1
|
Wang G, Liu HY, Meng XW, Chen Y, Zhao WM, Li WT, Xu HB, Peng K, Ji FH. Complement C1q-mediated microglial synaptic elimination by enhancing desialylation underlies sevoflurane-induced developmental neurotoxicity. Cell Biosci 2024; 14:42. [PMID: 38556890 PMCID: PMC10983687 DOI: 10.1186/s13578-024-01223-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Repeated neonatal sevoflurane exposures led to neurocognitive disorders in young mice. We aimed to assess the role of microglia and complement C1q in sevoflurane-induced neurotoxicity and explore the underlying mechanisms. METHODS Neonatal mice were treated with sevoflurane on postnatal days 6, 8, and 10, and the Morris water maze was performed to assess cognitive functions. For mechanistic explorations, mice were treated with minocycline, C1q-antibody ANX005, and sialidase-inhibitor N-acetyl-2,3-dehydro-2-deoxyneuraminic acid (NADNA) before sevoflurane exposures. Western blotting, RT-qPCR, Golgi staining, 3D reconstruction and engulfment analysis, immunofluorescence, and microglial morphology analysis were performed. In vitro experiments were conducted in microglial cell line BV2 cells. RESULTS Repeated neonatal sevoflurane exposures resulted in deficiencies in learning and cognition of young mice, accompanied by microglial activation and synapse loss. Sevoflurane enhanced microglia-mediated synapse elimination through C1q binding to synapses. Inhibition of microglial activation and phagocytosis with minocycline significantly reduced the loss of synapses. We further revealed the involvement of neuronal sialic acids in this process. The enhanced activity of sialidase by sevoflurane led to the loss of sialic acids, which facilitated C1q binding to synapses. Inhibition of C1q with ANX005 or inhibition of sialidase with NADNA significantly rescued microglia-mediated synapse loss and improved neurocognitive function. Sevoflurane enhanced the engulfment of BV2 cells, which was reversed by ANX005. CONCLUSIONS Our findings demonstrated that C1q-mediated microglial synaptic elimination by enhancing desialylation contributed to sevoflurane-induced developmental neurotoxicity. Inhibition of C1q or sialidase may be a potential therapeutic strategy for this neurotoxicity.
Collapse
Affiliation(s)
- Gang Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
- Ambulatory Surgery Center, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Ying Chen
- Departments of Neurology, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wei-Ming Zhao
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wen-Ting Li
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Han-Bing Xu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
2
|
Zhang H, Niu Y, Qiu L, Yang J, Sun J, Xia J. Melatonin-mediated mitophagy protects against long-term impairments after repeated neonatal sevoflurane exposures. Int Immunopharmacol 2023; 125:111210. [PMID: 37976600 DOI: 10.1016/j.intimp.2023.111210] [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: 08/23/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Melatonin is known to have protective effects in aging, neurodegenerative disorders and mitochondria-related diseases, while there is a poor understanding of the effects of melatonin treatment on mitophagy in neonatal cognitive dysfunction after repeated sevoflurane exposures. This study explores the protective effects of melatonin on mitophagy and cognition in developing rats exposed to sevoflurane. METHODS Postnatal day six (P6) neonatal rats were exposed to 3 % sevoflurane for 2 h daily from P6 to P8. In the intervention groups, rats received 3-Methyladenine (3-MA) intracerebroventricularly from P6 to P8 and melatonin intraperitoneally from P6 to P8 following water drinking once daily from P21 to P41, respectively. Behavioral tests, including open field (OF), novel object recognition (NOR), and fear conditioning (FC) tests, were performed to assess cognitive function during young adulthood. In another experiment, rat brains were harvested for biochemical, histopathological, and electron microscopy studies. RESULTS Rats exposed to sevoflurane showed disordered mitophagy and mitochondrial dysfunction as revealed by increased mitophagy marker proteins (microtubule-associated protein 1 light chain 3 (LC3) II/I, and parkin), decreased autophagy marker protein (sequestosome 1 (P62/SQSTM1)), electron transport chain (ETC) proteins and ATP levels. Immunofluorescent staining of LC3 was co-localized mostly with a neuronal marker and microglial marker but was not co-localized with a marker for astrocytes in rats exposed to sevoflurane. These rats had poorer performance in the NOR and FC tests than control rats during young adulthood. Melatonin treatment reversed the abnormal expression of mitophagy proteins, mitochondrial energy metabolism, the activity of microglia, and impaired cognition. These ameliorations were blocked by an autophagy inhibitor, 3-MA, except for the activation of microglia. CONCLUSION We have demonstrated that melatonin inhibits microglial activation by enhancing mitophagy and finally significantly reduces sevoflurane-induced deficits in cognition in neonatal rats. These results suggest that melatonin might be beneficial if considered when the anesthesia must be administered at a very young age.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yingqiao Niu
- Department of Anesthesiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Lili Qiu
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Jiaojiao Yang
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Jiangyan Xia
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
3
|
Mousavi S, Qiu H, Heinis FI, Bredahl EC, Ridwan Abid MS, Clifton AD, Andrews MT, Checco JW. Effects of Anesthetic Administration on Rat Hypothalamus and Cerebral Cortex Peptidome. ACS Chem Neurosci 2023; 14:3986-3992. [PMID: 37879091 PMCID: PMC10872895 DOI: 10.1021/acschemneuro.3c00499] [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] [Indexed: 10/27/2023] Open
Abstract
Prohormone-derived neuropeptides act as cell-cell signaling molecules to mediate a wide variety of biological processes in the animal brain. Mass spectrometry-based peptidomic experiments are valuable approaches to gain insight into the dynamics of individual peptides under different physiological conditions or experimental treatments. However, the use of anesthetics during animal procedures may confound experimental peptide measurements, especially in the brain, where anesthetics act. Here, we investigated the effects of the commonly used anesthetics isoflurane and sodium pentobarbital on the peptide profile in the rodent hypothalamus and cerebral cortex, as assessed by label-free quantitative peptidomics. Our results showed that neither anesthetic dramatically alters peptide levels, although extended isoflurane exposure did cause changes in a small number of prohormone-derived peptides in the cerebral cortex. Overall, our results demonstrate that acute anesthetic administration can be utilized in peptidomic experiments of the hypothalamus and cerebral cortex without greatly affecting the measured peptide profiles.
Collapse
Affiliation(s)
- Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Frazer I. Heinis
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Eric C. Bredahl
- Department of Exercise Science and Pre-Health Professions, Creighton University, Omaha, NE 68178, United States
| | - Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Ashley D. Clifton
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Matthew T. Andrews
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Mousavi S, Qiu H, Heinis FI, Abid MSR, Andrews MT, Checco JW. Short-Term Administration of Common Anesthetics Does Not Dramatically Change the Endogenous Peptide Profile in the Rat Pituitary. ACS Chem Neurosci 2022; 13:2888-2896. [PMID: 36126283 PMCID: PMC9547841 DOI: 10.1021/acschemneuro.2c00359] [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] [Indexed: 01/20/2023] Open
Abstract
Cell-cell signaling peptides (e.g., peptide hormones, neuropeptides) are among the largest class of cellular transmitters and regulate a variety of physiological processes. To identify and quantify the relative abundances of cell-cell signaling peptides in different physiological states, liquid chromatography-mass spectrometry-based peptidomics workflows are commonly utilized on freshly dissected tissues. In such animal experiments, the administration of general anesthetics is an important step for many research projects. However, acute anesthetic administration may rapidly change the measured abundance of transmitter molecules and metabolites, especially in the brain and endocrine system, which would confound experimental results. The aim of this study was to evaluate the effect of short-term (<5 min) anesthetic administration on the measured abundance of cell-cell signaling peptides, as evaluated by a typical peptidomics workflow. To accomplish this goal, we compared endogenous peptide abundances in the rat pituitary following administration of 5% isoflurane, 200 mg/kg sodium pentobarbital, or no anesthetic administration. Label-free peptidomics analysis demonstrated that acute use of isoflurane changed the levels of a small number of peptides, primarily degradation products of the hormone somatotropin, but did not influence the levels of most other peptide hormones. Acute use of sodium pentobarbital had negligible impact on the relative abundance of all measured peptides. Overall, our results suggest that anesthetics used in pituitary peptidomics studies do not dramatically confound observed results.
Collapse
Affiliation(s)
- Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Frazer I. Heinis
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Matthew T. Andrews
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| |
Collapse
|
7
|
Shu L, Du C. PHLDA1 promotes sevoflurane-induced pyroptosis of neuronal cells in developing rats through TRAF6-mediated activation of Rac1. Neurotoxicology 2022; 93:140-151. [PMID: 36155068 DOI: 10.1016/j.neuro.2022.09.007] [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: 06/16/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Sevoflurane anesthesia induces neurocognitive impairment and pyroptosis in the developing brain. Pleckstrin homology-like domain, family A, member 1 (PHLDA1) was involved in neuronal apoptosis, oxidative stress and inflammation during ischemic stroke. The role of PHLDA1 in sevoflurane-induced pyroptosis in developing rats was investigated. Firstly, neonatal rats at day 7 was exposed to 2.0% sevoflurane for 6 h to induce neurotoxicity. Pathological analysis showed that sevoflurane anesthesia induced hippocampal injury and reduced the number of neurons. The expression of PHLDA1 was elevated in hippocampus of sevoflurane-treated rats. Secondly, sevoflurane anesthesia-treated neonatal rats were injected with adeno-associated virus serotype (AAV) to mediate knockdown of PHLDA1. Injection with AAV-shPHLDA1 ameliorated sevoflurane-induced hippocampal injury and neurocognitive impairment in rats. Moreover, knockdown of PHLDA1 increased the number of neurons in sevoflurane-treated rats. Silence of PHLDA1 suppressed neuronal apoptosis, and inhibited pyroptosis in sevoflurane-treated rats. Thirdly, PHLDA1 was also elevated in sevoflurane-treated primary neuronal cells. Loss of PHLDA1 also enhanced cell viability and suppressed pyroptosis of sevoflurane-treated primary neuronal cells. Lastly, silence of PHLDA1 reduced protein expression of TRAF6 and p-Rac1 in sevoflurane-treated rats and neuronal cells. Over-expression of TRAF6 attenuated PHLDA1 silence-induced increase of cell viability and decreased pyroptosis in neuronal cells. In conclusion, loss of PHLDA1 protected against sevoflurane-induced pyroptosis in developing rats through inhibition of TRAF6-mediated activation of Rac1.
Collapse
Affiliation(s)
- Lijuan Shu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of ICU, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chunfu Du
- Department of Neurosurgery, Ya'an people's Hospital, Sichuan, Ya'an, Sichuan 625000, China.
| |
Collapse
|
8
|
Li Y, Li J, Shi Y, Zhou X, Feng W, Han L, Ma D, Jiang H, Yuan Y. Urinary Aromatic Amino Acid Metabolites Associated With Postoperative Emergence Agitation in Paediatric Patients After General Anaesthesia: Urine Metabolomics Study. Front Pharmacol 2022; 13:932776. [PMID: 35928271 PMCID: PMC9343964 DOI: 10.3389/fphar.2022.932776] [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: 04/30/2022] [Accepted: 06/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Emergence agitation (EA) is very common in paediatric patients during recovery from general anaesthesia, but underlying mechanisms remain unknown. This prospective study was designed to profile preoperative urine metabolites and identify potential biomarkers that can predict the occurrence of EA. Methods: A total of 224 patients were screened for recruitment; of those, preoperative morning urine samples from 33 paediatric patients with EA and 33 non-EA gender- and age-matched patients after being given sevoflurane general anaesthesia were analysed by ultra-high-performance liquid chromatography (UHPLC) coupled with a Q Exactive Plus mass spectrometer. Univariate analysis and orthogonal projection to latent structures squares-discriminant analysis (OPLS-DA) were used to analyse these metabolites. The least absolute shrinkage and selection operator (LASSO) regression was used to identify predictive variables. The predictive model was evaluated through the receiver operating characteristic (ROC) analysis and then further assessed with 10-fold cross-validation. Results: Seventy-seven patients completed the study, of which 33 (42.9%) patients developed EA. EA and non-EA patients had many differences in preoperative urine metabolic profiling. Sixteen metabolites including nine aromatic amino acid metabolites, acylcarnitines, pyridoxamine, porphobilinogen, 7-methylxanthine, and 5′-methylthioadenosine were found associated with an increased risk of EA, and they all exhibited higher levels in the EA group than in the non-EA group. The main metabolic pathways involved in these metabolic changes included phenylalanine, tyrosine and tryptophan metabolisms. Among these potential biomarkers, L-tyrosine had the best predictive value with an odds ratio (OR) (95% CI) of 5.27 (2.20–12.63) and the AUC value of 0.81 (0.70–0.91) and was robust with internal 10-fold cross-validation. Conclusion: Urinary aromatic amino acid metabolites are closely associated with EA in paediatric patients, and further validation with larger cohorts and mechanistic studies is needed. Clinical Trial Registration:clinicaltrials.gov, identifier NCT04807998
Collapse
Affiliation(s)
- Yueyue Li
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jingjie Li
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhuan Shi
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuhui Zhou
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqing Feng
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Han
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Hong Jiang
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Hong Jiang, ; Yongfang Yuan,
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- *Correspondence: Hong Jiang, ; Yongfang Yuan,
| |
Collapse
|
9
|
Yang H, Zhao L, Li Q. Echinacoside alleviates sevoflurane-induced cognitive dysfunction by activating FOXO1-mediated autophagy. Int J Dev Neurosci 2022; 82:339-348. [PMID: 35362638 DOI: 10.1002/jdn.10183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 11/07/2022] Open
Abstract
The current study aimed to examine the effects of echinacoside on cognitive impairment in mice after exposure to sevoflurane. To examine the role of FOXO1, si-FOXO1 and si-con were injected into the hippocampus through the left lateral cerebral ventricles. Sevoflurane-induced mice had serious cognitive dysfunction. However, pretreatment with echinacoside alleviated the cognitive dysfunction, as measured by a shortened escape latency time, and increased platform crossing times, the percentage of distance in the target quadrant and Y-maze spontaneous alternations. In addition, we found that echinacoside elevated FOXO1 expression in the hippocampus, increased the expression of autophagy-related proteins including Beclin 1, ATG5, ATG7 and LC3, and reduced P62 expression. Silencing of FOXO1 aggravated the cognitive deficits and reduced expression of the autophagy-related markers, while the effects of si-FOXO1 on memory were abrogated by echinacoside. Echinacoside attenuated the cognitive impairment in sevoflurane-induced mice through FOXO1-mediated autophagy.
Collapse
Affiliation(s)
- Huifang Yang
- Department of Anesthesia, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Zhao
- Department of Anesthesia, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qin Li
- Department of Respiration, Hebei Children's Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
10
|
Wang CM, Chen WC, Zhang Y, Lin S, He HF. Update on the Mechanism and Treatment of Sevoflurane-Induced Postoperative Cognitive Dysfunction. Front Aging Neurosci 2021; 13:702231. [PMID: 34305576 PMCID: PMC8296910 DOI: 10.3389/fnagi.2021.702231] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Sevoflurane is one of the most widely used anesthetics for the induction and maintenance of general anesthesia in surgical patients. Sevoflurane treatment may increase the incidence of postoperative cognitive dysfunction (POCD), and patients with POCD exhibit lower cognitive abilities than before the operation. POCD affects the lives of patients and places an additional burden on patients and their families. Understanding the mechanism of sevoflurane-induced POCD may improve prevention and treatment of POCD. In this paper, we review the diagnosis of POCD, introduce animal models of POCD in clinical research, analyze the possible mechanisms of sevoflurane-induced POCD, and summarize advances in treatment for this condition.
Collapse
Affiliation(s)
- Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia.,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| |
Collapse
|
11
|
Chen X, Chen D, Li Q, Wu S, Pan J, Liao Y, Zheng X, Zeng W. Dexmedetomidine Alleviates Hypoxia-Induced Synaptic Loss and Cognitive Impairment via Inhibition of Microglial NOX2 Activation in the Hippocampus of Neonatal Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6643171. [PMID: 33628369 PMCID: PMC7895593 DOI: 10.1155/2021/6643171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Perinatal hypoxia is a universal cause of death and neurological deficits in neonates worldwide. Activation of microglial NADPH oxidase 2 (NOX2) leads to oxidative stress and neuroinflammation, which may contribute to hypoxic damage in the developing brain. Dexmedetomidine has been reported to exert potent neuroprotection in several neurological diseases, but the mechanism remains unclear. We investigated whether dexmedetomidine acts through microglial NOX2 to reduce neonatal hypoxic brain damage. METHODS The potential role of microglial NOX2 in dexmedetomidine-mediated alleviation of hypoxic damage was evaluated in cultured BV2 microglia and neonatal rats subjected to hypoxia. In vivo, neonatal rats received dexmedetomidine (25 μg/kg, i.p.) 30 min before or immediately after hypoxia (5% O2, 2 h). Apocynin-mediated NOX inhibition and lentivirus-mediated NOX2 overexpression were applied to further assess the involvement of microglial NOX2 activation. RESULTS Pre- or posttreatment with dexmedetomidine alleviated hypoxia-induced cognitive impairment, restored damaged synapses, and increased postsynaptic density-95 and synaptophysin protein expression following neonatal hypoxia. Importantly, dexmedetomidine treatment suppressed hypoxia-induced microglial NOX2 activation and subsequent oxidative stress and the neuroinflammatory response, as reflected by reduced 4-hydroxynonenal and ROS accumulation, and decreased nuclear NF-κB p65 and proinflammatory cytokine levels in cultured BV2 microglia and the developing hippocampus. In addition, treating primary hippocampal neurons with conditioned medium (CM) from hypoxia-activated BV2 microglia resulted in neuronal damage, which was alleviated by CM from dexmedetomidine-treated microglia. Moreover, the neuroprotective effect of dexmedetomidine was reversed in NOX2-overexpressing BV2 microglia and diminished in apocynin-pretreated neonatal rats. CONCLUSION Dexmedetomidine targets microglial NOX2 to reduce oxidative stress and neuroinflammation and subsequently protects against hippocampal synaptic loss following neonatal hypoxia.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Dongtai Chen
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiang Li
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shuyan Wu
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Jiahao Pan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yanling Liao
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|