2023; 131:726-738. [PMID: 37537117 PMCID: PMC10541551 DOI: 10.1016/j.bja.2023.06.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The volatile anaesthetic sevoflurane induces time (single or multiple exposures)-dependent effects on tau phosphorylation and cognitive function in young mice. The underlying mechanism for this remains largely undetermined. METHODS Mice received 3% sevoflurane for 0.5 h or 2 h daily for 3 days on postnatal day (P) 6, 9, and 12. Another group of mice received 3% sevoflurane for 0.5 h or 1.5 h (3 × 0.5) on P6. We investigated effects of sevoflurane anaesthesia on tau phosphorylation on P6 or P12 mice, on cognitive function from P31 to P37, and on protein interactions, using in vivo studies, in vitro phosphorylation assays, and nanobeam single-molecule level interactions in vitro. RESULTS An initial sevoflurane exposure induced CaMKIIα phosphorylation (132 [11]% vs 100 [6]%, P<0.01), leading to tau phosphorylation at serine 262 (164 [7]% vs 100 [26]%, P<0.01) and tau detachment from microtubules. Subsequent exposures to the sevoflurane induced GSK3β activation, which phosphorylated detached or free tau (tau phosphorylated at serine 262) at serine 202 and threonine 205, resulting in cognitive impairment in young mice. In vitro phosphorylation assays also demonstrated sequential tau phosphorylation. Nanobeam analysis of molecular interactions showed different interactions between tau or free tau and CaMKIIα or GSK3β, and between tau and tubulin at a single-molecule level. CONCLUSIONS Multiple exposures to sevoflurane can induce sequential tau phosphorylation, leading to cognitive impairment in young mice, highlighting the need to investigate the underlying mechanisms of anaesthesia-induced tau phosphorylation in developing brain.
Collapse
Affiliation(s)
- Feng Liang
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Mengzhu Li
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Miao Xu
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiying Zhang
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yuanlin Dong
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Mary Ellen McCann
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Zhongcong Xie
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
27
|
Wang Y, Wu D, Li D, Zhou X, Fan D, Pan J. The role of PERK-eIF2α-ATF4-CHOP pathway in sevoflurane induced neuroapoptosis and cognitive dysfunction in aged mice. Cell Signal 2023; 110:110841. [PMID: 37549858 DOI: 10.1016/j.cellsig.2023.110841] [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: 05/28/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common surgical complication that causes additional pain in patients and affects their quality of life. To address this problem, emerging studies have focused on the POCD. Recent studies have shown that aging and anesthetic exposure are the two major risk factors for developing POCD. However, few reports described the exact molecular mechanisms underlying POCD in elderly patients. In the previous studies, the endoplasmic reticulum (ER) stress and neuroapoptosis in the hippocampus were associated with inducing POCD; however, no further information on the related signaling pathways could be disclosed. The PERK-eIF2α-ATF4-CHOP pathway is identified as the main regulatory pathway involved in ER stress and cell apoptosis. Therefore, we assume that the occurrence of POCD induced by sevoflurane inhalation may potentially result from ER stress and neuroapoptosis in the hippocampus of aged mice mediated by the PERK-eIF2α-ATF4-CHOP pathway. In our study, we found a relationship between sevoflurane inhalation concentration and memory decline in aged mice, with a 'ceiling effect'. We have confirmed that POCD induced by sevoflurane results from ER stress and neuroapoptosis in the hippocampus of aged mice, which is regulated by the over-expression of PERK-eIF2α-ATF4-CHOP pathway. Furthermore, we also showed that the dephosphorylation inhibitor of eIF2α (salubrinal) could down-regulate PERK-eIF2α-ATF4-CHOP pathway expression to inhibit ER stress and enhance the cognitive function of aged mice. In general, our study has elucidated one of the molecular mechanisms of sevoflurane-related cognitive dysfunction in aged groups and provided new strategies for treating sevoflurane-induced POCD.
Collapse
Affiliation(s)
- Yuhao Wang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Di Wu
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Danni Li
- Department of Anesthesiology, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, Chengdu 610072, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, PR China
| | - Xueer Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Dan Fan
- Department of Anesthesiology, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, Chengdu 610072, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, PR China.
| | - Jian Pan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
28
|
Zhong H, Ran X, Chen B, Xiong Y, Yu X. Apamin, an SK2 Inhibitor, Attenuated Neonatal Sevoflurane Exposures Caused Cognitive Deficits in Mice through the Regulation of Hippocampal Neuroinflammation. ACS Chem Neurosci 2023; 14:3409-3417. [PMID: 37647501 DOI: 10.1021/acschemneuro.3c00310] [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: 09/01/2023] Open
Abstract
Cognitive dysfunction induced by anesthesia in the infant is a crucial clinical issue that is still being debated and the focus of concern for the parents. However, the mechanism of cognitive decline caused by anesthesia and the corresponding treatment methods remain unclear. Postnatal day 7 (PND7) C57BL/6 mice included in the study were randomly divided into a control group (Control), a group with repeated exposure to sevoflurane (Sevo), and an Apamin intervention group (Sevo + Apamin). Apamin (0.5 μL at the concentration of 100 nmol/L) was injected into the bilateral hippocampus of mice. qRT-PCR, enzyme-linked immunosorbent assay (ELISA), and western blotting assay were used to evaluate the protein levels in the hippocampus. Object location memory (OLM) and novel object recognition (NOR) tasks, as well as elevated plus maze and contextual and cued fear conditioning tasks were used to evaluate the cognitive function of mice. Apamin mitigated sevoflurane-induced cognitive impairment of mice, sevoflurane-induced neuronal injury, and sevoflurane-induced activation of microglial in the hippocampus of the mice. Apamin inhibited M1-type polarization but promoted M2-type polarization of microglia after neonatal sevoflurane exposures in the hippocampus. In conclusion, Apamin attenuates neonatal sevoflurane exposures that cause cognitive deficits in mice through regulating hippocampal neuroinflammation.
Collapse
Affiliation(s)
- Heying Zhong
- Department of Anesthesiology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou 510000, Guangdong, China
| | - Xiaojuan Ran
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, Guizhou, China
| | - Bin Chen
- Department of Anesthesiology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yiqiang Xiong
- Department of Anesthesiology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Xiangdi Yu
- Department of Anesthesiology, Guizhou Provincial People's Hospital, No. 83 Zhongshan Road, Nanming District, Guiyang 550003, Guizhou, China
| |
Collapse
|
29
|
Qiu LL, Tan XX, Yang JJ, Ji MH, Zhang H, Zhao C, Xia JY, Sun J. Lactate Improves Long-term Cognitive Impairment Induced By Repeated Neonatal Sevoflurane Exposures Through SIRT1-mediated Regulation of Adult Hippocampal Neurogenesis and Synaptic Plasticity in Male Mice. Mol Neurobiol 2023; 60:5273-5291. [PMID: 37286723 DOI: 10.1007/s12035-023-03413-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
Repeated neonatal exposures to sevoflurane induce long-term cognitive impairment that has been reported to have sex-dependent differences. Exercise promotes learning and memory by releasing lactate from the muscle. The study tested the hypothesis that lactate may improve long-term cognitive impairment induced by repeated neonatal exposures to sevoflurane through SIRT1-mediated regulation of adult hippocampal neurogenesis and synaptic plasticity. C57BL/6 mice of both genders were exposed to 3% sevoflurane for 2 h daily from postnatal day 6 (P6) to P8. In the intervention experiments, mice received lactate at 1 g/kg intraperitoneally once daily from P21 to P41. Behavioral tests including open field (OF), object location (OL), novel object recognition (NOR), and fear conditioning (FC) tests were performed to assess cognitive function. The number of 5-Bromo-2'- deoxyuridine positive (BrdU+) cells and BrdU+/DCX+ (doublecortin) co-labeled cells, expressions of brain-derived neurotrophic factor (BDNF), activity-regulated cytoskeletal-associated protein (Arc), early growth response 1 (Egr-1), SIRT1, PGC-1α and FNDC5, and long-term potentiation (LTP) were evaluated in the hippocampus. Repeated exposures to sevoflurane induced deficits in OL, NOR and contextual FC tests in male but not female mice. Similarly, adult hippocampal neurogenesis, synaptic plasticity-related proteins and hippocampal LTP were impaired after repeated exposures to sevoflurane in male but not female mice, which could rescue by lactate treatment. Our study suggests that repeated neonatal exposures to sevoflurane inhibit adult hippocampal neurogenesis and induce defects of synaptic plasticity in male but not female mice, which may contribute to long-term cognitive impairment. Lactate treatment rescues these abnormalities through activation of SIRT1.
Collapse
Affiliation(s)
- Li-Li Qiu
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Xiao-Xiang Tan
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Jiao-Jiao Yang
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Mu-Huo Ji
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Zhang
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing, China
| | - Jiang-Yan Xia
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China.
| | - Jie Sun
- Department of Anesthesiology, Surgery and Pain Management, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
30
|
Yu Y, Yu Y, Zhang Y, Miao H. Editorial: Anesthetic neurotoxicity in developing brains: mechanisms, biomarkers, and therapeutic targets. Front Neurol 2023; 14:1279529. [PMID: 37719767 PMCID: PMC10501775 DOI: 10.3389/fneur.2023.1279529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- Yang Yu
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Huihui Miao
- Department of Anesthesia, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Hogarth K, Tarazi D, Maynes JT. The effects of general anesthetics on mitochondrial structure and function in the developing brain. Front Neurol 2023; 14:1179823. [PMID: 37533472 PMCID: PMC10390784 DOI: 10.3389/fneur.2023.1179823] [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: 03/04/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
The use of general anesthetics in modern clinical practice is commonly regarded as safe for healthy individuals, but exposures at the extreme ends of the age spectrum have been linked to chronic cognitive impairments and persistent functional and structural alterations to the nervous system. The accumulation of evidence at both the epidemiological and experimental level prompted the addition of a warning label to inhaled anesthetics by the Food and Drug Administration cautioning their use in children under 3 years of age. Though the mechanism by which anesthetics may induce these detrimental changes remains to be fully elucidated, increasing evidence implicates mitochondria as a potential primary target of anesthetic damage, meditating many of the associated neurotoxic effects. Along with their commonly cited role in energy production via oxidative phosphorylation, mitochondria also play a central role in other critical cellular processes including calcium buffering, cell death pathways, and metabolite synthesis. In addition to meeting their immense energy demands, neurons are particularly dependent on the proper function and spatial organization of mitochondria to mediate specialized functions including neurotransmitter trafficking and release. Mitochondrial dependence is further highlighted in the developing brain, requiring spatiotemporally complex and metabolically expensive processes such as neurogenesis, synaptogenesis, and synaptic pruning, making the consequence of functional alterations potentially impactful. To this end, we explore and summarize the current mechanistic understanding of the effects of anesthetic exposure on mitochondria in the developing nervous system. We will specifically focus on the impact of anesthetic agents on mitochondrial dynamics, apoptosis, bioenergetics, stress pathways, and redox homeostasis. In addition, we will highlight critical knowledge gaps, pertinent challenges, and potential therapeutic targets warranting future exploration to guide mechanistic and outcomes research.
Collapse
Affiliation(s)
- Kaley Hogarth
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Doorsa Tarazi
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Jason T. Maynes
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Zhu Y, Zhang M, Wang J, Wang Q. Knocking down Trim47 ameliorated sevoflurane-induced neuronal cell injury and cognitive impairment in rats. Exp Brain Res 2023; 241:1437-1446. [PMID: 37067562 DOI: 10.1007/s00221-023-06602-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/20/2023] [Indexed: 04/18/2023]
Abstract
Sevoflurane (SEV), usually causing neuronal damage and cognitive dysfunction, is one of the most commonly used anesthetics in clinical practice. However, the function of Trim47 in SEV-induced neuronal impairment remains elusive. The aim of this study was to study the effect of knocking down Trim47 on the nerve injury induced by SEV. Nerve injury was induced in rats by 3% SEV, and H19-7 was used to establish a pathological model, and sh-Trim47 was transfected into H19-7 to study the function of Trim47. The effects of SEV on the expression of Trim47 in the hippocampus and cognitive function of rats were studied by neurological function score and Moris water maze (MWM). The mRNA and protein expression of TNF-α, IL-1β and IL-6 in the cells, along with the neuronal apoptosis in the hippocampus of rats in each group were studied by TUNEL or WB. Flow cytometry was used to study the effect of knockdown of Trim47 on cell apoptosis. CCK-8 was used to detect cell viability of H19-7 cells. Finally, the potential signaling pathway affected by knockdown of Trim47 after abrogation of SEV induction was investigated by WB. The results showed that, knockdown of Trim47 ameliorated SEV-induced neurological damage and cognitive deficits, inflammation and neuronal cell apoptosis in rats, and promoted hippocampal neuronal activity. Knockdown of Trim47 can inhibit the NF-κB signaling pathway and improve neuronal cell damage and cognitive impairment induced by SEV in neonatal rats by regulating NF-κB signaling pathway, alleviating inflammatory response, and inhibiting neuronal apoptosis.
Collapse
Affiliation(s)
- Yingjun Zhu
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District, Huai'an, Jiangsu, China.
| | - Min Zhang
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District, Huai'an, Jiangsu, China
| | - Jiayu Wang
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District, Huai'an, Jiangsu, China
| | - Qingxiu Wang
- Department of Anesthesiology, The Affiliated Shanghai East Hospital of Tongji University, Shanghai, 200120, China
| |
Collapse
|
33
|
Lu P, Liang F, Dong Y, Xie Z, Zhang Y. Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration. Int J Mol Sci 2023; 24:ijms24076746. [PMID: 37047719 PMCID: PMC10095407 DOI: 10.3390/ijms24076746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
Collapse
Affiliation(s)
- Pan Lu
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
34
|
Liang J, Han S, Ye C, Zhu H, Wu J, Nie Y, Chai G, Zhao P, Zhang D. Minocycline Attenuates Sevoflurane-Induced Postoperative Cognitive Dysfunction in Aged Mice by Suppressing Hippocampal Apoptosis and the Notch Signaling Pathway-Mediated Neuroinflammation. Brain Sci 2023; 13:brainsci13030512. [PMID: 36979321 PMCID: PMC10046414 DOI: 10.3390/brainsci13030512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD), an important postoperative neurological complication, is very common and has an elevated incidence in elderly patients. Sevoflurane, an inhaled anesthetic, has been demonstrated to be associated with POCD in both clinical and animal studies. However, how to prevent POCD remains unclear. Minocycline, a commonly used antibiotic can cross the blood-brain barrier and exert an inhibitory effect on inflammation in the central nervous system. The present work aimed to examine the protective effect and mechanism of minocycline on sevoflurane-induced POCD in aged mice. We found that 3% sevoflurane administered 2 h a day for 3 consecutive days led to cognitive impairment in aged animals. Further investigation revealed that sevoflurane impaired synapse plasticity by causing apoptosis and neuroinflammation and thus induced cognitive dysfunction. However, minocycline pretreatment (50 mg/kg, i.p, 1 h prior to sevoflurane exposure) significantly attenuated learning and memory impairments associated with sevoflurane in aged animals by suppressing apoptosis and neuroinflammation. Moreover, a mechanistic analysis showed that minocycline suppressed sevoflurane-triggered neuroinflammation by inhibiting Notch signaling. Similar results were also obtained in vitro. Collectively, these findings suggested minocycline may be an effective drug for the prevention of sevoflurane-induced POCD in elderly patients.
Collapse
Affiliation(s)
- Junjie Liang
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi 214002, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Shanshan Han
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi 214002, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Chao Ye
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Haimeng Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Jiajun Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Gaoshang Chai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Dengxin Zhang
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi 214002, China
| |
Collapse
|
35
|
Fu N, Wang Y, Zhu R, Li N, Zeng S, Miao M, Yang Y, Sun M, Zhang J. Bicuculline and Bumetanide Attenuate Sevoflurane-Induced Impairment of Myelination and Cognition in Young Mice. ACS Chem Neurosci 2023; 14:1146-1155. [PMID: 36802490 DOI: 10.1021/acschemneuro.2c00764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Sevoflurane (Sevo) is one of the most commonly used general anesthetics for infants and young children. We investigated whether Sevo impairs neurological functions, myelination, and cognition via the γ-aminobutyric acid A receptor (GABAAR) and Na+-K+-2Cl- cotransporter (NKCC1) in neonatal mice. On postnatal days 5-7, mice were exposed to 3% Sevo for 2 h. On postnatal day 14, mouse brains were dissected, and oligodendrocyte precursor cell line level lentivirus knockdown of GABRB3, immunofluorescence, and transwell migration assays were performed. Finally, behavioral tests were conducted. Multiple Sevo exposure groups exhibited increased neuronal apoptosis levels and decreased neurofilament protein levels in the mouse cortex compared with the control group. Sevo exposure inhibited the proliferation, differentiation, and migration of the oligodendrocyte precursor cells, thereby affecting their maturation process. Electron microscopy revealed that Sevo exposure reduced myelin sheath thickness. The behavioral tests showed that multiple Sevo exposures induced cognitive impairment. GABAAR and NKCC1 inhibition provided protection against Sevo-induced neurotoxicity and cognitive dysfunction. Thus, bicuculline and bumetanide can protect against Sevo-induced neuronal injury, myelination impairment, and cognitive dysfunction in neonatal mice. Furthermore, GABAAR and NKCC1 may be mediators of Sevo-induced myelination impairment and cognitive dysfunction.
Collapse
Affiliation(s)
- Ningning Fu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yangyang Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ruilou Zhu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ningning Li
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shuang Zeng
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengrong Miao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yitian Yang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
36
|
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
|
37
|
Zhong Y, Zhang C, Wang Y, Tang C, Ren J, Wang M, Liu D, Zhu Z. Multiple exposures to sevoflurane across postnatal development may cause cognitive deficits in older age. Pediatr Res 2023; 93:838-844. [PMID: 35804157 DOI: 10.1038/s41390-022-01943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND The aim of the study was to determine the effects of repeated anesthesia exposure across postnatal development. METHODS Seventy-two newborn Sprague-Dawley rats were randomly divided into Sev group and Con-aged group. Sev groups were exposed to 2.6% sevoflurane for 2 h on postnatal day (P) 7, P14, and P21; the Con groups only received carrier gas for 2 h. Learning and memory were evaluated using the MWM test at P31 (juvenile), P91 (adult), and 18 months postnatally (aged). The relative expression of APP and Mapt mRNA was detected by RT-PCR, while Aβ, tau, and P-tau protein levels were analyzed by immunohistochemistry. RESULTS After repeated inhalation of sevoflurane, MWM test performance was significantly decreased in the Sev-aged group compared to the Con-aged group (P > 0.05). The relative expression of APP and Mapt mRNA was not significantly different between groups in each growth period (P > 0.05). The tau expression in the juvenile hippocampal CA1, CA3, and dentate gyrus regions increased markedly in the Sev group, while P-tau only increased in the hippocampal CA3 region in the Sev-adult group. The expression of tau, P-tau, and Aβ in the hippocampal regions was upregulated in the Sev-aged group. CONCLUSIONS Multiple exposures to sevoflurane across postnatal development can induce or aggravate cognitive impairment in old age. IMPACT Whether multiple sevoflurane exposures across postnatal development cause cognitive impairment in childhood, adulthood, or old age, as well as the relationship between sevoflurane and the hippocampal Aβ, tau, and P-tau proteins, remains unknown. This study's results demonstrate that multiple exposures to sevoflurane across postnatal development do not appear to affect cognitive function in childhood and adulthood; however, multiple exposures may lead to a cognitive function deficit in old age. The underlying mechanism may involve overexpression of the tau, P-tau, and Aβ proteins in the hippocampus.
Collapse
Affiliation(s)
- Yuanping Zhong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, PR China
| | - Chao Zhang
- Guizhou Key Laboratory of Basic Research of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, Guizhou, PR China
| | - Yi Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, PR China
| | - Chunchun Tang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, PR China
| | - Juanjuan Ren
- Affiliated Hospital of Jining Medical University, Jining, 272000, Shan Dong, PR China
| | - Mengmeng Wang
- Women and Children's Hospital, Qingdao University, Qing Dao, 266000, Shan Dong, PR China
| | - Dexing Liu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, PR China
| | - Zhaoqiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, PR China.
- Guizhou Key Laboratory of Basic Research of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, Guizhou, PR China.
| |
Collapse
|
38
|
Li J, Li L, He J, Xu J, Bao F. The NLRP3 inflammasome is a potential mechanism and therapeutic target for perioperative neurocognitive disorders. Front Aging Neurosci 2023; 14:1072003. [PMID: 36688154 PMCID: PMC9845955 DOI: 10.3389/fnagi.2022.1072003] [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: 10/17/2022] [Accepted: 11/21/2022] [Indexed: 01/06/2023] Open
Abstract
Perioperative neurocognitive disorders (PNDs) are frequent complications associated with cognitive impairment during the perioperative period, including acute postoperative delirium and long-lasting postoperative cognitive dysfunction. There are some risk factors for PNDs, such as age, surgical trauma, anesthetics, and the health of the patient, but the underlying mechanism has not been fully elucidated. Pyroptosis is a form of programmed cell death that is mediated by the gasdermin protein and is involved in cognitive dysfunction disorders. The canonical pathway induced by nucleotide oligomerization domain (NOD)-, leucine-rich repeat (LRR)- and pyrin domain-containing protein 3 (NLRP3) inflammasomes contributes to PNDs, which suggests that targeting NLRP3 inflammasomes may be an effective strategy for the treatment of PNDs. Therefore, inhibiting upstream activators and blocking the assembly of the NLRP3 inflammasome may attenuate PNDs. The present review summarizes recent studies and systematically describes the pathogenesis of NLRP3 activation and regulation and potential therapeutics targeting NLRP3 inflammasomes in PNDs patients.
Collapse
Affiliation(s)
- Jiayue Li
- Department of Anesthesiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Li Li
- Department of Anesthesiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Jiannan He
- Department of Anesthesiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Jianhong Xu
- Department of Anesthesiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Fangping Bao
- Department of Anesthesiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China,Department of Anesthesiology, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Fangping Bao,
| |
Collapse
|
39
|
Emekli-Alturfan E, Alturfan AA. The emerging relationship between vitamin K and neurodegenerative diseases: a review of current evidence. Mol Biol Rep 2023; 50:815-828. [PMID: 36329336 DOI: 10.1007/s11033-022-07925-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/06/2022] [Indexed: 11/06/2022]
Abstract
Neurodegenerative disease refers to a group of disorders that predominantly damage the neurons in the brain. Despite significant progress in the knowledge of neurodegenerative diseases, there is currently no disease-modifying drug available. Vitamin K was first established for its involvement in blood clotting, but there is now compelling evidence indicating its role in the neurological system. In particular, the results of recent studies on the effects of vitamin K2 on preventing apoptosis, oxidative stress, and microglial activation in neuron cells through its role in electron transport are very promising against Alzheimer's disease. In addition to its protective effect on cognitive functions, its inhibitory effects on inflammation and α-synuclein fibrillization in Parkinson's disease, which has been revealed in recent years, are remarkable. Although there are many studies on the mechanism and possible treatment methods of neurodegenerative diseases, especially Parkinson's and Alzheimer's disease, studies on the relationship between vitamin K and neurodegenerative diseases are very limited, yet have promising findings. Vitamin K has also been proposed for therapeutic use in multiple sclerosis patients to lower the intensity or to slow down the progression of the disease. Accordingly, the aim of this study is to review the current evidence for the use of vitamin K supplementation in neurodegenerative diseases, in particular Alzheimer's disease, Parkinson's disease, and multiple sclerosis.
Collapse
Affiliation(s)
- Ebru Emekli-Alturfan
- Faculty of Dentistry, Department of Basic Medical Sciences, Marmara University, Istanbul, Turkey.
| | - A Ata Alturfan
- Faculty of Medicine, Department of Biochemistry, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
40
|
Zhao L, Wang Z, Chen H, Du Y, Ma W, Tao Q, Ma X, Wu Z, Peng J. Effects of lncRNA HOXA11-AS on Sevoflurane-Induced Neuronal Apoptosis and Inflammatory Responses by Regulating miR-98-5p/EphA4. Mediators Inflamm 2023; 2023:7750134. [PMID: 37064501 PMCID: PMC10098412 DOI: 10.1155/2023/7750134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/19/2022] [Accepted: 03/20/2023] [Indexed: 04/18/2023] Open
Abstract
Objective To explore the molecular mechanism of sevoflurane-induced neurotoxicity and to determine whether lncRNA HOXA11-AS affects sevoflurane-induced neuronal apoptosis and inflammation by regulating miR-98-5p/EphA4. Methods Morris water maze (MWM) test was used to detect the learning and memory ability of rats, HE staining was used to observe hippocampal pathology, TUNEL staining was used to detect the level of neuronal apoptosis, and RT-qPCR was used to detect the expression of HOXA11-AS, miR-98-5p, IL-6, IL-1β, and TNF-α. At the same time, the contents of IL-6, IL-1β, and TNF-α in serum were detected by ELISA. The expressions of apoptosis-related proteins EphA4, Bax, Cleaved caspase 3, and Bcl-2 were detected by Western blot. The dual-luciferase gene reporter verified the targeting relationship between HOXA11-AS and miR-98-5p and the targeting relationship between miR-98-5p and EphA4. Results The expression of HOXA11-AS was observed in sevoflurane-treated rats or cells and promoted neuronal apoptosis and inflammation. HOXA11-AS was knocked out alone, or miR-98-5p was overexpressed which attenuates neuronal apoptosis and inflammatory inflammation after sevoflurane treatment. Furthermore, knockdown of HOXA11-AS alone was partially restored by knockdown of miR-98-5p or overexpression of EphA4. Conclusion Inhibition of lncRNA HOXA11-AS attenuates sevoflurane-induced neuronal apoptosis and inflammatory responses via miR-98-5p/EphA4.
Collapse
Affiliation(s)
- Li Zhao
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Zhonghui Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Haitao Chen
- Department of Ultrasound, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Yaxi Du
- Department of Molecular Diagnosis Center, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Weihao Ma
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Qunfen Tao
- Department of Operation Room, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Yunnan, China
| | - Xiang Ma
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Zeming Wu
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| | - Jing Peng
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118 Yunnan, China
| |
Collapse
|
41
|
Chen Z, Wang S, Meng Z, Ye Y, Shan G, Wang X, Zhao X, Jin Y. Tau protein plays a role in the mechanism of cognitive disorders induced by anesthetic drugs. Front Neurosci 2023; 17:1145318. [PMID: 36937655 PMCID: PMC10015606 DOI: 10.3389/fnins.2023.1145318] [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: 01/16/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Cognitive disorders are mental health disorders that can affect cognitive ability. Surgery and anesthesia have been proposed to increase the incidence of cognitive dysfunction, including declines in memory, learning, attention and executive function. Tau protein is a microtubule-associated protein located in the axons of neurons and is important for microtubule assembly and stability; its biological function is mainly regulated by phosphorylation. Phosphorylated tau protein has been associated with cognitive dysfunction mediated by disrupting the stability of the microtubule structure. There is an increasing consensus that anesthetic drugs can cause cognitive impairment. Herein, we reviewed the latest literature and compared the relationship between tau protein and cognitive impairment caused by different anesthetics. Our results substantiated that tau protein phosphorylation is essential in cognitive dysfunction caused by anesthetic drugs, and the possible mechanism can be summarized as "anesthetic drugs-kinase/phosphatase-p-Tau-cognitive impairment".
Collapse
|
42
|
Feng J, Lin H, Zhao Y, Yang Y, Zhuang X, Yu Y, Yu Y. Tandem mass tag-based quantitative proteomic analysis of effects of multiple sevoflurane exposures on the cerebral cortex of neonatal and adult mice. Front Neurol 2022; 13:1056947. [PMID: 36582614 PMCID: PMC9792844 DOI: 10.3389/fneur.2022.1056947] [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: 09/29/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction Sevoflurane is the most commonly used general anesthetic in pediatric surgery, but it has the potential to be neurotoxic. Previous research found that long-term or multiple sevoflurane exposures could cause cognitive deficits in newborn mice but not adult mice, whereas short-term or single inhalations had little effect on cognitive function at both ages. The mechanisms behind these effects, however, are unclear. Methods In the current study, 6- and 60-day-old C57bl mice in the sevoflurane groups were given 3% sevoflurane plus 60% oxygen for three consecutive days, each lasting 2 hours, while those in the control group only got 60% oxygen. The cortex tissues were harvested on the 8th or 62nd day. The tandem mass tags (TMT)pro-based quantitative proteomics combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis, Golgi staining, and western blotting analysis were applied to analyze the influences of multiple sevoflurane anesthesia on the cerebral cortex in mice with various ages. The Morris water maze (MWM) test was performed from postnatal day (P)30 to P36 or P84 to P90 after control or multiple sevoflurane treatment. Sevoflurane anesthesia affected spatial learning and memory and diminished dendritic spines primarily in newborn mice, whereas mature animals exhibited no significant alterations. Results A total of 6247 proteins were measured using the combined quantitative proteomics methods of TMTpro-labeled and LC-MS/MS, 443 of which were associated to the age-dependent neurotoxic mechanism of repeated sevoflurane anesthesia. Furthermore, western blotting research revealed that sevoflurane-induced brain damage in newborn mice may be mediated by increasing the levels of protein expression of CHGB, PTEN, MAP2c, or decreasing the level of SOD2 protein expression. Conclusion Our findings would help to further the mechanistic study of age-dependent anesthetic neurotoxicity and contribute to seek for effective protection in the developing brain under general anesthesia.
Collapse
Affiliation(s)
- Jingyu Feng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China
| | - Hua Lin
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yue Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yongyan Yang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China
| | - Xiaoli Zhuang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China,*Correspondence: Yang Yu
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Anesthesiology, Tianjin, China,Yonghao Yu
| |
Collapse
|
43
|
Wang Y, Zhou WL, Feng DX. Application of different concentrations of sevoflurane with remifentanil in radical surgery for gastrointestinal tumors: Effects on intraoperative hemodynamics and postoperative anesthetic recovery. Shijie Huaren Xiaohua Zazhi 2022; 30:1039-1045. [DOI: 10.11569/wcjd.v30.i23.1039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022] Open
Abstract
BACKGROUND The combination of remifentanil and sevoflurane can provide patients with sufficient analgesic and sedative effects. Anesthesia for gastrointestinal tumor surgery not only needs to reach the required depth of anesthesia for surgery, but also needs to reduce the surgical stress to ensure rapid recovery after surgery.
AIM To explore the application of different concentrations of sevoflurane with remifentanil in radical surgery for gastrointestinal tumors and the effects on intraoperative hemodynamics and postoperative anesthesia recovery.
METHODS Eighty-six patients undergoing radical surgery for gastrointestinal tumors at our hospital from January 2020 to December 2021 were selected and divided into two groups by random number table method, with 43 cases in each group. Sevoflurane at a 1.0 minimum alveolar effective concentration (MAC) with remifentanil was adopted in group A, and 1.5 MAC sevoflurane with remifentanil was adopted in group B. The quality of anesthesia, intraoperative hemodynamics [mean arterial pressure (MAP) and heart rate (HR)], cerebral oxygen metabolism [cerebral oxygen uptake rate (CERO2) and jugular venous oxygen content (SjvO2)] at different time points, adverse events during anesthesia maintenance, and postoperative anesthesia recovery were recorded in the two groups.
RESULTS During maintenance of anesthesia, the rate of adjustment of remifentanil pumping rate was lower in group A than in group B (32.56% vs 67.44%, P < 0.05). There was no significant difference in HR or MAP at each time point between the two groups (P > 0.05), and both HR and MAP were within the normal range. There was no significant difference in CERO2 or SjvO2 at each time point between the two groups (P > 0.05); CERO2 at T2, T3, T4, T5, and T6 in both groups was lower than that at T1, and SjvO2 was higher than that at T1 (P < 0.05). The incidence of hypotension during anesthesia maintenance in group A was lower than that in group B (P < 0.05). The times to open eyes on command, recovery of spontaneous breathing, extubation, and exit from the room during anesthesia awakening were shorter in group A than in group B (P < 0.05).
CONCLUSION Both 1.0 MAC and 1.5 MAC sevoflurane can meet the demand for anesthesia maintenance in surgery for radical gastrointestinal tumors; however, 1.0 MAC sevoflurane can provide better quality of anesthesia maintenance with less effect on cerebral oxygen metabolism, which can significantly improve the quality of awakening and shorten the anesthesia awakening time, and is conducive to faster clinical turnaround.
Collapse
Affiliation(s)
- Ying Wang
- Panan County People's Hospital of Jinhua City, Jinhua 322300, Zhejiang Province, China
| | - Wei-Lan Zhou
- Panan County People's Hospital of Jinhua City, Jinhua 322300, Zhejiang Province, China
| | - Ding-Xiang Feng
- Panan County People's Hospital of Jinhua City, Jinhua 322300, Zhejiang Province, China
| |
Collapse
|
44
|
Zhu Y, Zhang M, Wang J, Wang Q. Lin28A Reduced Sevoflurane-Induced Nerve Injury and Cognitive Dysfunction by Inhibiting Tau Acetylation and Phosphorylation via Activating SIRT1 in Elderly Rats. Neurotox Res 2022; 40:1913-1923. [PMID: 36322362 DOI: 10.1007/s12640-022-00594-4] [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: 05/17/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
Abstract
Sevoflurane (Sev) might cause neurotoxicity in elderly rats. However, the role of Lin28A in Sev-induced neurotoxicity remains unclear in elderly rats. In this study, elderly rats were used to construct an Sev-induced nerve injury model. Learning and memory abilities were assessed by Morris water maze (MWM) trainings; pathological alterations in hippocampal region were assessed by HE staining; neuronal apoptosis was assessed by TUNEL; related protein expression was analyzed by immunofluorescence, immunohistochemistry, and Western blotting. Results of this study showed that Sev treatment caused nerve injury and cognitive dysfunction in elderly rats, with increased neuronal apoptosis and decreased Lin28A levels. Pathological impairment and learning and memory abilities of elderly rats were significantly improved after forced overexpression of Lin28A using AAV, accompanied by decreased expression of CD68, Iba-1, and GFAP. TUNEL analysis showed that Lin28A overexpression significantly reversed Sev-induced neuronal apoptosis. Further mechanistic analysis showed that Lin28A significantly promoted SIRT1 expression, which further reversed Sev-induced Tau acetylation at lysine 280 and 686 and Tau hyperphosphorylation, thereby alleviating nerve injury and cognitive dysfunction in elderly rats. The introduction of SIRT1 inhibitor EX527 further confirmed the involvement of SIRT1 in the regulation of Lin28A in elderly rats. In conclusion, our findings demonstrated that Lin28A reduced sevoflurane-induced nerve injury and cognitive dysfunction by inhibiting Tau acetylation and phosphorylation via activating SIRT1 in elderly rats.
Collapse
Affiliation(s)
- Yingjun Zhu
- Department of Anesthesiology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District , 223300, Huai'an, Jiangsu, China.
| | - Min Zhang
- Department of Anesthesiology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District , 223300, Huai'an, Jiangsu, China
| | - Jiayu Wang
- Department of Anesthesiology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, No. 6 West Beijing Road, Huaiyin District , 223300, Huai'an, Jiangsu, China
| | - Qingxiu Wang
- Department of Anesthesiology, The Affiliated Shanghai East Hospital of Tongji University, Shanghai, 200120, China
| |
Collapse
|
45
|
Chen YR, Zhang SX, Fang M, Zhang P, Zhou YF, Yu X, Zhang XN, Chen G. Egr2 contributes to age-dependent vulnerability to sevoflurane-induced cognitive deficits in mice. Acta Pharmacol Sin 2022; 43:2828-2840. [PMID: 35577909 PMCID: PMC9622904 DOI: 10.1038/s41401-022-00915-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/18/2022] [Indexed: 01/27/2023] Open
Abstract
Sevoflurane inhalation is prone to initiate cognitive deficits in infants. The early growth response-2 (Egr-2) gene is DNA-binding transcription factor, involving in cognitive function. In this study we explored the molecular mechanisms underlying the vulnerability to cognitive deficits after sevoflurane administration. Six-day-old (young) and 6-week-old (early adult) mice received anesthesia with 3% sevoflurane for 2 h daily for 3 days. We showed that multiple exposures of sevoflurane induced significant learning ability impairment in young but not early adult mice, assessed in Morris water maze test on postnatal days 65. The integrated differential expression analysis revealed distinct transcription responses of Egr family members in the hippocampus of the young and early adult mice after sevoflurane administration. Particularly, Egr2 was significantly upregulated after sevoflurane exposure only in young mice. Microinjection of Egr2 shRNA recombinant adeno-associated virus into the dentate gyrus alleviated sevoflurane-induced cognitive deficits, and abolished sevoflurane-induced dendritic spins loss and BDNF downregulation in young mice. On the contrary, microinjection of the Egr2 overexpression virus in the dentate gyrus aggravated learning ability impairment induced by sevoflurane in young mice but not early adult mice. Furthermore, we revealed that sevoflurane markedly upregulated the nuclear factors of activated T-cells NFATC1 and NFATC2 in young mice, which were involved in Egr2 regulation. In conclusion, Egr2 serves as a critical factor for age-dependent vulnerability to sevoflurane-induced cognitive deficits.
Collapse
Affiliation(s)
- Ye-Ru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Shu-Xia Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Man Fang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Piao Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - You-Fa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiang-Nan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
46
|
Chen BZ, Jiang LH, Zhou W, Shang YC, Li F, Liu B. Repeated Sevoflurane Exposure in Neonatal Rats Enhances the Sensitivity to Pain and Traumatic Stress Later in Juvenile Life. J Pain Res 2022; 15:3171-3178. [PMID: 36258761 PMCID: PMC9572549 DOI: 10.2147/jpr.s365253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Purposeː Sevoflurane exposure in the neonatal period of rodent animals was reported to be associated with neuroendocrine dysregulations later in life. We tested the hypothesis that repeated sevoflurane exposure in neonatal rats enhances the sensitivity to pain and acute traumatic stress response later in juvenile life and investigated whether the neonatal brain depolarizing γ-aminobutyric acid type A receptor (GABAAR) activity is involved in mediating these abnormalities. Methodsː The postnatal 6 days (P6) Sprague-Dawley male rat pups pretreated with vehicle or the NKCC1 inhibitor, bumetanide, received sequential exposures to 2.1% sevoflurane exposure for 2 hours daily in 3 consecutive days. Resultsː The results showed that repeated exposures to sevoflurane in neonatal rats significantly reduced the paw withdrawal thermal latency (PWTL) at P9, P45. Repeated exposures to sevoflurane in neonatal rats did not significantly affect the basal secretion of serum corticosterone at juvenile period P45, whereas the level of corticosterone for neonatal sevoflurane-exposed rats at P45 was significantly higher than the CON group after subject to conditioned fear traumatic stress (CFTS). The resulting NKCC1/KCC2 mRNA ratio was significantly increased immediately after the neonatal rats received the last sevoflurane exposure, which was alleviated by pretreated with the NKCC1 inhibitor bumetanide. Conclusionː Repeated exposures to sevoflurane in neonatal rats enhanced the sensitivity to pain and acute traumatic stress response in juvenile life. The neonatal brain depolarizing GABAAR activity is involved in mediating these abnormalities.
Collapse
Affiliation(s)
- Ben-Zhen Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China,Department of Anesthesiology, Sichuan Provincial Women’s and Children’s Hospital, Chengdu, People’s Republic of China
| | - Li-Hua Jiang
- Department of Operating Room Nursing, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Wenqin Zhou
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yu-Chao Shang
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Fang Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Bin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China,Correspondence: Bin Liu, Department of Anesthesiology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, 610041, People’s Republic of China, Tel +86-13408669172, Email
| |
Collapse
|
47
|
Identification and Validation of Ferroptosis-Related Genes in Sevoflurane-Induced Hippocampal Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4435161. [PMID: 36238640 PMCID: PMC9553355 DOI: 10.1155/2022/4435161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022]
Abstract
Background Sevoflurane is one of the most popular inhalational anesthetics during perioperative period but presenting neurotoxicity among pediatric and aged populations. Recent experiments in vivo and in vitro have indicated that ferroptosis may contribute to the neurotoxicity of sevoflurane anesthesia. However, the exact mechanism is still unclear. Methods In current study, we explored the differential expressed genes (DEGs) in HT-22 mouse hippocampal neuronal cells after sevoflurane anesthesia using RNA-seq. Differential expressed ferroptosis-related genes (DEFRGs) were screened and analyzed by Gene Ontology (GO) and pathway enrichment analysis. Protein-to-protein interaction (PPI) network was constructed by the Search Tool for the Retrieval of Interacting Genes (STRING). Significant modules and the hub genes were identified by using Cytoscape. The Connectivity Map (cMAP) was used for screening drug candidates targeting the identified DEFRGs. Potential TF-gene network and drug-gene pairs were established towards the hub genes. In final, we validated these results in experiments. Results A total of 37 ferroptosis-related genes (18 upregulated and 19 downregulated) after sevoflurane exposure in hippocampal neuronal cells were finally identified. These differentially expressed genes were mainly involved into the biological processes of cellular response to oxidative stress. Pathway analysis indicated that these genes were involved in ferroptosis, mTOR signaling pathway, and longevity-regulating pathway. PPI network was constructed. 10 hub genes including Prkaa2, Chac1, Arntl, Tfrc, Slc7a11, Atf4, Mgst1, Lpin1, Atf3, and Sesn2 were found. Top 10 drug candidates, gene-drug networks, and TFs targeting these genes were finally identified. These results were validated in experiments. Conclusion Our results suggested that ferroptosis-related genes play roles in sevoflurane anesthesia-related hippocampal neuron injury and offered the hub genes and potential therapeutic agents for investigating and treatment of this neurotoxicity after sevoflurane exposure. Finally, therapeutic effect of these drug candidates and function of potential ferroptosis targets should be further investigated for treatment and clarifying mechanisms of sevoflurane anesthesia-induced neuron injury in future research.
Collapse
|
48
|
Sheng Z, Liu Q, Cheng C, Li M, Barash J, Kofke WA, Shen Y, Xie Z. Fentanyl induces autism-like behaviours in mice by hypermethylation of the glutamate receptor gene Grin2b. Br J Anaesth 2022; 129:544-554. [PMID: 35697546 DOI: 10.1016/j.bja.2022.04.027] [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: 12/07/2021] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Environmental factors contribute to autism spectrum disorder. Fentanyl, one of the most widely used opioid analgesics in anaesthesia, can induce neurotoxicity, but its role in autism remains unknown. We determined whether fentanyl induced autism-like behaviours in young mice and the underlying mechanisms. METHODS Young male and female mice received fentanyl at postnatal days 6, 8, and 10, and performed behavioural tests, including three-chamber social preference, elevated plus maze, grooming behaviour, and open-field test, from postnatal days 30-32. Expression of Grin2b, the gene encoding the GluN2B subunit of the N-methyl-d-aspartate receptor, was assessed in the anterior cingulate cortex of male mice using fluorescence in situ hybridisation histochemistry. We used bisulfite target sequencing to determine Grin2b hypermethylation sites after fentanyl treatment. In the specific activation and rescue experiments, we injected the mu opioid receptor agonist [D-Ala,2 N-MePhe,4 Gly-ol]-enkephalin (DAMGO) or Grin2b overexpression lentivirus into the anterior cingulate cortex of male mice. RESULTS Fentanyl induced autism-like behaviours in both young male and female mice, and downregulated Grin2b expression (0.49-fold [0.08] vs 1.00-fold [0.09]; P<0.01) and GluN2B protein amounts (0.38-fold [0.07] vs 1.00-fold [0.12]; P<0.01) in the anterior cingulate cortex through hypermethylation of Grin2b. The mu-opioid receptor antagonist naloxone and overexpression of Grin2b in anterior cingulate cortex attenuated the fentanyl-induced effects, whereas DAMGO injection into the anterior cingulate cortex induced autism-like behaviours. CONCLUSIONS These data suggest that fentanyl induces autism-like behaviours in young mice via an epigenetic mechanism. Further research is required to determine possible clinical relevance to autism risk.
Collapse
Affiliation(s)
- Zhihao Sheng
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qidong Liu
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chun Cheng
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mengzhu Li
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jed Barash
- Department of Medicine, Soldiers' Home, Chelsea, MA, USA
| | - W Andrew Kofke
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Shen
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
49
|
Zhu R, Zeng S, Li N, Fu N, Wang Y, Miao M, Yang Y, Sun M, Zhang J. Sevoflurane exposure induces neurotoxicity by regulating mitochondrial function of microglia due to NAD insufficiency. Front Cell Neurosci 2022; 16:914957. [PMID: 36212689 PMCID: PMC9532507 DOI: 10.3389/fncel.2022.914957] [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: 04/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Developmental neurons received with sevoflurane, the commonly used inhalational anesthetic agent in clinical surgery, several times tend to be destroyed. Microglia, the resident immune cells of the central nervous system (CNS), are activated after sevoflurane exposure, accompanied by releasing proinflammatory cytokines that damage developing neurons. The sevoflurane-induced neurotoxicity could be attributed to activated microglia presenting proinflammatory and anti-inflammatory functions. Proinflammatory microglia release cytokines to impair the CNS, while anti-inflammatory microglia engulf damaged neurons to maintain CNS homeostasis. Sevoflurane exposure promotes the secretion of proinflammatory cytokines by microglia, inhibiting the microglial phagocytic function. Microglia with poor phagocytic function cannot engulf damaged neurons, leading to the accumulation of damaged neurons. The mechanism underlying poor phagocytic function may be attributed to mitochondrial dysfunction of microglia induced by sevoflurane exposure, in which affected mitochondria cannot generate adequate ATP and NAD to satisfy the energy demand. We discovered that sevoflurane treatment impaired the mitochondrial metabolism of microglia, which resulted in NAD deficiency and couldn’t produce sufficient energy to clear damaged neurons to maintain CNS development. Our findings provide an explanation of a new mechanism underlying sevoflurane-induced neurotoxicity.
Collapse
|
50
|
Shang J, Li B, Fan H, Liu P, Zhao W, Chen T, Chen P, Yang L. Sevoflurane promotes premature differentiation of dopaminergic neurons in hiPSC-derived midbrain organoids. Front Cell Dev Biol 2022; 10:941984. [PMID: 36176283 PMCID: PMC9513420 DOI: 10.3389/fcell.2022.941984] [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: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Conventional animal models used in corresponding basic studies are distinct from humans in terms of the brain’s development trajectory, tissue cytoarchitecture and cell types, making it difficult to accurately evaluate the potential adverse effects of anesthetic treatments on human fetal brain development. This study investigated the effects of sevoflurane on the midbrain’s development and cytopathology using human physiologically-relevant midbrain organoids. Methods: Monolayer human induced pluripotent stem cells (hiPSC)-derived human floor plate cells and three-dimensional hiPSC-derived midbrain organoids (hMBOs) were exposed to 2% (v/v) sevoflurane for 2 or 6 h, followed by expansion or differentiation culture. Then, immunofluorescence, real-time PCR, EdU assay, Tunnel assay, and transcriptome sequencing were performed to examine the effects of sevoflurane on the midbrain’s development. Results: We found that 2% sevoflurane exposure inhibited hFPCs’ proliferation (differentiation culture: 7.2% ± 0.3% VS. 13.3% ± 0.7%, p = 0.0043; expansion culture: 48% ± 2.2% VS. 35.2% ± 1.4%, p = 0.0002) and increased their apoptosis, but did not affect their differentiation into human dopaminergic neurons After 6 h, 2% sevoflurane exposure inhibited cell proliferation (62.8% ± 5.6% VS. 100% ± 5.5%, p = 0.0065) and enhanced the premature differentiation of hMBOs (246% ± 5.2% VS. 100% ± 28%, p = 0.0065). The RNA-seq results showed long-term exposure to sevoflurane up regulates some transcription factors in the differentiation of dopaminergic neurons, while short-term exposure to sevoflurane has a weak up-regulation effect on these transcription factors. Conclusion: This study revealed that long-term exposure to sevoflurane could promote the premature differentiation of hMBOs, while short-term exposure had negligible effects, suggesting that long-term exposure to sevoflurane in pregnant women may lead to fetals’ midbrain development disorder.
Collapse
Affiliation(s)
- Jia Shang
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| | - Bin Li
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Han Fan
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Peidi Liu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Wen Zhao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Tao Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei, China
- *Correspondence: Longqiu Yang, ; Pu Chen,
| | - Longqiu Yang
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
- Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- *Correspondence: Longqiu Yang, ; Pu Chen,
| |
Collapse
|