1
|
Yang M, Bai M, Zhuang Y, Lu S, Ge Q, Li H, Deng Y, Wu H, Xu X, Niu F, Dong X, Zhang B, Liu B. High-dose dexamethasone regulates microglial polarization via the GR/JAK1/STAT3 signaling pathway after traumatic brain injury. Neural Regen Res 2025; 20:2611-2623. [PMID: 39314167 DOI: 10.4103/nrr.nrr-d-23-01772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00023/figure1/v/2024-11-05T132919Z/r/image-tiff Although microglial polarization and neuroinflammation are crucial cellular responses after traumatic brain injury, the fundamental regulatory and functional mechanisms remain insufficiently understood. As potent anti-inflammatory agents, the use of glucocorticoids in traumatic brain injury is still controversial, and their regulatory effects on microglial polarization are not yet known. In the present study, we sought to determine whether exacerbation of traumatic brain injury caused by high-dose dexamethasone is related to its regulatory effects on microglial polarization and its mechanisms of action. In vitro cultured BV2 cells and primary microglia and a controlled cortical impact mouse model were used to investigate the effects of dexamethasone on microglial polarization. Lipopolysaccharide, dexamethasone, RU486 (a glucocorticoid receptor antagonist), and ruxolitinib (a Janus kinase 1 antagonist) were administered. RNA-sequencing data obtained from a C57BL/6 mouse model of traumatic brain injury were used to identify potential targets of dexamethasone. The Morris water maze, quantitative reverse transcription-polymerase chain reaction, western blotting, immunofluorescence and confocal microscopy analysis, and TUNEL, Nissl, and Golgi staining were performed to investigate our hypothesis. High-throughput sequencing results showed that arginase 1, a marker of M2 microglia, was significantly downregulated in the dexamethasone group compared with the traumatic brain injury group at 3 days post-traumatic brain injury. Thus dexamethasone inhibited M1 and M2 microglia, with a more pronounced inhibitory effect on M2 microglia in vitro and in vivo . Glucocorticoid receptor plays an indispensable role in microglial polarization after dexamethasone treatment following traumatic brain injury. Additionally, glucocorticoid receptor activation increased the number of apoptotic cells and neuronal death, and also decreased the density of dendritic spines. A possible downstream receptor signaling mechanism is the GR/JAK1/STAT3 pathway. Overactivation of glucocorticoid receptor by high-dose dexamethasone reduced the expression of M2 microglia, which plays an anti-inflammatory role. In contrast, inhibiting the activation of glucocorticoid receptor reduced the number of apoptotic glia and neurons and decreased the loss of dendritic spines after traumatic brain injury. Dexamethasone may exert its neurotoxic effects by inhibiting M2 microglia through the GR/JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Mengshi Yang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Miao Bai
- Department of Neurology, The First Hospital of Tsinghua University, Beijing, China
| | - Yuan Zhuang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenghua Lu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qianqian Ge
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Deng
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongbin Wu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fei Niu
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xinlong Dong
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Center for Nerve Injury and Repair, Beijing Institute of Brain Disorders, China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
2
|
Ma C, Wang Y. BHLHE40 regulates microglia polarization after spinal cord injury via the NF-κB pathway. Brain Res Bull 2025; 220:111139. [PMID: 39586332 DOI: 10.1016/j.brainresbull.2024.111139] [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: 08/11/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Spinal cord injury (SCI) is a devastating disease characterized by neuroinflammation and irreversible neuronal loss. The basic helix-loop-helix family member e40 (Bhlhe40) is a stress-responsive transcription factor involved in the pathological process of inflammation. However, Bhlhe40 expression and its role in SCI are largely unknown. SCI rat models were established with an aneurysm clip and then the rats were injected with lentiviral Bhlhe40 shRNA to knock down Bhlhe40 expression. In vitro, BV2 microglia cells were stimulated with LPS and IFN-γ to promote M1 microglia polarization. The results showed that Bhlhe40 expression was significantly elevated in the injured spinal cord tissue. Bhlhe40 deficiency reduced neuroinflammation and neuronal loss, and then promoted the recovery of neurological function. Additionally, Bhlhe40 knockdown alleviated neuronal apoptosis by regulating microglia polarization. In our study, Bhlhe40 knockdown inhibited M1 microglia polarization and the secretion of pro-inflammatory factors (TNF-α, IL-1β, and IL-6). Meanwhile, the NF-κB pathway was inhibited after the Bhlhe40 knockdown in SCI rats. To further explore the functional role of Bhlhe40, we performed in vitro experiments. Bhlhe40 knockdown decreased M1 microglia polarization by inhibiting the NF-κB pathway. In conclusion, our study indicates that Bhlhe40 knockdown can alleviate the progression of SCI and its underlying mechanism in regulating macrophage polarization through the NF-κB pathway.
Collapse
Affiliation(s)
- Chao Ma
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Mei B, Yang X, Yang YY, Weng JT, Cao SD, Yang R, Xu G. Intraoperative Dexmedetomidine Infusion Improved Postoperative Sleep Quality and Melatonin Secretion in Patients Undergoing Elective Thoracoscopic Lung Surgery: A Prospective, Randomized Study. Nat Sci Sleep 2024; 16:2009-2020. [PMID: 39677826 PMCID: PMC11645892 DOI: 10.2147/nss.s491084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Background Dexmedetomidine has been reported to improve postoperative sleep quality. However, the underlying mechanism remains unclear. This study aimed to investigate the effect of intraoperative dexmedetomidine infusion on postoperative sleep quality and changes in melatonin secretion in older patients undergoing elective thoracoscopic lung surgery. Methods A total of 126 older patients were randomly divided into two groups: dexmedetomidine group (Group D), which received continuous dexmedetomidine infusion at 0.3-0.5 µg/(kg·h) combined with propofol during surgery, and propofol group (Group P), which received propofol alone. The primary outcome was the postoperative sleep quality on the first postoperative night, assessed by the Richards-Campbell Sleep Questionnaire (RCSQ). Secondary outcomes included sleep quality scores on the second and third postoperative nights, melatonin concentrations postoperatively, and the incidence of delirium on the first and seventh postoperative days (discharge day). Results On the first postoperative night, Group D had a higher sleep quality score compared to Group P (57±11.4 vs 53±10.3; [95% CI, 1.1 to 8.7];P = 0.012), with no difference between the groups on the second and third postoperative nights. There was no statistically significant difference in the preoperative and postoperative night 3 urine 6-SMT concentrations between the two groups (P > 0.05); however, Group D had significantly higher urine 6-SMT concentrations on postoperative nights 1 and 2 compared to Group P (27 (24, 30) vs 21 (17, 24); [95% CI, -8.56 to -4.73]; P = 0.000. 28 (25, 30) vs 26 (21, 27); [95% CI, -4.37 to -1.65]; P = 0.000). There was no significant difference in the incidence of postoperative delirium between the two groups (P=0.65). Conclusion Continuous intraoperative infusion of dexmedetomidine can effectively improve sleep quality during the first postoperative night by promoting melatonin secretion over the first two postoperative nights.
Collapse
Affiliation(s)
- Bin Mei
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Xiao Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Yue-yue Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Jun-tao Weng
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - San-dong Cao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Rui Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Guanghong Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| |
Collapse
|
4
|
Sharma KK, Chauhan B. Integrated dexmedetomidine-sevoflurane algorithm for anesthetic induction - A viable asset for neurosurgery. Surg Neurol Int 2024; 15:455. [PMID: 39777190 PMCID: PMC11704436 DOI: 10.25259/sni_934_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Affiliation(s)
- Kunal Kumar Sharma
- Department of Anesthesia, Neuroanesthesia Cell, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India
| | - Bharti Chauhan
- Department of Anesthesia and Critical Care, Neuroanesthesia Division Under, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
5
|
Suo Z, Xiao T, Qu Y, Zheng Y, Xu W, Zhou B, Yang J, Yu J, Zheng H, Ni C. Aged hippocampal single-cell atlas screening unveils disrupted neuroglial system in postoperative cognitive impairment. Aging Cell 2024:e14406. [PMID: 39540334 DOI: 10.1111/acel.14406] [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: 06/08/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Glia-neuron interaction is a crucial feature in aged hippocampus during the occurrence of postoperative cognitive impairment. However, the regulatory effects of microglia, astrocytes, and oligodendrocytes in this glia-neuron interaction, the potential mechanisms and gene targets are still to be elucidated. Here, single-cell RNA sequencing was performed to detect the perioperative genomic expression characteristics of neuroglial system in the hippocampus of aged mice, and to investigate the potential cross-cellular mechanisms and valuable treatment options for glia-neuron interaction-related cognitive impairment. We found that postoperative neurons and glia cells exhibited protein dysmetabolism and mitochondrial electron misrouting. Impaired autophagy and circadian rhythm worsened microglia activation/neuroinflammation, and exacerbated these metabolic alterations. Reactive microglia also aggravated astrocyte and oligodendrocyte cytotoxicity through the PGD2/DP and complement pathways, altering glutamate level and synaptic function via the "tripartite synapses" model, and affecting neuronal myelination. Ligand-receptor communication also indicated these synaptic and axonal dysfunctions via enhanced MDK and PTN pathways. Additionally, we found that anesthetic dexmedetomidine hold therapeutic potential within the disrupted neuroglial system. It enhanced neuronal metabolic rebalance (Atf3-related) and reduced neuroinflammation from a multicellular perspective, therefore improving postoperative cognitive impairment. Together, our study proposes an aged hippocampal cell atlas and provides insights into the role of disrupted glia-neuron cycle in postoperative cognitive impairment. Our findings also elucidate the therapeutic potential and mechanism of dexmedetomidine intervention.
Collapse
Affiliation(s)
- Zizheng Suo
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Xiao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinyin Qu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yuxiang Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjie Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bowen Zhou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jie Yu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Xue H, Ding Z, Chen X, Yang X, Jia Y, Zhao P, Wu Z. Dexmedetomidine Improves Long-term Neurological Outcomes by Promoting Oligodendrocyte Genesis and Myelination in Neonatal Rats Following Hypoxic-ischemic Brain Injury. Mol Neurobiol 2024:10.1007/s12035-024-04564-z. [PMID: 39496877 DOI: 10.1007/s12035-024-04564-z] [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: 07/19/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024]
Abstract
Neonatal hypoxic-ischemic brain injury (HIBI) can lead to white matter damage, which significantly contributes to cognitive dysfunction, emotional disorders, and sensorimotor impairments. Although dexmedetomidine enhances neurobehavioral outcomes, its impact on oligodendrocyte genesis and myelination following hypoxic-ischemic events, as well as the underlying mechanisms, remain poorly understood. Dexmedetomidine was administered 15 min post-HIBI. We assessed neurobehavioral deficits using various tests: surface righting, negative geotaxis, forelimb grip strength, cliff avoidance, sensory reflexes, novel object recognition, T-maze, and three-chamber social interaction. We also investigated the relationship between myelination and neurobehavioral outcomes. Measurements included oligodendrocyte precursor cell (OPC) proliferation and survival 24 h post-injury, early myelination, and oligodendrocyte differentiation by postnatal day 14. Furthermore, we evaluated microglial activation towards the M2 phenotype and the extent of neuroinflammation during the acute phase. Dexmedetomidine significantly ameliorated long-term neurological deficits caused by HIBI. Pearson linear regression analysis revealed a strong correlation between long-term neurological outcomes and myelin maturity. The treatment notably mitigated the long-term deterioration of myelin formation and maturation following HIBI. This protective effect was primarily due to enhanced OPC proliferation and survival post-HIBI during the acute phase and, to a lesser extent, to the modulation of microglial activity towards the M2 phenotype and a reduction in neuroinflammation. Dexmedetomidine offers substantial protection against long-term neurobehavioral disabilities induced by HIBI, primarily by revitalizing the impaired survival and maturation of oligodendrocyte progenitor cells and promoting myelination.
Collapse
Affiliation(s)
- Hang Xue
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zixuan Ding
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xiaoyan Chen
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xu Yang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yufei Jia
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
7
|
Tan Y, Wang Q, Guo Y, Zhang N, Xu Y, Bai X, Liu J, Bi X. Dexmedetomidine mitigates lidocaine-induced spinal cord injury by repressing ferritinophagy-mediated ferroptosis by increasing CISD2 expression in rat models. J Bioenerg Biomembr 2024; 56:517-530. [PMID: 39168950 PMCID: PMC11455791 DOI: 10.1007/s10863-024-10034-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: 05/08/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Dexmedetomidine (DEX) has been confirmed to exert neuroprotective effects in various nerve injury models by regulating ferroptosis, including spinal cord injury (SCI). Although it has been established that CDGSH iron sulfur domain 2 (CISD2) can regulate ferroptosis, whether DEX can regulate ferroptosis by CISD2 in SCI remains unclear. Lidocaine was used to induce PC12 cells and stimulate rats to establish SCI models in vitro and in vivo. MTT assays were performed to analyze cell viability. Ferroptosis was assessed by determining the levels of cellular reactive axygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and Fe2+. Ferritinophagy was analyzed by LysoTracker staining, FerroOrange staining, and immunofluorescence. Western blotting was carried out to quantify the levels of several proteins. Fluorescence microscopy was also used to observe cell autophagy. The morphology of mitochondria within the tissue was observed under transmission electron microscopy (TEM). DEX treatment weakened lidocaine-induced elevation of ROS, Fe2+, and MDA and reduced GSH in PC12 cells, indicating that DEX treatment weakened lidocaine-induced ferroptosis in PC12 cells. Similarly, lidocaine promoted autophagy, Fe2+, and microtubule-associated protein 1 light chain 3 (LC3) in PC12 cells and suppressed ferritin and p62 protein levels, indicating that DEX could weaken lidocaine-induced ferritinophagy in PC12 cells. DEX treatment improved the BBB score, reduced tissue damage, increased the number of neurons, and alleviated mitochondrial damage by inhibiting ferroptosis and ferritinophagy in lidocaine-induced SCI rat models. The decreased CISD2, ferritin heavy chain 1 (FTH1), solute carrier family 7-member 11-glutathione (SLC7A11), and glutathione peroxidase 4 (GPX4) protein levels and the elevated nuclear receptor coactivator 4 (NCOA4) protein levels in rat models in the lidocaine group were weakened by DEX treatment. Moreover, CISD2 inhibition reversed the inhibitory effects of DEX treatment on lidocaine-induced ferroptosis and ferritinophagy in PC12 cells significantly. Taken together, DEX treatment could impair lidocaine-induced SCI by inhibiting ferroptosis and ferritinophagy by upregulating CISD2 in rat models.
Collapse
Affiliation(s)
- Yonghong Tan
- Department of Anesthesiology, Liuzhou Hospital, Guangzhou Women and Children's Medical Center, No. 50 Boyuan Avenue, Yufeng District, Liuzhou, Guangxi, 545000, China
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qiong Wang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yubing Guo
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Na Zhang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yingyi Xu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xue Bai
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jianhua Liu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaobao Bi
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
8
|
Then J, Tawfik S, Law T, Brown A, Carnegie V, Udy A, Jeffcote T. The Impact of Sedative Choice in the Management of Aneurysmal Subarachnoid Hemorrhage: A Scoping Review. Neurocrit Care 2024:10.1007/s12028-024-02111-1. [PMID: 39266867 DOI: 10.1007/s12028-024-02111-1] [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: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is characterized by high mortality and morbidity. This scoping review assesses the current evidence regarding the use of sedatives and analgesics in the acute intensive care unit management of aSAH. We conducted a systematic search of Ovid MEDLINE, Ovid Embase, Ovid EmCare, APA PsycInfo, CINAHL, and the Cochrane Database of Systematic Reviews from inception to June 2023. Studies were included if they enrolled intensive care unit patients aged 18 or older with a significant proportion (> 20%) who had aSAH and evaluated the impact of one or more commonly used analgosedatives on physiological parameters in the management of aSAH. The methodological quality of the studies was assessed using the Methodological Index for Nonrandomized Studies score. Of 2,583 articles, 11 met the inclusion criteria. The median sample size was 47 (interquartile range 10-127), and the median Methodological Index for Nonrandomized Studies score was 9.5 (interquartile range 8-11). The studies' publication years ranged from 1980 to 2023. Dexmedetomidine and ketamine showed potential benefits in reducing the incidence of cortical spreading depolarization and delayed cerebral ischemia. Propofol and opioids appeared safe but lacked robust evidence for efficacy. Benzodiazepines were associated with increased delayed cerebral ischemia-related cerebral infarctions and cortical spreading depolarization events. The evidence available to guide the use of analgosedative medications in aSAH is critically inadequate. Dexmedetomidine and ketamine warrant further exploration in large-scale prospective studies because of their potential benefits. Improved study designs with consistent definitions and a focus on patient-centered outcomes are necessary to inform clinical practice.
Collapse
Affiliation(s)
- James Then
- Department of Intensive Care, The Royal Melbourne Hospital, Melbourne, VIC, Australia.
| | - Samuel Tawfik
- The Victorian Brain and Spine Centre, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Timothy Law
- Department of Intensive Care, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Alastair Brown
- Department of Intensive Care, St. Vincent's Hospital, Melbourne, VIC, Australia
- Australia and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Vanessa Carnegie
- Department of Intensive Care, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Andrew Udy
- Department of Intensive Care, The Alfred Hospital, Melbourne, VIC, Australia
| | - Toby Jeffcote
- Department of Intensive Care, The Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Zhang Y, Xiao S, Dan F, Yao G, Hong S, Liu J, Liu Z. Phillygenin inhibits neuroinflammation and promotes functional recovery after spinal cord injury via TLR4 inhibition of the NF-κB signaling pathway. J Orthop Translat 2024; 48:133-145. [PMID: 39220679 PMCID: PMC11363727 DOI: 10.1016/j.jot.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Background Spinal cord injuries (SCIs) trigger a cascade of detrimental processes, encompassing neuroinflammation and oxidative stress (OS), ultimately leading to neuronal damage. Phillygenin (PHI), isolated from forsythia, is used in a number of biomedical applications, and is known to exhibit anti-neuroinflammation activity. In this study, we investigated the role and mechanistic ability of PHI in the activation of microglia-mediated neuroinflammation and subsequent neuronal apoptosis following SCI. Methods A rat model of SCI was used to investigate the impact of PHI on inflammation, axonal regeneration, neuronal apoptosis, and the restoration of motor function. In vitro, neuroinflammation models were induced by stimulating microglia with lipopolysaccharide (LPS); then, we investigated the influence of PHI on pro-inflammatory mediator release in LPS-treated microglia along with the underlying mechanisms. Finally, we established a co-culture system, featuring microglia and VSC 4.1 cells, to investigate the role of PHI in the activation of microglia-mediated neuronal apoptosis. Results In vivo, PHI significantly inhibited the inflammatory response and neuronal apoptosis while enhancing axonal regeneration and improving motor function recovery. In vitro, PHI inhibited the release of inflammation-related factors from polarized BV2 cells in a dose-dependent manner. The online Swiss Target Prediction database predicted that toll-like receptor 4 (TLR4) was the target protein for PHI. In addition, Molecular Operating Environment software was used to perform molecular docking for PHI with the TLR4 protein; this resulted in a binding energy interaction of -6.7 kcal/mol. PHI inhibited microglia-mediated neuroinflammation, the production of reactive oxygen species (ROS), and activity of the NF-κb signaling pathway. PHI also increased mitochondrial membrane potential (MMP) in VSC 4.1 neuronal cells. In BV2 cells, PHI attenuated the overexpression of TLR4-induced microglial polarization and significantly suppressed the release of inflammatory cytokines. Conclusion PHI ameliorated SCI-induced neuroinflammation by modulating the TLR4/MYD88/NF-κB signaling pathway. PHI has the potential to be administered as a treatment for SCI and represents a novel candidate drug for addressing neuroinflammation mediated by microglial cells. The translational potential of this article We demonstrated that PHI is a potential drug candidate for the therapeutic management of SCI with promising developmental and translational applications.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
| | - Shining Xiao
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fan Dan
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
| | - Geliang Yao
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
| | - Shu'e Hong
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jiaming Liu
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
| | - Zhili Liu
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, China
| |
Collapse
|
10
|
Wongsripuemtet P, Ohnuma T, Temkin N, Barber J, Komisarow J, Manley GT, Hatfield J, Treggiari M, Colton K, Sasannejad C, Chaikittisilpa N, Ivins-O'Keefe K, Grandhi R, Laskowitz D, Mathew JP, Hernandez A, James ML, Raghunathan K, Miller J, Vavilala M, Krishnamoorthy V. Association of early dexmedetomidine exposure with brain injury biomarker levels following moderate - Severe traumatic brain injury: A TRACK-TBI study. J Clin Neurosci 2024; 126:338-347. [PMID: 39029302 DOI: 10.1016/j.jocn.2024.07.003] [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: 03/27/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) triggers autonomic dysfunction and inflammatory response that can result in secondary brain injuries. Dexmedetomidine is an alpha-2 agonist that may modulate autonomic function and inflammation and has been increasingly used as a sedative agent for critically ill TBI patients. We aimed to investigate the association between early dexmedetomidine exposure and blood-based biomarker levels in moderate-to-severe TBI (msTBI). METHODS We conducted a retrospective cohort study using data from the Transforming Clinical Research and Knowledge in Traumatic Brain Injury Study (TRACK-TBI), which enrolled acute TBI patients prospectively across 18 United States Level 1 trauma centers between 2014-2018. Our study population focused on adults with msTBI defined by Glasgow Coma Scale score 3-12 after resuscitation, who required mechanical ventilation and sedation within the first 48 h of ICU admission. The study's exposure was early dexmedetomidine utilization (within the first 48 h of admission). Primary outcome included brain injury biomarker levels measured from circulating blood on day 3 following injury, including glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase-L1 (UCH-L1), neuron-specific enolase (NSE), S100 calcium-binding protein B (S100B) and the inflammatory biomarker C-reactive protein (CRP). Secondary outcomes assessed biomarker levels on days 5 and 14. Linear mixed-effects regression modelling of the log-transformed response variable was used to analyze the association of early dexmedetomidine exposure with brain injury biomarker levels. RESULTS Among the 352 TRACK-TBI subjects that met inclusion criteria, 50 (14.2 %) were exposed to early dexmedetomidine, predominantly male (78 %), white (81 %), and non-Hispanic (81 %), with mean age of 39.8 years. Motor vehicle collisions (27 %) and falls (22 %) were common causes of injury. No significant associations were found between early dexmedetomidine exposure with day 3 brain injury biomarker levels (GFAP, ratio = 1.46, 95 % confidence interval [0.90, 2.34], P = 0.12; UCH-L1; ratio = 1.17 [0.89, 1.53], P = 0.26; NSE, ratio = 1.19 [0.92, 1.53], P = 0.19; S100B, ratio = 1.01 [0.95, 1.06], P = 0.82; hs-CRP, ratio = 1.29 [0.91, 1.83], P = 0.15). The hs-CRP level at day 14 in the dexmedetomidine group was higher than that of the non-exposure group (ratio = 1.62 [1.12, 2.35], P = 0.012). CONCLUSIONS There were no significant associations between early dexmedetomidine exposure and day 3 brain injury biomarkers in msTBI. Our findings suggest that early dexmedetomidine use is not correlated with either decrease or increase in brain injury biomarkers following msTBI. Further research is necessary to confirm these findings.
Collapse
Affiliation(s)
- Pattrapun Wongsripuemtet
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Tetsu Ohnuma
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Nancy Temkin
- Department of Biostatistics, University of Washington, Seattle, WA, United States; Department of Neurosurgery, University of Washington, Seattle, WA, United States
| | - Jason Barber
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Jordan Komisarow
- Department of Neurosurgery, Duke University, Durham, NC, United States
| | - Geoffrey T Manley
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, United States
| | - Jordan Hatfield
- Department of Neurosurgery, Duke University, Durham, NC, United States; Duke University School of Medicine, Durham, NC, United States
| | - Miriam Treggiari
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Katharine Colton
- Department of Neurology, Duke University, Durham, NC, United States
| | - Cina Sasannejad
- Department of Neurology, Duke University, Durham, NC, United States
| | - Nophanan Chaikittisilpa
- Department of Anesthesiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kelly Ivins-O'Keefe
- Department of Anesthesiology, Duke University, Durham, NC, United States; Duke University School of Medicine, Durham, NC, United States
| | - Ramesh Grandhi
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, United States
| | - Daniel Laskowitz
- Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Neurosurgery, Duke University, Durham, NC, United States; Department of Neurology, Duke University, Durham, NC, United States
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Adrian Hernandez
- Department of Medicine, Duke University, Durham, NC, United States
| | - Michael L James
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Neurology, Duke University, Durham, NC, United States
| | - Karthik Raghunathan
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Population Health Sciences, Duke University, Durham, NC, United States
| | - Joseph Miller
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI, United States
| | - Monica Vavilala
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| | - Vijay Krishnamoorthy
- Critical Care and Perioperative Population Health Research (CAPER) Program, Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Anesthesiology, Duke University, Durham, NC, United States; Department of Population Health Sciences, Duke University, Durham, NC, United States
| |
Collapse
|
11
|
Wang X, Hong CG, Duan R, Pang ZL, Zhang MN, Xie H, Liu ZZ. Transplantation of olfactory mucosa mesenchymal stromal cells repairs spinal cord injury by inducing microglial polarization. Spinal Cord 2024; 62:429-439. [PMID: 38849489 DOI: 10.1038/s41393-024-01004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
STUDY DESIGN Animal studies OBJECTIVES: To evaluate the therapeutic effect of olfactory mucosa mesenchymal stem cell (OM-MSCs) transplantation in mice with spinal cord injury (SCI) and to explore the mechanism by which OM-MSCs inhibit neuroinflammation and improve SCI. SETTING Xiangya Hospital, Central South University; Affiliated Hospital of Guangdong Medical University. METHODS Mice (C57BL/6, female, 6-week-old) were randomly divided into sham, SCI, and SCI + OM-MSC groups. The SCI mouse model was generated using Allen's method. OM-MSCs were immediately delivered to the lateral ventricle after SCI using stereotaxic brain injections. One day prior to injury and on days 1, 5, 7, 14, 21, and 28 post-injury, the Basso Mouse Scale and Rivlin inclined plate tests were performed. Inflammation and microglial polarization were evaluated using histological staining, immunofluorescence, and qRT-PCR. RESULTS OM-MSCs originating from the neuroectoderm have great potential in the management of SCI owing to their immunomodulatory effects. OM-MSCs administration improved motor function, alleviated inflammation, promoted the transformation of the M1 phenotype of microglia into the M2 phenotype, facilitated axonal regeneration, and relieved spinal cord injury in SCI mice. CONCLUSIONS OM-MSCs reduced the level of inflammation in the spinal cord tissue, protected neurons, and repaired spinal cord injury by regulating the M1/M2 polarization of microglia.
Collapse
Affiliation(s)
- Xin Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chun-Gu Hong
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ran Duan
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhi-Lin Pang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Min-Na Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hui Xie
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zheng-Zhao Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
12
|
Bao D, Wang Y, Xiong W, Zhang D, Qiao L, Zheng N, Li L, Jin X. Intraoperative application of low-dose dexmedetomidine or lidocaine for postoperative analgesia in pediatric patients following craniotomy: a randomized double-blind placebo-controlled trial. Front Surg 2024; 11:1371588. [PMID: 38978991 PMCID: PMC11228272 DOI: 10.3389/fsurg.2024.1371588] [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: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 07/10/2024] Open
Abstract
Background Postoperative pain is a common occurrence in pediatric patients following craniotomy, often leading to negative outcomes. Intravenous dexmedetomidine and lidocaine are commonly used adjuvant medicines in general anesthesia to reduce perioperative opioid consumption and relieve postoperative pain in adults. While they show promise for use in pediatrics, the evidence of their application in pediatric craniotomy patients is limited. Therefore, we aimed to compare the effects of dexmedetomidine and lidocaine on postoperative pain in pediatric patients following craniotomy. Methods We conducted a randomized, double-blind, single-center trial on children scheduled for craniotomy. The 255 recruited participants aged 1-12 years were randomly assigned to intraoperatively receive a loading intravenous dose of either dexmedetomidine 1 μg·kg-1 or lidocaine 2 mg·kg-1 or normal saline for 15 min followed by dexmedetomidine 0.5 μg·kg-1·h-1 or lidocaine 1 mg·kg-1·h-1 or normal saline until the sutures of endocranium were completed. The primary outcome was the cumulative sufentanil consumption within 24 h post-surgery. Results A total of 241 patients were included in the statistical analysis. The primary outcome did not show any significant differences among the three groups (median (IQR) lidocaine group: 3.36 (1.32-5.64) μg vs. dexmedetomidine group: 3.12 (1.36-6.39) μg vs. control group 3.46 (1.77-7.62) μg, p = 0.485). Among the secondary outcomes, there was a statistically significant but small reduction in sufentanil consumption within 2 h, postoperative FLACC/WBFS/NRS pain scores within 4 h after surgery and postoperative Ramsay sedation scores in dexmedetomidine group (p < 0.05). Regarding postoperative complications, the incidence of electrolyte disturbance within 24 and 48 h after surgery was significantly higher in control group compared to the other two groups. There were no significant differences in intraoperative opioid consumption, postoperative frequency of remedy medication, or length of hospitalization among the three groups. No adverse events related to lidocaine or dexmedetomidine were observed. Conclusions There were no significant differences in the primary outcome among the three groups. Although dexmedetomidine showed some benefits in reducing postoperative opioid consumption within the first 2 h and pain intensity within the first 4 h post-surgery, these findings should be interpreted with caution. Further research is required to comprehensively assess the outcomes and determine the optimal administration strategy. Clinical Trial Registration [http://www.chictr.org.cn/index.aspx], identifier [ChiCTR1800019411].
Collapse
Affiliation(s)
- Di Bao
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaxin Wang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Xiong
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Di Zhang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lanxin Qiao
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Na Zheng
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lu Li
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xu Jin
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Feng H, Hu X, Lin Y, Xiao J, Dai C, Hu Z, Feng H, Qin J, Chen L. Dexmedetomidine attenuates acute stress-impaired learning and memory in mice by maintaining the homeostasis of intestinal flora. Eur J Med Res 2024; 29:271. [PMID: 38711117 DOI: 10.1186/s40001-024-01832-5] [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: 01/07/2024] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Dexmedetomidine (Dex) has been used in surgery to improve patients' postoperative cognitive function. However, the role of Dex in stress-induced anxiety-like behaviors and cognitive impairment is still unclear. In this study, we tested the role of Dex in anxiety-like behavior and cognitive impairment induced by acute restrictive stress and analyzed the alterations of the intestinal flora to explore the possible mechanism. Behavioral and cognitive tests, including open field test, elevated plus-maze test, novel object recognition test, and Barnes maze test, were performed. Intestinal gut Microbe 16S rRNA sequencing was analyzed. We found that intraperitoneal injection of Dex significantly improved acute restrictive stress-induced anxiety-like behavior, recognition, and memory impairment. After habituation in the environment, mice (male, 8 weeks, 18-23 g) were randomly divided into a control group (control, N = 10), dexmedetomidine group (Dex, N = 10), AS with normal saline group (AS + NS, N = 10) and AS with dexmedetomidine group (AS + Dex, N = 10). By the analysis of intestinal flora, we found that acute stress caused intestinal flora disorder in mice. Dex intervention changed the composition of the intestinal flora of acute stress mice, stabilized the ecology of the intestinal flora, and significantly increased the levels of Blautia (A genus of anaerobic bacteria) and Coprobacillus. These findings suggest that Dex attenuates acute stress-impaired learning and memory in mice by maintaining the homeostasis of intestinal flora.
Collapse
Affiliation(s)
- Hao Feng
- Department of Dermatology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410000, People's Republic of China
| | - Xing Hu
- Department of Dermatology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410000, People's Republic of China
| | - Yizi Lin
- Department of Radiology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325200, People's Republic of China
| | - Jingni Xiao
- Department of Nephrology, Hengyang Medical School, University of South China Affiliated Changsha Central Hospital, No. 161 Shaoshan South Road, Changsha, Hunan, 410004, People's Republic of China
| | - Chao Dai
- Department of Nephrology, Hengyang Medical School, University of South China Affiliated Changsha Central Hospital, No. 161 Shaoshan South Road, Changsha, Hunan, 410004, People's Republic of China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, Hunan, 410011, People's Republic of China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Jiao Qin
- Department of Nephrology, Hengyang Medical School, University of South China Affiliated Changsha Central Hospital, No. 161 Shaoshan South Road, Changsha, Hunan, 410004, People's Republic of China.
| | - Li Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Wenzhou Medical University, No.108 Wansong Road, Wenzhou, Zhejiang, 325200, People's Republic of China.
| |
Collapse
|
14
|
Pan JZ, Wang Z, Sun W, Pan P, Li W, Sun Y, Chen S, Lin A, Tan W, He L, Greene J, Yao V, An L, Liang R, Li Q, Yu J, Zhang L, Kyritsis N, Fernandez XD, Moncivais S, Mendoza E, Fung P, Wang G, Niu X, Du Q, Xiao Z, Chang Y, Lv P, Huie JR, Torres‐Espin A, Ferguson AR, Hemmerle DD, Talbott JF, Weinstein PR, Pascual LU, Singh V, DiGiorgio AM, Saigal R, Whetstone WD, Manley GT, Dhall SS, Bresnahan JC, Maze M, Jiang X, Singhal NS, Beattie MS, Su H, Guan Z. ATF3 is a neuron-specific biomarker for spinal cord injury and ischaemic stroke. Clin Transl Med 2024; 14:e1650. [PMID: 38649772 PMCID: PMC11035380 DOI: 10.1002/ctm2.1650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Although many molecules have been investigated as biomarkers for spinal cord injury (SCI) or ischemic stroke, none of them are specifically induced in central nervous system (CNS) neurons following injuries with low baseline expression. However, neuronal injury constitutes a major pathology associated with SCI or stroke and strongly correlates with neurological outcomes. Biomarkers characterized by low baseline expression and specific induction in neurons post-injury are likely to better correlate with injury severity and recovery, demonstrating higher sensitivity and specificity for CNS injuries compared to non-neuronal markers or pan-neuronal markers with constitutive expressions. METHODS In animal studies, young adult wildtype and global Atf3 knockout mice underwent unilateral cervical 5 (C5) SCI or permanent distal middle cerebral artery occlusion (pMCAO). Gene expression was assessed using RNA-sequencing and qRT-PCR, while protein expression was detected through immunostaining. Serum ATF3 levels in animal models and clinical human samples were measured using commercially available enzyme-linked immune-sorbent assay (ELISA) kits. RESULTS Activating transcription factor 3 (ATF3), a molecular marker for injured dorsal root ganglion sensory neurons in the peripheral nervous system, was not expressed in spinal cord or cortex of naïve mice but was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Additionally, ATF3 protein levels in mouse blood significantly increased 1 day after SCI or ischemic stroke. Importantly, ATF3 protein levels in human serum were elevated in clinical patients within 24 hours after SCI or ischemic stroke. Moreover, Atf3 knockout mice, compared to the wildtype mice, exhibited worse neurological outcomes and larger damage regions after SCI or ischemic stroke, indicating that ATF3 has a neuroprotective function. CONCLUSIONS ATF3 is an easily measurable, neuron-specific biomarker for clinical SCI and ischemic stroke, with neuroprotective properties. HIGHLIGHTS ATF3 was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Serum ATF3 protein levels are elevated in clinical patients within 24 hours after SCI or ischemic stroke. ATF3 exhibits neuroprotective properties, as evidenced by the worse neurological outcomes and larger damage regions observed in Atf3 knockout mice compared to wildtype mice following SCI or ischemic stroke.
Collapse
Affiliation(s)
- Jonathan Z. Pan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhanqiang Wang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyCangzhou People's HospitalCangzhouChina
| | - Wei Sun
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Peipei Pan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Wei Li
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Yongtao Sun
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyQianfoshan Hospital, Shandong UniversityJinanChina
| | - Shoulin Chen
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyThe Second Affiliated Hospital, Nanchang UniversityNanchangChina
| | - Amity Lin
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Wulin Tan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyGuangzhou Medical UniversityGuangzhouChina
| | - Liangliang He
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Pain ManagementXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jacob Greene
- Medical SchoolUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Virginia Yao
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lijun An
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyNo. 1 People's HospitalHuaianChina
| | - Rich Liang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Qifeng Li
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
| | - Jessica Yu
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Nikolaos Kyritsis
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Xuan Duong Fernandez
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sara Moncivais
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Esmeralda Mendoza
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Pamela Fung
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Gongming Wang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Xinhuan Niu
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Qihang Du
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Zhaoyang Xiao
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyThe Second Affiliated Hospital, Dalian Medical UniversityDalianChina
| | - Yuwen Chang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Peiyuan Lv
- Department of AnesthesiologyThe Second Affiliated Hospital, Dalian Medical UniversityDalianChina
- Department of NeurologyHebei Medical UniversityShijiazhuangChina
| | - J. Russell Huie
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Abel Torres‐Espin
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Adam R. Ferguson
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Debra D. Hemmerle
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jason F. Talbott
- Department of RadiologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Philip R. Weinstein
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lisa U. Pascual
- Department of Orthopedic SurgeryOrthopaedic Trauma InstituteUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Vineeta Singh
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Anthony M. DiGiorgio
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Rajiv Saigal
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - William D. Whetstone
- Department of Emergency MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Geoffrey T. Manley
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sanjay S. Dhall
- Department of NeurosurgeryHarbor UCLA Medical CenterTorranceCaliforniaUSA
| | - Jacqueline C. Bresnahan
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Mervyn Maze
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Xiangning Jiang
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Neel S. Singhal
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Michael S. Beattie
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Hua Su
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
15
|
Zhu C, Cao D, Chen L, Zheng X, Lin D. The effect of dexmedetomidine on postoperative wound healing in neurosurgical patients: A meta-analysis. Int Wound J 2024; 21:e14585. [PMID: 38148721 PMCID: PMC10961872 DOI: 10.1111/iwj.14585] [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: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023] Open
Abstract
This meta-analysis investigates the effect of dexmedetomidine on postoperative wound healing in neurosurgical patients. A thorough literature search resulted in the selection of seven studies from an initial pool of 1546 records. The analysis focused on wound healing outcomes, evaluated by the Redness, Oedema, Ecchymosis, Discharge, Approximation (REEDA) scale and the Manchester Scar Scale (MSS). Results indicated significant improvements in the dexmedetomidine group: the REEDA scale scores at day seven post-surgery showed a Standardized Mean Difference group (SMD = -16.18, 95% CI: [-22.30, -10.06], p < 0.01), and the MSS scores at 3 months post-operation demonstrated an (SMD = -8.95, 95% CI: [-14.27, -3.62], p < 0.01). These findings suggest that dexmedetomidine may enhance wound healing and reduce scar formation in neurosurgical patients. Bias assessment indicated a low risk of bias across the studies.
Collapse
Affiliation(s)
- Congying Zhu
- Department of AnesthesiologyTaizhou Hospital Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Donghang Cao
- Department of AnesthesiologyTaizhou Hospital Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Lingyang Chen
- Department of AnesthesiologyTaizhou Hospital Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Xuhang Zheng
- Department of AnesthesiologyTaizhou Hospital Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Danjiang Lin
- Department of AnesthesiologyTaizhou Hospital Affiliated to Wenzhou Medical UniversityLinhaiChina
| |
Collapse
|
16
|
Mei B, Xu X, Weng J, Yang Y, Wang P, Qiu G, Zhang C, Zhang Q, Lu Y, Liu X. Activating astrocytic α2A adrenoceptors in hippocampus reduces glutamate toxicity to attenuate sepsis-associated encephalopathy in mice. Brain Behav Immun 2024; 117:376-398. [PMID: 38320682 DOI: 10.1016/j.bbi.2024.02.005] [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: 09/09/2023] [Revised: 12/09/2023] [Accepted: 02/02/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Glutamate metabolism disorder is an important mechanism of sepsis-associated encephalopathy (SAE). Astrocytes regulate glutamate metabolism. In septic mice, α2A adrenoceptor (α2A-AR) activation in the central nervous system provides neuroprotection. α2A-ARs are expressed abundantly in hippocampal astrocytes. This study was performed to determine whether hippocampal astrocytic α2A-AR activation confers neuroprotection against SAE and whether this protective effect is astrocyte specific and achieved by the modulation of glutamate metabolism. METHODS Male C57BL/6 mice with and without α2A-AR knockdown were subjected to cecal ligation and puncture (CLP). They were treated with intrahippocampal guanfacine (an α2A-AR agonist) or intraperitoneal dexmedetomidine in the presence or absence of dihydrokainic acid [DHK; a glutamate transporter 1 (GLT-1) antagonist] and/or UCPH-101 [a glutamate/aspartate transporter (GLAST) antagonist]. Hippocampal tissue was collected for the measurement of astrocyte reactivity, GLT-1 and GLAST expression, and glutamate receptor subunit 2B (GluN2B) phosphorylation. In vivo real-time extracellular glutamate concentrations in the hippocampus were measured by ultra-performance liquid chromatography tandem mass spectrometry combined with microdialysis, and in vivo real-time hippocampal glutamatergic neuron excitability was assessed by calcium imaging. The mice were subjected to the Barnes maze and fear conditioning tests to assess their learning and memory. Golgi staining was performed to assess changes in the hippocampal synaptic structure. In vitro, primary astrocytes with and without α2A-AR knockdown were stimulated with lipopolysaccharide (LPS) and treated with guanfacine or dexmedetomidine in the presence or absence of 8-bromo- cyclic adenosine monophosphate (8-Br-cAMP, a cAMP analog). LPS-treated primary and BV2 microglia were also treated with guanfacine or dexmedetomidine. Astrocyte reactivity, PKA catalytic subunit, GLT-1 an GLAST expression were determined in primary astrocytes. Interleukin-1β, interleukin-6 and tumor necrosis factor-alpha in the medium of microglia culture were measured. RESULTS CLP induced synaptic injury, impaired neurocognitive function, increased astrocyte reactivity and reduced GLT-1 and GLAST expression in the hippocampus of mice. The extracellular glutamate concentration, phosphorylation of GluN2B at Tyr-1472 and glutamatergic neuron excitability in the hippocampus were increased in the hippocampus of septic mice. Intraperitoneal dexmedetomidine or intrahippocampal guanfacine administration attenuated these effects. Hippocampal astrocytes expressed abundant α2A-ARs; expression was also detected in neurons but not microglia. Specific knockdown of α2A-ARs in hippocampal astrocytes and simultaneous intrahippocampal DHK and UCPH-101 administration blocked the neuroprotective effects of dexmedetomidine and guanfacine. Intrahippocampal administration of DHK or UCPH-101 alone had no such effect. In vitro, guanfacine or dexmedetomidine inhibited astrocyte reactivity, reduced PKA catalytic subunit expression, and increased GLT-1 and GLAST expression in primary astrocytes but not in primary astrocytes that received α2A-AR knockdown or were treated with 8-Br-cAMP. Guanfacine or dexmedetomidine inhibited microglial reactivity in BV2 but not primary microglia. CONCLUSIONS Our results suggest that neurocognitive protection against SAE after hippocampal α2A-AR activation is astrocyte specific. This protection may involve the inhibition of astrocyte reactivity and alleviation of glutamate neurotoxicity, thereby reducing synaptic injury. The cAMP/protein kinase A (PKA) signaling pathway is a potential cellular mechanism by which activating α2A-AR modulates astrocytic function.
Collapse
Affiliation(s)
- Bin Mei
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China.
| | - Xiaoxia Xu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Juntao Weng
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Yueyue Yang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Peng Wang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Gaolin Qiu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Chi Zhang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, 230001, China
| | - Qunlin Zhang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, 230001, China
| | - Yao Lu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Xuesheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
17
|
Diao S, Wang D, Chang Y, Ni C, Fu D, Liu J, Gao S, Jia X, Wang T, Nan X, Cao H, Liu Z, Zhang X. Clinical study of the combined use of dexmedetomidine and remifentanil in patients with coronary heart disease undergoing 3D laparoscopic surgery with EEG bispectral index monitoring. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2023; 16:378-385. [PMID: 38188350 PMCID: PMC10767478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/03/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE This study sought to investigate the safety and clinical outcomes associated with the combined administration of dexmedetomidine (Dex) and remifentanil (Rem) in patients with coronary heart disease undergoing three-dimensional (3D) laparoscopic surgery, with concurrent monitoring of the electroencephalography (EEG) bispectral index. METHODS This study is of a retrospective nature and involved a total of 60 patients with coronary heart disease who underwent 3D laparoscopic surgery at our hospital between June 2020 and September 2021. In a double-blind manner, these patients were randomly assigned to two groups: the control group (Group I), which consisted of 30 patients, and the treatment group (Group II) receiving a combination of Dex and Rem, also comprising 30 patients. The study's primary objective was to compare and assess the treatment outcomes in these two patient groups. RESULTS Patients in Group II who developed postoperative coronary heart disease experienced a significant reduction in blood pressure, heart rate, and electrocardiogram values (P<0.05). Additionally, Group II exhibited lower bispectral index (BIS) and visual analog scale (VAS) values (P<0.05). CONCLUSION In patients with coronary heart disease undergoing 3D laparoscopic surgery, the intraoperative use of Dex combined with Rem anesthesia offers several advantages. It helps stabilize hemodynamics, reducing the risk of myocardial ischemia, and significantly alleviates postoperative pain, all without increasing the likelihood of adverse postoperative reactions. Furthermore, this approach effectively dampens the intraoperative and postoperative stress response, facilitating enhanced recovery after surgery (ERAS). Overall, the clinical impact is positive, safe, and reliable.
Collapse
Affiliation(s)
- Shiqi Diao
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Dongxin Wang
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Ying Chang
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Chunyan Ni
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Dongmei Fu
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Jixin Liu
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Song Gao
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Xiunan Jia
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Tongrao Wang
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Xi Nan
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Hongling Cao
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Zongming Liu
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| | - Xitong Zhang
- Department of Anesthesiology, Jilin Cancer Hospital Changchun 130000, Jilin, P. R. China
| |
Collapse
|
18
|
Gurnani PK, Barlow B, Boling B, Busse LW, Diaz-Gomez JL, Ford J, Gibson GA, Khanna AK, Lee JS, Rivosecchi RM, Spezzano KM, Thornton N, Vallabhajosyula S, Witenko CJ, Wieruszewski PM. Major Publications in the Critical Care Pharmacotherapy Literature: 2022. Crit Care Explor 2023; 5:e0981. [PMID: 37753239 PMCID: PMC10519466 DOI: 10.1097/cce.0000000000000981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
OBJECTIVES A number of trials related to critical care pharmacotherapy were published in 2022. We aimed to summarize the most influential publications related to the pharmacotherapeutic care of critically ill patients in 2022. DATA SOURCES PubMed/Medical Literature Analysis and Retrieval System Online and the Clinical Pharmacy and Pharmacology Pharmacotherapy Literature Update. STUDY SELECTION Randomized controlled trials, prospective studies, or systematic review/meta-analyses of adult critically ill patients assessing a pharmacotherapeutic intervention and reporting clinical endpoints published between January 1, 2022, and December 31, 2022, were included in this article. DATA EXTRACTION Articles from a systematic search and the Clinical Pharmacy and Pharmacology Pharmacotherapy Literature Update were included and stratified into clinical domains based upon consistent themes. Consensus was obtained on the most influential publication within each clinical domain utilizing an a priori defined three-round modified Delphi process with the following considerations: 1) overall contribution to scientific knowledge and 2) novelty to the literature. DATA SYNTHESIS The systematic search and Clinical Pharmacy and Pharmacology Pharmacotherapy Literature Update yielded a total of 704 articles, of which 660 were excluded. The remaining 44 articles were stratified into the following clinical domains: emergency/neurology, cardiovascular, gastroenterology/fluids/nutrition, hematology, infectious diseases/immunomodulation, and endocrine/metabolic. The final article selected from each clinical domain was summarized following a three-round modified Delphi process and included three randomized controlled trials and three systematic review/meta-analyses. Article topics summarized included dexmedetomidine versus other sedatives during mechanical ventilation, beta-blocker treatment in the critically ill, restriction of IV fluids in septic shock, venous thromboembolism prophylaxis in critically ill adults, duration of antibiotic therapy for Pseudomonas aeruginosa ventilator-associated pneumonia, and low-dose methylprednisolone treatment in severe community-acquired pneumonia. CONCLUSIONS This concise review provides a perspective on articles published in 2022 that are relevant to the pharmacotherapeutic care of critically ill patients and their potential impact on clinical practice.
Collapse
Affiliation(s)
- Payal K Gurnani
- Department of Pharmacy, Memorial Hermann The Woodlands Medical Center, The Woodlands, TX
| | - Brooke Barlow
- Department of Pharmacy, Memorial Hermann The Woodlands Medical Center, The Woodlands, TX
| | - Bryan Boling
- Department of Anesthesiology, Division of Critical Care Medicine, University of Kentucky, Lexington, KY
| | | | - Jose L Diaz-Gomez
- Department of Anesthesiology and Critical Care Medicine, Texas Heart Institute, Baylor College of Medicine, Houston, TX
| | - Jenna Ford
- Department of Neurology, University of Florida, Gainesville, FL
| | | | - Ashish K Khanna
- Department of Anesthesiology, Section of Critical Care Medicine, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC
| | | | | | | | - Nathan Thornton
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Saraschandra Vallabhajosyula
- Department of Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Corey J Witenko
- Department of Pharmacy, New York-Presbyterian Hospital, New York, NY
| | | |
Collapse
|
19
|
Zhang S, Zhang Y, Zheng Y, Zhu S, Sun J, Deng Y, Wang Q, Zhai Q. Dexmedetomidine attenuates sleep deprivation-induced inhibition of hippocampal neurogenesis via VEGF-VEGFR2 signaling and inhibits neuroinflammation. Biomed Pharmacother 2023; 165:115085. [PMID: 37392656 DOI: 10.1016/j.biopha.2023.115085] [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: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023] Open
Abstract
Long periods of sleep deprivation (SD) have serious effects on health. While the α2 adrenoceptor agonist dexmedetomidine (DEX) can improve sleep quality for patients who have insomnia, the effect of DEX on cognition and mechanisms after SD remains elusive. C57BL/6 mice were subjected to 20 h SD daily for seven days. DEX (100 μg/kg) was administered intravenously twice daily (at 1:00 p.m. and 3:00 p.m.) during seven days of SD. We found that systemic administration of DEX attenuated cognitive deficits by performing the Y maze and novel object recognition tests and increased DCX+, SOX2+, Ki67+, and BrdU+NeuN+/NeuN+ cell numbers in the dentate gyrus (DG) region of SD mice by using immunofluorescence, western blotting, and BrdU staining. DEX did not reverse the decrease in DCX+, SOX2+, or Ki67+ cell numbers in SD mice after administration of the α2A-adrenoceptor antagonist BRL-44408. Furthermore, the vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) expression was upregulated in SD+DEX mice compared with SD mice. Luminex analysis showed that the neurogenic effects of DEX were possibly related to the inhibition of neuroinflammation, including IL-1α, IL-2, CCL5, and CXCL1. Our results suggested that DEX alleviated the impaired learning and memory of SD mice potentially by inducing hippocampal neurogenesis via the VEGF-VEGFR2 signaling pathway and by suppressing neuroinflammation, and α2A adrenoceptors are required for the neurogenic effects of DEX after SD. This novel mechanism may add to our knowledge of DEX in the clinical treatment of impaired memory caused by SD.
Collapse
Affiliation(s)
- Shuyue Zhang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ying Zhang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yige Zheng
- The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Shan Zhu
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jianyu Sun
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yingying Deng
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Qian Zhai
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
20
|
Qiao L, Li G, Yuan HX. Dexmedetomidine mediates the mechanism of action of ferroptosis in mice with Alzheimer's disease by regulating the mTOR-TFR1 pathway. World J Psychiatry 2023; 13:511-523. [PMID: 37701546 PMCID: PMC10494775 DOI: 10.5498/wjp.v13.i8.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/06/2023] [Accepted: 07/27/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disorder, and there are currently no effective drugs to delay progression of the disease. Ferroptosis may play a vital part in AD, and is therefore receiving increasing attention by researchers. AIM To investigate the effects of dexmedetomidine (Dex) on ferroptosis in AD mouse hippocampus. METHODS Hippocampal neurons (HNs) HT22 were induced by amyloid β-protein (Aβ) and both in vitro and in vivo AD mouse models were prepared via injections. The cell-counting kit-8 assay and immunofluorescence technique were adopted to determine cell proliferation activity and intracellular Fe2+ levels, and the TBA method and microplate method were employed for malondialdehyde and glutathione measurements, respectively. Hippocampal tissue damage was determined using hematoxylin and eosin and Nissl staining. Mouse learning and memory ability in each group was assessed by the Morris water maze test, and the expression levels of mammalian target of rapamycin (mTOR) signal molecules and ferroptosis-related proteins transferrin receptor 1 (TFR1), SLC7A11 and glutathione peroxidase 4 were examined by western blotting. RESULTS Dex enhanced lipid peroxidation and iron influx in mouse HNs in both in vitro and in vivo experiments, while inhibition of the mTOR axis blocked this process. These findings demonstrate that Dex can inhibit ferroptosis-induced damage in mouse HNs by activating mTOR-TFR1 signaling to regulate ferroptosis-associated proteins, thus alleviating cognitive dysfunction in AD mice. CONCLUSION Dex can activate the mTOR-TFR1 axis to inhibit ferroptosis in mouse HNs, thereby improving the learning and memory ability of mice.
Collapse
Affiliation(s)
- Li Qiao
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Gang Li
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Hong-Xun Yuan
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| |
Collapse
|
21
|
Cao S, Wu Y, Gao Z, Tang J, Xiong L, Hu J, Li C. Automated phenotyping of postoperative delirium-like behaviour in mice reveals the therapeutic efficacy of dexmedetomidine. Commun Biol 2023; 6:807. [PMID: 37532767 PMCID: PMC10397202 DOI: 10.1038/s42003-023-05149-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023] Open
Abstract
Postoperative delirium (POD) is a complicated and harmful clinical syndrome. Traditional behaviour analysis mostly focuses on static parameters. However, animal behaviour is a bottom-up and hierarchical organizational structure composed of time-varying posture dynamics. Spontaneous and task-driven behaviours are used to conduct comprehensive profiling of behavioural data of various aspects of model animals. A machine-learning based method is used to assess the effect of dexmedetomidine. Fourteen statistically different spontaneous behaviours are used to distinguish the non-POD group from the POD group. In the task-driven behaviour, the non-POD group has greater deep versus shallow investigation preference, with no significant preference in the POD group. Hyperactive and hypoactive subtypes can be distinguished through pose evaluation. Dexmedetomidine at a dose of 25 μg kg-1 reduces the severity and incidence of POD. Here we propose a multi-scaled clustering analysis framework that includes pose, behaviour and action sequence evaluation. This may represent the hierarchical dynamics of delirium-like behaviours.
Collapse
Affiliation(s)
- Silu Cao
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434, China
| | - Yiling Wu
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Zilong Gao
- School of Life Sciences and Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, 310024, China
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Jinxuan Tang
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434, China
| | - Ji Hu
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Cheng Li
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434, China.
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434, China.
| |
Collapse
|
22
|
Zhou M, Luo Q, Xu Y. As an inhibitor of norepinephrine release, dexmedetomidine provides no improvement on stroke-associated pneumonia in mice. Front Pharmacol 2023; 14:1203646. [PMID: 37601052 PMCID: PMC10433391 DOI: 10.3389/fphar.2023.1203646] [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: 04/11/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Background: Dexmedetomidine (DEX) is commonly employed as a sedative agent to attenuate sympathetic tone and reduce norepinephrine (NE) levels. In the context of stroke-associated pneumonia (SAP), which is believed to arise from heightened sympathetic nervous system activity and elevated NE release, the precise influence of DEX remains uncertain. Methods: In this study, we generated an SAP model using middle cerebral artery occlusion (MCAO) and examined NE levels, immunological statuses in the brain and periphery, pneumonia symptoms, and extent of infarction. We aimed to determine the effects of DEX on SAP and explore the underlying. Despite its potential to reduce NE levels, DEX did not alleviate SAP symptoms or decrease the infarct area. Interestingly, DEX led to an increase in spleen size and spleen index. Furthermore, we observed a decrease in the CD3+ T cell population in both the blood and brain, but an increase in the spleen following DEX administration. The precise mechanism linking decreased CD3+ T cells and DEX's role in SAP requires further investigation. Conclusion: The clinical use of DEX in stroke patients should be approached with caution, considering its inability to alleviate SAP symptoms and reduce the infarct area. Further research is necessary to fully understand the relationship between decreased CD3+ T cells and DEX's influence on SAP.
Collapse
Affiliation(s)
- Miaomiao Zhou
- Anesthesiology Department, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiong Luo
- Anesthesiology Department, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Younian Xu
- Anesthesiology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Zhong Y, Wang S, Yin Y, Yu J, Liu Y, Gao H. Dexmedetomidine suppresses hippocampal astrocyte pyroptosis in cerebral hypoxic-ischemic neonatal rats by upregulating microRNA-148a-3p to inactivate the STAT/JMJD3 axis. Int Immunopharmacol 2023; 121:110440. [PMID: 37327511 DOI: 10.1016/j.intimp.2023.110440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Dexmedetomidine (DEX), a selective α2-adrenoceptor agonist, is an anesthetic and sedative agent and has been reported to confer neuroprotective effects after cerebral hypoxic ischemia (CHI). This study was undertaken to elucidate the mechanisms by which microRNA (miR)-148a-3p is involved in the neuroprotective effect of DEX on hypoxic-ischemic brain damage in neonatal rats. METHODS Neonatal rats were exposed to CHI conditions, a miR-148a-3p inhibitor, and DEX. Hippocampal astrocytes were isolated to construct an oxygen-glucose deprivation (OGD) model. qRT-PCR and western blot were utilized to inspect miR-148a-3p, STAT1, STAT3, JMJD3, cleaved-Caspase-1, ASC, NLRP3, GSDMD, and GSDMD-N expression in rats and astrocytes. TUNEL staining was employed to measure astrocyte apoptosis rate, immunofluorescence to inspect cleaved-Caspase-1 and ASC levels, and ELISA to determine IL-1β and IL-18 expression. The target genes of miR-148a-3p were predicted using online software and verified by a dual-luciferase reporter gene assay. RESULTS A prominent increase in astrocyte apoptosis rate and the expression of pyroptosis- and inflammation-related factors were found in rats with CHI and OGD-treated astrocytes. DEX suppressed astrocyte apoptosis rate and decreased expression of pyroptosis- and inflammation-related factors. Knockdown of miR-148a-3p facilitated astrocyte pyroptosis, indicating that DEX exerted its protective effect by upregulating miR-148a-3p. miR-148a-3p negatively mediated STAT to inactivate JMJD3. Overexpression of STAT1 and STAT3 facilitated pyroptosis in astrocytes, which was negated by the overexpression of miR-148a-3p. CONCLUSION DEX inhibited hippocampal astrocyte pyroptosis by upregulating miR-148a-3p to inactivate the STAT/JMJD3 axis, thereby alleviating cerebral damage in neonatal rats with CHI.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Shengzhao Wang
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yongqiang Yin
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Jialu Yu
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yang Liu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Hong Gao
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou 558000, PR China.
| |
Collapse
|
24
|
Zwolinski NM, Patel KS, Vadivelu N, Kodumudi G, Kaye AD. ERAS Protocol Options for Perioperative Pain Management of Substance Use Disorder in the Ambulatory Surgical Setting. Curr Pain Headache Rep 2023; 27:65-79. [PMID: 37079258 PMCID: PMC10116112 DOI: 10.1007/s11916-023-01108-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/21/2023]
Abstract
Even prior to the COVID-19 pandemic, rates of ambulatory surgeries and ambulatory patients presenting with substance use disorder were increasing, and the end of lockdown has further catalyzed the increasing rates of ambulatory patients presenting for surgery with substance use disorder (SUD). Certain subspecialty groups of ambulatory procedures have already established protocols to optimize early recovery after surgery (ERAS), and these groups have subsequently enjoyed improved efficiency and reduced adverse outcomes as a result. In this present investigation, we review the literature as it relates to substance use disorder patients, with a particular focus on pharmacokinetic and pharmacodynamic profiles, and their resulting impact on the acute- or chronic user ambulatory patient. The systematic literature review findings are organized and summarized. We conclude by identifying areas of opportunity for further study, specifically with the aim of developing a dedicated ERAS protocol for substance use disorder patients in the ambulatory surgery setting. - Healthcare in the USA has seen an increase in rates of both substance use disorder patients and separately in ambulatory surgery cases. - Specific perioperative protocols to optimize outcomes for patients who suffer from substance use disorder have been described in recent years. - Agents of interest like opioids, cannabis, and amphetamines are the top three most abused substances in North America. - A protocol and recommend further work should be done to integrate with concrete clinical data, in which strategies should be employed to confer benefits to patient outcomes and hospital quality metrics like those enjoyed by ERAS protocol in other settings.
Collapse
Affiliation(s)
- Nicholas M Zwolinski
- Department of Anesthesiology, Yale University School of Medicine, 333, Cedar Street, New Haven, CT, 06520, USA
| | - Kaiwal S Patel
- Department of Anesthesiology, Yale University School of Medicine, 333, Cedar Street, New Haven, CT, 06520, USA
| | - Nalini Vadivelu
- Department of Anesthesiology, Yale University School of Medicine, 333, Cedar Street, New Haven, CT, 06520, USA
| | - Gopal Kodumudi
- Department of Anesthesiology, LSU School of Medicine, 1542 Tulane Avenue Room 659, New Orleans, LA, 70112, USA
| | - Alan David Kaye
- Department of Anesthesiology, LSU School of Medicine, 1542 Tulane Avenue Room 659, New Orleans, LA, 70112, USA.
- Department of Anesthesiology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71103, USA.
| |
Collapse
|
25
|
Xu X, Zhuo L, Zhang L, Peng H, Lyu Y, Sun H, Zhai Y, Luo D, Wang X, Li X, Li L, Zhang Y, Ma X, Wang Q, Li Y. Dexmedetomidine alleviates host ADHD-like behaviors by reshaping the gut microbiota and reducing gut-brain inflammation. Psychiatry Res 2023; 323:115172. [PMID: 36958092 DOI: 10.1016/j.psychres.2023.115172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is one of the most prevalent psychiatric disorders that affects children and even continues into adulthood. Dexmedetomidine (DEX), a short-term sedative, can selectively activate the α2-adrenoceptor. Treatment with α2-adrenergic agonists in patients with ADHD is becoming increasingly common. However, the therapeutic potential of DEX for the treatment of ADHD is unknown. Here, we evaluated the effect of DEX on ADHD-like behavior in spontaneously hypertensive rats (SHRs), a widely used animal model of ADHD. DEX treatment ameliorated hyperactivity and spatial working memory deficits and normalized θ electroencephalogram (EEG) rhythms in SHRs. We also found that DEX treatment altered the gut microbiota composition and promoted the enrichment of beneficial gut bacterial genera associated with anti-inflammatory effects in SHRs. The gut pathological scores and permeability and the level of inflammation observed in the gut and brain were remarkably improved after DEX administration. Moreover, transplantation of fecal microbiota from DEX-treated SHRs produced effects that mimicked the therapeutic effects of DEX administration. Therefore, DEX is a promising treatment for ADHD that functions by reshaping the composition of the gut microbiota and reducing inflammation in the gut and brain.
Collapse
Affiliation(s)
- Xiangzhao Xu
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Anesthesiology, The People's Hospital of Nanchuan, Chongqing 408400, China
| | - Lixia Zhuo
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Linjuan Zhang
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Huan Peng
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yixuan Lyu
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Huan Sun
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yifang Zhai
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Danlei Luo
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaodan Wang
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xinyang Li
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Liya Li
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ying Zhang
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qiang Wang
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yan Li
- Department of Anesthesiology and Perioperative Medicine and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
26
|
Pilipović I, Stojić-Vukanić Z, Leposavić G. Adrenoceptors as potential target for add-on immunomodulatory therapy in multiple sclerosis. Pharmacol Ther 2023; 243:108358. [PMID: 36804434 DOI: 10.1016/j.pharmthera.2023.108358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
This review summarizes recent findings related to the role of the sympathetic nervous system (SNS) in pathogenesis of multiple sclerosis (MS) and its commonly used experimental model - experimental autoimmune encephalomyelitis (EAE). They indicate that noradrenaline, the key end-point mediator of the SNS, acting through β-adrenoceptor, has a contributory role in the early stages of MS/EAE development. This stage is characterized by the SNS hyperactivity (increased release of noradrenaline) reflecting the net effect of different factors, such as the disease-associated inflammation, stress, vitamin D hypovitaminosis, Epstein-Barr virus infection and dysbiosis. Thus, the administration of propranolol, a non-selective β-adrenoceptor blocker, readily crossing the blood-brain barrier, to experimental rats before the autoimmune challenge and in the early (preclinical/prodromal) phase of the disease mitigates EAE severity. This phenomenon has been ascribed to the alleviation of neuroinflammation (due to attenuation of primarily microglial activation/proinflammatory functions) and the diminution of the magnitude of the primary CD4+ T-cell autoimmune response (the effect associated with impaired autoantigen uptake by antigen presenting cells and their migration into draining lymph nodes). The former is partly related to breaking of the catecholamine-dependent self-amplifying microglial feed-forward loop and the positive feedback loop between microglia and the SNS, leading to down-regulation of the SNS hyperactivity and its enhancing influence on microglial activation/proinflammatory functions and the magnitude of autoimmune response. The effects of propranolol are shown to be more prominent in male EAE animals, the phenomenon important as males (like men) are likely to develop clinically more severe disease. Thus, these findings could serve as a firm scientific background for formulation of a new sex-specific immune-intervention strategy for the early phases of MS (characterized by the SNS hyperactivity) exploiting anti-(neuro)inflammatory and immunomodulatory properties of propranolol and other relatively cheap and safe adrenergic drugs with similar therapeutic profile.
Collapse
Affiliation(s)
- Ivan Pilipović
- Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- University of Belgrade-Faculty of Pharmacy, Department of Microbiology and Immunology, Belgrade, Serbia
| | - Gordana Leposavić
- University of Belgrade-Faculty of Pharmacy, Department of Pathobiology, Belgrade, Serbia.
| |
Collapse
|
27
|
Hong Z, Cheng J, Ye Y, Chen X, Zhang F. MicroRNA-451 Attenuates the Inflammatory Response of Activated Microglia by Downregulating Nucleotide Binding Oligomerization Domain-Like Receptor Protein 3. World Neurosurg 2022; 167:e1128-e1137. [PMID: 36087911 DOI: 10.1016/j.wneu.2022.08.139] [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: 04/18/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Spinal cord injury is the most common problem encountered during spinal surgery. After the initial trauma, the disruption of the blood-brain barrier and subsequent microglia activation result in extensive inflammatory responses. Inflammasomes are large protein complexes that are essential during inflammation. One of the most studied inflammasome components, nucleotide binding oligomerization domain-like receptor protein 3 (NLRP; nucleotide binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing 3), is widely expressed in the central nervous system. Previous research has shown that microRNA-451 (miR-451) might play a role in regulating inflammatory conditions. METHODS Using bioinformatics analysis, we found that NLRP3 is a direct target of miR-451. This in silico prediction was confirmed using dual-luciferase reporter gene assays. To further demonstrate that miR-451 influenced microglial NLRP3 production, we activated microglial cells with lipopolysaccharides. RESULTS Activating microglial cells with lipopolysaccharides resulted in the production of NLRP3 inflammasomes and the secretion of the proinflammatory cytokines interleukin-1β and interleukin-18. We were able to demonstrate that overexpression of miR-451 suppressed this NLRP3-induced proinflammatory cascade of events. CONCLUSIONS Our findings have highlighted the potential anti-inflammatory role of miR-451 in reducing the secondary neuronal damage after spinal cord injury.
Collapse
Affiliation(s)
- Zhou Hong
- Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaqi Cheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Ye
- Medical School of Nantong University, Nantong, China
| | - Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
28
|
Ge MM, Chen N, Zhou YQ, Yang H, Tian YK, Ye DW. Galectin-3 in Microglia-Mediated Neuroinflammation: Implications for Central Nervous System Diseases. Curr Neuropharmacol 2022; 20:2066-2080. [PMID: 35105290 PMCID: PMC9886847 DOI: 10.2174/1570159x20666220201094547] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/27/2021] [Accepted: 01/29/2022] [Indexed: 11/22/2022] Open
Abstract
Microglial activation is one of the common hallmarks shared by various central nervous system (CNS) diseases. Based on surrounding circumstances, activated microglia play either detrimental or neuroprotective effects. Galectin-3 (Gal-3), a group of β-galactoside-binding proteins, has been cumulatively revealed to be a crucial biomarker for microglial activation after injuries or diseases. In consideration of the important role of Gal-3 in the regulation of microglial activation, it might be a potential target for the treatment of CNS diseases. Recently, Gal-3 expression has been extensively investigated in numerous pathological processes as a mediator of neuroinflammation, as well as in cell proliferation. However, the underlying mechanisms of Gal-3 involved in microgliamediated neuroinflammation in various CNS diseases remain to be further investigated. Moreover, several clinical studies support that the levels of Gal-3 are increased in the serum or cerebrospinal fluid of patients with CNS diseases. Thus, we summarized the roles and underlying mechanisms of Gal-3 in activated microglia, thus providing a better insight into its complexity expression pattern, and contrasting functions in CNS diseases.
Collapse
Affiliation(s)
- Meng-Meng Ge
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Nan Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Hui Yang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; ,Address correspondence to these authors at the Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. E-mail: ., Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. E-mail:
| | - Da-Wei Ye
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; ,Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China,Address correspondence to these authors at the Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. E-mail: ., Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. E-mail:
| |
Collapse
|
29
|
Zhang Z, Mu X, Zhou X. Dexmedetomidine alleviates inflammatory response and oxidative stress injury of vascular smooth muscle cell via α2AR/GSK-3β/MKP-1/NRF2 axis in intracranial aneurysm. BMC Pharmacol Toxicol 2022; 23:81. [PMID: 36273189 PMCID: PMC9588221 DOI: 10.1186/s40360-022-00607-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 08/08/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) phenotypic modulation regulates the initiation and progression of intracranial aneurysm (IA). Dexmedetomidine (DEX) is suggested to play neuroprotective roles in patients with craniocerebral injury. Therefore, we investigated the biological functions of DEX and its mechanisms against IA formation and progression in the current study. The rat primary VSMCs were isolated from Sprague-Dawley rats. IA and superficial temporal artery (STA) tissue samples were obtained from patients with IA. Flow cytometry was conducted to identify the characteristics of isolated VSMCs. Hydrogen peroxide (H2O2) was used to mimic IA-like conditions in vitro. Cell viability was detected using CCK-8 assays. Wound healing and Transwell assays were performed to detect cell motility. ROS production was determined by immunofluorescence using DCFH-DA probes. Western blotting and RT-qPCR were carried out to measure gene expression levels. Inflammation responses were determined by measuring inflammatory cytokines. Immunohistochemistry staining was conducted to measure α2-adrenergic receptor levels in tissue samples. DEX alleviated the H2O2-induced cytotoxicity, attenuated the promoting effects of H2O2 on cell malignancy, and protected VSMCs against H2O2-induced oxidative damage and inflammation response. DEX regulated the GSK-3β/MKP-1/NRF2 pathway via the α2AR. DEX alleviates the inflammatory responses and oxidative damage of VSMCs by regulating the GSK-3β/MKP-1/NRF2 pathway via the α2AR in IA.
Collapse
Affiliation(s)
- Ze Zhang
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| | - Xiue Mu
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| | - Xiaohui Zhou
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
30
|
Singh N, Anandan V, Ahmad SR. Effect of Dexmedetomidine as an adjuvant in quadratus lumborum block in patient undergoing caesarean section - A randomised controlled study. J Clin Anesth 2022; 81:110892. [PMID: 35671598 DOI: 10.1016/j.jclinane.2022.110892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
STUDY OBJECTIVE This study was conducted to evaluate the effect of Dexmedetomidine as an adjuvant in quadratus lumborum block (QLB) for postoperative pain relief at rest in patients undergoing caesarean section (CS). The primary objective was to compare the time to the first request of rescue analgesia. Secondary objectives were to compare the amount of rescue analgesia, patient satisfaction, Numeric rating scale (NRS), and Ramsay sedation score (RSS) during the first 24 h. DESIGN A randomised, double-blinded study. SETTING The study was conducted at AIIMS Bhubaneswar from December 2019 to February 2021in the Operating Theatre complex (for the immediate postoperative follow-up) and in the Obstetric Ward (for follow-up at the later time points). PATIENTS A total of 70 patients were enrolled with singleton term pregnancies scheduled for CS under spinal anaesthesia after written informed consent. INTERVENTION Bilateral QLB was given in the recovery area. Group A received 30 ml of 0.25% Bupivacaine and group B received 30 ml 0.25% bupivacaine with Dexmedetomidine 1 μg/kg. They received inj. Paracetamol 15 mg/kg intravenously TDS and Inj. Tramadol 1 mg/kg as rescue analgesia (if Numeric rating scale (NRS) Score ≥ 4). We also compared the rescue analgesia in the first 24 h, patient satisfaction scores, Ramsay sedation score (RSS), and NRS scores at 2, 4, 6, 8, 12, 18, and 24 h. MAIN RESULTS The time to request the first rescue analgesia was significantly prolonged in group B [Mean ± SD (95% CI)] 880 ± 351 (720-1040) min. vs group A 439 ± 208 (368-510) min., p < 0.001). There was a significant decrease in the amount of rescue analgesia [(Inj. Tramadol (1 mg/ kg)] used in the group with dexmedetomidine [group B Mean ± SD (95% CI) (57 ± 18 (49-65) mg. vs group A - 81 ± 25 (73-90)] mg., p < 0.001]. A significant difference was seen in patient satisfaction scores and pain scores between the groups up to 18 h. (p < 0.05) but not in RSS. CONCLUSION Dexmedetomidine can be considered an effective adjuvant for QLB in CS in the absence of intrathecal morphine.
Collapse
Affiliation(s)
- Neha Singh
- Department of Anesthesiology and Critical Care, All India Institute of Medical Sciences, Bhubaneswar 751019, Odisha, India.
| | - Vishnu Anandan
- Department of Anesthesiology and Critical Care, All India Institute of Medical Sciences, Bhubaneswar 751019, Odisha, India
| | - Suma Rabab Ahmad
- Department of Anesthesiology and Critical Care, All India Institute of Medical Sciences, Bhubaneswar 751019, Odisha, India
| |
Collapse
|
31
|
Kim JE, Yang SJ. miR-30a-5p Augments the Anti-inflammatory Effects of Dexmedetomidine in LPS-induced BV2 Cells. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2022. [DOI: 10.15324/kjcls.2022.54.3.201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Ji-Eun Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Korea
| |
Collapse
|
32
|
Luo SM, Li LY, Guo LZ, Wang L, Wang YF, Chen N, Wang E. Dexmedetomidine exerts an anti-inflammatory effect via α2 adrenoceptors to alleviate cognitive dysfunction in 5xFAD mice. Front Aging Neurosci 2022; 14:978768. [PMID: 36204551 PMCID: PMC9531904 DOI: 10.3389/fnagi.2022.978768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Inflammation promotes the progression of Alzheimer’s disease (AD). In this study, we explored the effect of dexmedetomidine on inflammation and cognitive function in a mouse model of AD. Methods: 5xFAD mice were intragastrically administered saline, dexmedetomidine, or dexmedetomidine and yohimbine for 14 days. The effects of dexmedetomidine on the acquisition and retention of memory in the Morris water-maze test and Y maze were evaluated. The deposition of amyloid beta protein (Abeta) and cytokine levels in the hippocampus were assessed. The expression of Bace1 protein and NFκB-p65 protein was assessed by Western blotting. Results: Compared with WT mice, 5xFAD mice exhibited cognitive impairment in the Morris water maze test and Y maze test. Cognitive decline was alleviated by dexmedetomidine and this was reversed by the α2 adrenoceptor antagonist yohimbine. Compared with saline treatment, dexmedetomidine led to a reduction in the Abeta deposition area (p < 0.05) and in the mean gray value (p < 0.01) in the hippocampus of 5xFAD mice. Compared with saline treatment, dexmedetomidine inhibited the activation of astrocytes and microglia in the hippocampal DG of 5xFAD mice and reduced the area of GFAP (p < 0.01) and IBA1 (p < 0.01). The level of IL-1β in the hippocampus decreased significantly after dexmedetomidine treatment compared with saline treatment in 5xFAD mice (p < 0.01). Yohimbine neutralized the effects of dexmedetomidine. Dexmedetomidine inhibited the expression of BACE1 and NF-κB p65 (p < 0.01), and these changes were reversed by yohimbine treatment. Conclusion: Dexmedetomidine alleviates cognitive decline, inhibits neuroinflammation, and prevents the deposition of Abeta in 5xFAD mice. The effect is mediated by the α2 adrenoceptor-mediated anti-inflammatory pathway. Dexmedetomidine may be effective for the treatment of AD and a better choice for the sedation of AD.
Collapse
Affiliation(s)
- Su-mei Luo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Long-yan Li
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| | - Li-zhe Guo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Yan-feng Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Na Chen
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- *Correspondence: E. Wang
| |
Collapse
|
33
|
Huang H, Zhu Y, Zhang Y, Hou B, Zhang Q, Shi X, Min J. Dexmedetomidine suppresses the isoflurane-induced neurological damage by upregulating Heme Oxygenase-1 via activation of the mitogen-activated protein kinase kinase 1/extracellular regulated protein kinases 1/nuclear factor erythroid 2-related factor 2 axis in aged rats. Chem Biol Interact 2022; 367:110114. [PMID: 36027947 DOI: 10.1016/j.cbi.2022.110114] [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: 01/24/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022]
Abstract
Dexmedetomidine (DEX) displays a neuroprotective role in aged rats with isoflurane (ISO)-induced cognitive impairment through antioxidant, and anti-inflammatory, and anti-apoptotic effects. Therefore, the present study was performed to define the molecular mechanism of DEX on ISO-induced neurological impairment in aged rats in relation to the MEK1/ERK1/Nrf2/HO-1 axis. The study enrolled elderly patients undergoing ISO anesthesia. Patient cognitive function following treatment with DEX was evaluated using mini-mental state examination (MMSE). The results revealed that DEX supplementation of anesthesia contributed to higher MMSE scores in patients one week post treatment. Rat model of neurological impairment was also induced in 18-month-age Wistar rats by ISO, followed by DEX treatment. Based on the results of Morris water maze experiment, ELISA, and TUNEL and hematoxylin-eosin staining, in vivo experiments confirmed that DEX could reduce the oxidative stress and neurological damage induced by ISO in rats. DEX activated the nuclear factor erythroid 2-related factor (Nrf2)/Heme Oxygenase 1 (HO-1) pathway. DEX upregulated the expression of Nrf2 and HO-1 by activating the MEK1/ERK1 pathway, whereby attenuating the ISO-caused oxidative stress and neurological damage in rats. Collectively, DEX suppresses the ISO-induced neurological impairment in the aged rats by promoting HO-1 through activation of the MEK1/ERK1/Nrf2 axis.
Collapse
Affiliation(s)
- Haijin Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Yunsheng Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Benchao Hou
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Qin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xiaoyun Shi
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Jia Min
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
34
|
Chen J, Chen B, Chen A. Dexmedetomidine improved respiratory dynamics and arterial blood gas indices in patients with esophageal cancer after induction of anesthesia. Am J Transl Res 2022; 14:5915-5922. [PMID: 36105043 PMCID: PMC9452352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To retrospectively analyze the effects of dexmedetomidine after induction of anesthesia on intraoperative indices in patients with esophageal cancer. METHODS The clinical data of 93 patients with esophageal cancer that admitted to our hospital from January 2019 to December 2020 were retrospectively analyzed. The patients were divided into control group (n=31), case group A (n=31, continuous intravenous infusion of 0.3 μg/(kg∙h) dexmedetomidine hydrochloride) and case group B (n=31, continuous intravenous infusion of 0.5 μg/(kg∙h) dexmedetomidine hydrochloride) according to the application condition of dexmedetomidine hydrochloride. Heart rate, blood pressure, arterial blood gas indicators (all measured by blood gas analyzer), respiratory mechanics index (measured by mechanical ventilation), ephedrine and atropine utilization rate of the three groups were compared. RESULTS The plateau pressure, peak pressure and airway resistance at the end of one-lung ventilation and at chest closure in case groups A and B were lower than those in the control group, and the pulmonary compliance in case group B was higher than that in the control group (P < 0.05). PaO2, P(A-a)O2, and RI before the start of OLV, at the end of OLV, and at chest closure in the three groups were significantly increased compared with those before induction of anesthesia (P < 0.05). Compared with the control group, PaO2 significantly increased, while P(A-a)O2 and RI significantly decreased at the end of OLV and at chest closure in the case group B. CONCLUSION Dexmedetomidine can improve respiratory dynamics and arterial blood gas indices after anesthesia induction of esophageal cancer, showing high safety and clinical feasibility.
Collapse
Affiliation(s)
- Jian Chen
- Department of Anesthesiology, Hainan Western Central Hospital Danzhou City 571700, Hainan Province, China
| | - Baoli Chen
- Department of Anesthesiology, Hainan Western Central Hospital Danzhou City 571700, Hainan Province, China
| | - Ailuan Chen
- Department of Anesthesiology, Hainan Western Central Hospital Danzhou City 571700, Hainan Province, China
| |
Collapse
|
35
|
α-Cyperone Improves Rat Spinal Cord Tissue Damage via Akt/Nrf2 and NF-κB Pathways. J Surg Res 2022; 276:331-339. [DOI: 10.1016/j.jss.2022.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/07/2022] [Accepted: 02/10/2022] [Indexed: 12/18/2022]
|
36
|
Blocking the Aryl Hydrocarbon Receptor Alleviates Myocardial Ischemia/Reperfusion Injury in Rats. Curr Med Sci 2022; 42:966-973. [DOI: 10.1007/s11596-022-2601-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
|
37
|
Wang J, Xin Y, Chu T, Liu C, Xu A. Dexmedetomidine attenuates perioperative neurocognitive disorders by suppressing hippocampal neuroinflammation and HMGB1/RAGE/NF-κB signaling pathway. Biomed Pharmacother 2022; 150:113006. [PMID: 35486975 DOI: 10.1016/j.biopha.2022.113006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 11/02/2022] Open
Abstract
Surgical trauma can induce an inflammatory response in the central nervous system. Neuroinflammation is a crucial pathological mechanism of perioperative neurocognitive disorders (PND). Dexmedetomidine (Dex) is an alpha (α)-2 adrenoceptor agonist that is widely used in the perioperative period. Previous studies have shown that Dex has neuroprotection in various nerve injury models, but its role in PND remains unclear. Our study aimed to observe the neuroprotective effect of Dex pretreatment on postoperative cognitive change and explore the effects of hippocampal neuroinflammation, microglial polarization and HMGB1/RAGE/NF-κB signaling pathway involved in Dex on PND in rats. Rats were pretreated with Dex alone or in combination with yohimbine (α-2 adrenoceptor antagonist) before surgery. Behavioral tests results showed that Dex ameliorated surgery-induced cognitive impairment in rats. Nissl, immunohistochemistry and TUNEL-NeuN staining results indicated that Dex reduced hippocampus damage and neuronal apoptosis caused by surgery. Dex preconditioning reduced the expression of the proinflammatory cytokines IL-1β, TNF-α and IL-6 in hippocampus. Immunohistochemical and immunofluorescence results showed that Dex preconditioning inhibited the activation of glial cells induced by surgery. Western blot analysis showed that Dex preconditioning downregulated the expression of M1 phenotype markers (CD86 and iNOS), HMGB1, RAGE and nuclear NF-κB and upregulated the expression of M2 phenotype markers (Arginase 1 and CD206) and cytoplasmic NF-κB. Yohimbine could inhibit the neuroprotective effect of Dex. These results indicated that Dex pretreatment could improve postoperative short-term cognitive impairment, and the neuroprotective mechanism may involve the suppression of hippocampal neuroinflammation, regulation of M1/M2 polarization, and inhibition of HMGB1/RAGE/NF-κB signal transduction.
Collapse
Affiliation(s)
- Jinxu Wang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yueyang Xin
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiantian Chu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cheng Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Aijun Xu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
38
|
Li J, Tang H, Tu W. Mechanism of dexmedetomidine preconditioning on spinal cord analgesia in rats with functional chronic visceral pain. Acta Cir Bras 2022; 37:e370203. [PMID: 35507967 PMCID: PMC9064187 DOI: 10.1590/acb370203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/03/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose: To analyze the effect and mechanism of dexmedetomidine (DEX) analgesia pretreatment on functional chronic visceral pain in rats. Methods: Rats were divided into six groups: W1, W2, W3, W4, W5, and W6. The behavioral changes and electrophysiological indexes of rats in each group before and after DEX treatment were detected. Results: The levels of abdominal withdrawal reflex (AWR) in W5 and W6 groups were significantly lower than those in group W3, while the levels of thermal withdrawal latency (TWL) and mechanical withdrawal threshold (MWT) were significantly higher than those in group W3 (p < 0.05). The electromyographic signals of W1, W5, and W6 groups showed little fluctuation, while those of groups W2, W3, and W4 showed obvious fluctuation. TLR4 mRNA expression, IRF3, P65, and phosphorylation levels in W4, W5, and W6 groups were significantly lower than those in group W2 (p < 0.05). Conclusions: Dexmedetomidine epidural anesthesia pretreatment could significantly inhibit visceral pain response in rats with functional chronic visceral pain, and its mechanism was related to the activation of TLR4 in spinal dorsal horn tissue of rats and the activation inhibition of IRF3 and P65 in the downstream key signals.
Collapse
Affiliation(s)
- Jun Li
- Southern Medical University, China
| | - Huizhong Tang
- People’s Hospital of Guangxi Zhuang Autonomous Region, China
| | - Weifeng Tu
- General Hospital of Southern Theatre Command, China
| |
Collapse
|
39
|
Chen H, Feng Z, Min L, Deng W, Tan M, Hong J, Gong Q, Zhang D, Liu H, Hou J. Vagus Nerve Stimulation Reduces Neuroinflammation Through Microglia Polarization Regulation to Improve Functional Recovery After Spinal Cord Injury. Front Neurosci 2022; 16:813472. [PMID: 35464311 PMCID: PMC9022634 DOI: 10.3389/fnins.2022.813472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/11/2022] [Indexed: 01/02/2023] Open
Abstract
BackgroundSpinal cord injury (SCI) is a devastating disease that lacks effective treatment. Interestingly, recent studies indicated that vagus nerve stimulation (VNS), neuromodulation that is widely used in a variety of central nervous system (CNS) diseases, improved motor function recovery after SCI. But the exact underlying mechanism of how VNS ameliorates SCI is unclear. This study aimed to confirm the efficacy and further explore the potential therapeutic mechanism of VNS in SCI.MethodA T10 spinal cord compression model was established in adult female Sprague-Dawley rats. Then the stimulation electrode was placed in the left cervical vagus nerve (forming Sham-VNS, VNS, and VNS-MLA groups). Basso-Beattie-Bresnahan (BBB) behavioral scores and Motor evoked potentials (MEPs) analysis were used to detect motor function. A combination of histological and molecular methods was used to clarify the relevant mechanism.ResultsCompared with the Sham-VNS group, the VNS group exhibited better functional recovery, reduced scar formation (both glial and fibrotic scars), tissue damage, and dark neurons, but these beneficial effects of VNS were diminished after alpha 7 nicotinic acetylcholine receptor (α7nAchR) blockade. Specifically, VNS inhibited the pro-inflammatory factors TNF-α, IL-1β, and IL-6 and increased the expression of the anti-inflammatory factors IL-10. Furthermore, we found that VNS promotes the shift of M1-polarized Iba-1+/CD86+ microglia to M2-polarized Iba-1+/CD206+ microglia via upregulating α7nAchR to alleviate neuroinflammation after SCI.ConclusionOur results demonstrated that VNS promotes microglial M2 polarization through upregulating α7nAChR to reduce neuroinflammation, thus improving motor function recovery after SCI. These findings indicate VNS might be a promising neuromodulation strategy for SCI.
Collapse
|
40
|
Fauss GNK, Hudson KE, Grau JW. Role of Descending Serotonergic Fibers in the Development of Pathophysiology after Spinal Cord Injury (SCI): Contribution to Chronic Pain, Spasticity, and Autonomic Dysreflexia. BIOLOGY 2022; 11:234. [PMID: 35205100 PMCID: PMC8869318 DOI: 10.3390/biology11020234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022]
Abstract
As the nervous system develops, nerve fibers from the brain form descending tracts that regulate the execution of motor behavior within the spinal cord, incoming sensory signals, and capacity to change (plasticity). How these fibers affect function depends upon the transmitter released, the receptor system engaged, and the pattern of neural innervation. The current review focuses upon the neurotransmitter serotonin (5-HT) and its capacity to dampen (inhibit) neural excitation. A brief review of key anatomical details, receptor types, and pharmacology is provided. The paper then considers how damage to descending serotonergic fibers contributes to pathophysiology after spinal cord injury (SCI). The loss of serotonergic fibers removes an inhibitory brake that enables plasticity and neural excitation. In this state, noxious stimulation can induce a form of over-excitation that sensitizes pain (nociceptive) circuits, a modification that can contribute to the development of chronic pain. Over time, the loss of serotonergic fibers allows prolonged motor drive (spasticity) to develop and removes a regulatory brake on autonomic function, which enables bouts of unregulated sympathetic activity (autonomic dysreflexia). Recent research has shown that the loss of descending serotonergic activity is accompanied by a shift in how the neurotransmitter GABA affects neural activity, reducing its inhibitory effect. Treatments that target the loss of inhibition could have therapeutic benefit.
Collapse
Affiliation(s)
| | | | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77843, USA; (G.N.K.F.); (K.E.H.)
| |
Collapse
|
41
|
Liu X, Li Y, Kang L, Wang Q. Recent Advances in the Clinical Value and Potential of Dexmedetomidine. J Inflamm Res 2022; 14:7507-7527. [PMID: 35002284 PMCID: PMC8724687 DOI: 10.2147/jir.s346089] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has sedative, anxiolytic, analgesic, sympatholytic, and opioid-sparing properties and induces a unique sedative response which shows an easy transition from sleep to wakefulness, thus allowing a patient to be cooperative and communicative when stimulated. Recent studies indicate several emerging clinical applications via different routes. We review recent data on dexmedetomidine studies, particularly exploring the varying routes of administration, experimental implications, clinical effects, and comparative advantages over other drugs. A search was conducted on the PubMed and Web of Science libraries for recent studies using different combinations of the words “dexmedetomidine”, “route of administration”, and pharmacological effect. The current routes, pharmacological effects, and application categories of dexmedetomidine are presented. It functions by stimulating pre- and post-synaptic α2-adrenoreceptors within the central nervous system, leading to hyperpolarization of noradrenergic neurons, induction of an inhibitory feedback loop, and reduction of norepinephrine secretion, causing a sympatholytic effect, in addition to its anti-inflammation, sleep induction, bowel recovery, and sore throat reduction effects. Compared with similar α2-adrenoceptor agonists, dexmedetomidine has both pharmacodynamics advantage of a significantly greater α2:α1-adrenoceptor affinity ratio and a pharmacokinetic advantage of having a significantly shorter elimination half-life. In its clinical application, dexmedetomidine has been reported to present a significant number of benefits including safe sedation for various surgical interventions, improvement of intraoperative and postoperative analgesia, sedation for compromised airways without respiratory depression, nephroprotection and stability of hypotensive hemodynamics, reduction of postoperative nausea and vomiting and postoperative shivering incidence, and decrease of intraoperative blood loss. Although the clinical application of dexmedetomidine is promising, it is still limited and further research is required to enhance understanding of its pharmacological properties, patient selection, dosage, and adverse effects.
Collapse
Affiliation(s)
- Xiaotian Liu
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yueqin Li
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Li Kang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| |
Collapse
|
42
|
Yu Q, Jiang X, Liu X, Shen W, Mei X, Tian H, Wu C. Glutathione-modified macrophage-derived cell membranes encapsulated metformin nanogels for the treatment of spinal cord injury. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112668. [DOI: 10.1016/j.msec.2022.112668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/20/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022]
|
43
|
Pang QM, Chen SY, Xu QJ, Fu SP, Yang YC, Zou WH, Zhang M, Liu J, Wan WH, Peng JC, Zhang T. Neuroinflammation and Scarring After Spinal Cord Injury: Therapeutic Roles of MSCs on Inflammation and Glial Scar. Front Immunol 2021; 12:751021. [PMID: 34925326 PMCID: PMC8674561 DOI: 10.3389/fimmu.2021.751021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Transected axons are unable to regenerate after spinal cord injury (SCI). Glial scar is thought to be responsible for this failure. Regulating the formation of glial scar post-SCI may contribute to axonal regrow. Over the past few decades, studies have found that the interaction between immune cells at the damaged site results in a robust and persistent inflammatory response. Current therapy strategies focus primarily on the inhibition of subacute and chronic neuroinflammation after the acute inflammatory response was executed. Growing evidences have documented that mesenchymal stem cells (MSCs) engraftment can be served as a promising cell therapy for SCI. Numerous studies have shown that MSCs transplantation can inhibit the excessive glial scar formation as well as inflammatory response, thereby facilitating the anatomical and functional recovery. Here, we will review the effects of inflammatory response and glial scar formation in spinal cord injury and repair. The role of MSCs in regulating neuroinflammation and glial scar formation after SCI will be reviewed as well.
Collapse
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qi-Jing Xu
- Department of Human Anatomy, Zunyi Medical University, Zunyi, China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi-Chun Yang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wang-Hui Zou
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Juan Liu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia-Chen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
44
|
Wen W, Gong X, Cheung H, Yang Y, Cai M, Zheng J, Tong X, Zhang M. Dexmedetomidine Alleviates Microglia-Induced Spinal Inflammation and Hyperalgesia in Neonatal Rats by Systemic Lipopolysaccharide Exposure. Front Cell Neurosci 2021; 15:725267. [PMID: 34955749 PMCID: PMC8692868 DOI: 10.3389/fncel.2021.725267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
Noxious stimulus and painful experience in early life can induce cognitive deficits and abnormal pain sensitivity. As a major component of the outer membrane of gram-negative bacteria, lipopolysaccharide (LPS) injection mimics clinical symptoms of bacterial infections. Spinal microglial activation and the production of pro-inflammatory cytokines have been implicated in the pathogenesis of LPS-induced hyperalgesia in neonatal rats. Dexmedetomidine (DEX) possesses potent anti-neuroinflammatory and neuroprotective properties through the inhibition of microglial activation and microglial polarization toward pro-inflammatory (M1) phenotype and has been widely used in pediatric clinical practice. However, little is known about the effects of DEX on LPS-induced spinal inflammation and hyperalgesia in neonates. Here, we investigated whether systemic LPS exposure has persistent effects on spinal inflammation and hyperalgesia in neonatal rats and explored the protective role of DEX in adverse effects caused by LPS injection. Systemic LPS injections induced acute mechanical hyperalgesia, increased levels of pro-inflammatory cytokines in serum, and short-term increased expressions of pro-inflammatory cytokines and M1 microglial markers in the spinal cord of neonatal rats. Pretreatment with DEX significantly decreased inflammation and alleviated mechanical hyperalgesia induced by LPS. The inhibition of M1 microglial polarization and microglial pro-inflammatory cytokines expression in the spinal cord may implicate its neuroprotective effect, which highlights a new therapeutic target in the treatment of infection-induced hyperalgesia in neonates and preterm infants.
Collapse
Affiliation(s)
- Wen Wen
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingrui Gong
- Department of Anesthesiology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Hoiyin Cheung
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyan Yang
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meihua Cai
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Tong
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mazhong Zhang
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Zhang Y, Bao D, Chi D, Li L, Liu B, Zhang D, Qiao L, Liang Y, Wang Y, Jin X. Dexmedetomidine vs. lidocaine for postoperative analgesia in pediatric patients undergoing craniotomy: a protocol for a prospective, randomized, double-blinded, placebo-controlled trial. Trials 2021; 22:800. [PMID: 34774098 PMCID: PMC8590361 DOI: 10.1186/s13063-021-05774-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022] Open
Abstract
Background Postoperative pain is a common problem that occurs in pediatric patients following neurosurgery which may lead to severe complications. Dexmedetomidine is a commonly used adjuvant medicine in craniotomy owing to its sedative, amnestic, analgesic, and neuroprotective properties. Besides, studies suggest that lidocaine has similar effects on sedation, analgesia, and neuroprotection. Both two adjuvants can reduce postoperative pain after neurosurgery in adults. However, it is still unknown whether dexmedetomidine or lidocaine can reduce postoperative pain in children undergoing craniotomy, and if yes, which is a better medicine choice. Therefore, we aimed to compare the effect of dexmedetomidine vs. lidocaine on postoperative pain in pediatric patients after craniotomy. Methods/design We will perform a randomized (1:1:1), double-blind, placebo-controlled, single-center trial. Children aged 1–12 years scheduled for craniotomy will be eligible for inclusion. The 255 recruited participants will be stratified by age in two strata (1–6 years and 7–12 years), and then each stratum will be equally randomized to three groups: group D (infusion of dexmedetomidine [intervention group]), group L (infusion of lidocaine [intervention group]), and group C (infusion of normal saline [control group]). Patients will be followed up at 1 h, 2 h, 4 h, 24 h, and 48 h after surgery. The primary outcome will be total sufentanil consumption within 24 h after surgery. Discussion In this clinical trial, we expect to clarify and compare the postoperative analgesic effect of dexmedetomidine vs. lidocaine infusion on pediatric patients undergoing craniotomy. We believe that the results of this trial will provide more choices for postoperative analgesia for the pediatric population. Trial registration Chinese ClinicalTrials.gov ChiCTR1800019411. Registered on 10 November 2018
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Di Bao
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Dongmei Chi
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Lu Li
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Bin Liu
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Di Zhang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Lanxin Qiao
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Yi Liang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Yaxin Wang
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Xu Jin
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.
| |
Collapse
|
46
|
Liu YB, Liu WF, Chen WC, Li W, Lin YL, Xu CJ, He HF. Dexmedetomidine alleviates traumatic spinal cord injury in rats via inhibiting apoptosis induced by endoplasmic reticulum stress. Neurol Res 2021; 44:275-284. [PMID: 34533101 DOI: 10.1080/01616412.2021.1979750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the protective effect of dexmedetomidine (Dex) on traumatic spinal cord injury (TSCI) and to evaluate the involvement of inhibition of endoplasmic reticulum (ER) stress response in the potential mechanism. METHOD Sprague-Dawley rats were randomly divided into five groups. The hind limb locomotor function of rats was evaluated at 1, 3 and 7 days after the operation. At 7 days after the operation, spinal cord specimens were obtained for hematoxylin and eosin (H&E), Nissl and TUNEL staining, as well as immunofluorescence and Western blot analyses to detect the level of apoptosis and the levels of proteins related to ER stress. RESULTS 7 days after the operation, Dex treatment promoted the recovery and also inhibited apoptosis of neurons in the spinal cord. Additionally, Dexinhibited the expression of proteins related to ER stress response after spinal cord injury. CONCLUSIONS Dex improves the neurological function of rats with TSCI and reduces apoptosis of spinal cord neurons. The potential mechanism is related to the inhibition of the ER stress response.
Collapse
Affiliation(s)
- Yi-Bin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Feng Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei Li
- Department of ICU, Wuhan Third Hospital, Wuhan University, Wuhan, China**
| | - Yan-Ling Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chong-Jun Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
47
|
Xie X, Shen Z, Hu C, Zhang K, Guo M, Wang F, Qin K. Dexmedetomidine Ameliorates Postoperative Cognitive Dysfunction in Aged Mice. Neurochem Res 2021; 46:2415-2426. [PMID: 34159456 DOI: 10.1007/s11064-021-03386-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/20/2022]
Abstract
Neuroinflammation and oxidative stress coexist and interact in the progression of postoperative cognitive dysfunction (POCD) and other neurodegenerative disease. Mounting studies reveal that Dexmedetomidine (Dex) possesses anti-inflammatory and antioxidant properties. Nevertheless, whether Dex exerts neuroprotective effect on the cognitive sequelae of oxidative stress and inflammatory process remains unclear. A mouse model of abdominal exploratory laparotomy-induced cognitive dysfunction was employed to explore the underlying mechanism of neuroprotective effects exerted by Dex in POCD. Aged mice were treated with Dex (20 µg/kg) 20 min prior to surgery. Open field test (OFT) and Morris water maze (MWM) were employed to examine the cognitive function on postoperative day 3 (POD 3) or POD 7. In the present study, mice underwent surgery exhibited cognitive impairment without altering spontaneous locomotor activity, while the surgery-induced cognitive impairment could be alleviated by Dex pretreatment. Dex inhibited surgery-induced pro-inflammatory cytokines accumulation and microglial activation in the hippocampi of mice. Furthermore, Dex decreased MDA levels, enhanced SOD activity, modulated CDK5 activity and increased BDNF expression in the hippocampus. In addition, Dex remarkably reduced the surgery-induced increased ratio of Bax/Bcl-2 and apoptotic neurons in the hippocampi of aged mice. Collectively, our study provides evidence that Dex may exert neuroprotective effects against surgery-induced cognitive impairment through mechanisms involving its anti-inflammatory and antioxidant properties, as well as the suppression on the mitochondrial permeability transition pore and apoptosis-related pathway.
Collapse
Affiliation(s)
- Xiaolan Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Zhiwen Shen
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Mingyan Guo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Fei Wang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Kai Qin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
48
|
Kii N, Sawada A, Yoshikawa Y, Tachibana S, Yamakage M. Dexmedetomidine Ameliorates Perioperative Neurocognitive Disorders by Suppressing Monocyte-Derived Macrophages in Mice With Preexisting Traumatic Brain Injury. Anesth Analg 2021; 134:869-880. [PMID: 34319918 DOI: 10.1213/ane.0000000000005699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) initiates immune responses involving infiltration of monocyte-derived macrophages (MDMs) in the injured brain tissue. These MDMs play a key role in perioperative neurocognitive disorders (PNDs). We tested the hypothesis that preanesthetic treatment with dexmedetomidine (DEX) could suppress infiltration of MDMs into the hippocampus of TBI model mice, ameliorating PND. METHODS We first performed bone marrow transplantation from green fluorescent protein-transgenic mice to C57BL/6 mice to identify MDMs. We used only male mice for homogeneity. Four weeks after transplantation, a controlled cortical impact model of TBI was created using recipient mice. Four weeks after TBI, mice received pretreatment with DEX before general anesthesia (GA). Mice performed the Barnes maze test (8-12 mice/group) 2 weeks after GA and were euthanized for immunohistochemistry (4-5 mice/group) or immunoblotting (7 mice/group) 4 weeks after GA. RESULTS In Barnes maze tests, TBI model mice showed longer primary latency (mean difference, 76.5 [95% confidence interval, 41.4-111.6], P < .0001 versus Naïve), primary path length (431.2 [98.5-763.9], P = .001 versus Naïve), and more primary errors (5.7 [0.62-10.7], P = .017 versus Naïve) than Naïve mice on experimental day 3. Expression of MDMs in the hippocampus was significantly increased in TBI mice compared to Naïve mice (2.1 [0.6-3.7], P = .003 versus Naïve). Expression of monocyte chemotactic protein-1 (MCP1)-positive areas in the hippocampus was significantly increased in TBI mice compared to Naïve mice (0.38 [0.09-0.68], P = .007 versus Naïve). Immunoblotting indicated significantly increased expression of interleukin-1β in the hippocampus in TBI mice compared to Naïve mice (1.59 [0.08-3.1], P = .035 versus Naïve). In contrast, TBI mice pretreated with DEX were rescued from these changes and showed no significant difference from Naïve mice. Yohimbine, an α2 receptor antagonist, mitigated the effects of DEX (primary latency: 68.3 [36.5-100.1], P < .0001 versus TBI-DEX; primary path length: 414.9 [120.0-709.9], P = .0002 versus DEX; primary errors: 6.6 [2.1-11.2], P = .0005 versus TBI-DEX; expression of MDMs: 2.9 [1.4-4.4], P = .0001 versus TBI-DEX; expression of MCP1: 0.4 [0.05-0.67], P = .017 versus TBI-DEX; expression of interleukin-1β: 1.8 [0.34-3.35], P = .01 versus TBI-DEX). CONCLUSIONS Preanesthetic treatment with DEX suppressed infiltration of MDMs in the hippocampus and ameliorated PND in TBI model mice. Preanesthetic treatment with DEX appears to suppress infiltration of MDMs in the hippocampus and may lead to new treatments for PND in patients with a history of TBI.
Collapse
Affiliation(s)
- Natsumi Kii
- From the Department of Anesthesiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | |
Collapse
|
49
|
Scott MC, Bedi SS, Olson SD, Sears CM, Cox CS. Microglia as therapeutic targets after neurological injury: strategy for cell therapy. Expert Opin Ther Targets 2021; 25:365-380. [PMID: 34029505 DOI: 10.1080/14728222.2021.1934447] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Microglia is the resident tissue macrophages of the central nervous system. Prolonged microglial activation often occurs after traumatic brain injury and is associated with deteriorating neurocognitive outcomes. Resolution of microglial activation is associated with limited tissue loss and improved neurocognitive outcomes. Limiting the prolonged pro-inflammatory response and the associated secondary tissue injury provides the rationale and scientific premise for considering microglia as a therapeutic target. AREAS COVERED In this review, we discuss markers of microglial activation, such as immunophenotype and microglial response to injury, including cytokine/chemokine release, free radical formation, morphology, phagocytosis, and metabolic shifts. We compare the origin and role in neuroinflammation of microglia and monocytes/macrophages. We review potential therapeutic targets to shift microglial polarization. Finally, we review the effect of cell therapy on microglia. EXPERT OPINION Dysregulated microglial activation after neurologic injury, such as traumatic brain injury, can worsen tissue damage and functional outcomes. There are potential targets in microglia to attenuate this activation, such as proteins and molecules that regulate microglia polarization. Cellular therapeutics that limit, but do not eliminate, the inflammatory response have improved outcomes in animal models by reducing pro-inflammatory microglial activation via secondary signaling. These findings have been replicated in early phase clinical trials.
Collapse
Affiliation(s)
- M Collins Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston (Uthealth), USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Candice M Sears
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
50
|
Li LC, Tian Y, Xiao J, Yang Y, Wu JN, Chen Y, Zhang PH, Gao-Smith F, Wang JG, Jin SW. Dexmedetomidine promotes inflammation resolving through TGF-β1 secreted by F4/80 +Ly6G + macrophage. Int Immunopharmacol 2021; 95:107480. [PMID: 33676148 DOI: 10.1016/j.intimp.2021.107480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist, which can regulate inflammatory responses. However, whether DEX interferes with the inflammation resolving remains unclear. Here, we reported the effects of DEX on zymosan-induced generalized inflammation in mice during resolution. Mice were administered intraperitoneally with DEX after the initiation of sepsis. The resolution interval (Ri), a vital resolution indice, decreased from twelve hours to eight hours after the administration of DEX. The induction of peritoneal pro-inflammatory interleukin [IL] - 1β and tumour necrosis factor-α (TNF-α) appeared to be inhibited. Of interest, the anti-inflammatory transforming growth factor-β1 (TGF-β1) but not IL-10 levels were up-regulated at twenty-four hours in the DEX group along with 1.0 mg/mice zymosan A (ZyA) treatment. The expression levels of multiple genes related to protective immune processes and clearance functions were detected and revealed the same trends. DEX markedly increased the F4/80+Ly6G+ macrophage population. Additionally, the adequate apoptotic neutrophil clearance from injury after DEX installation could be reverse by opsonization or co-instillation of TGF-β1 neutralizing antibody in vivo, promoting the inflammation-resolution programs. In conclusion, DEX post-treatment, via the increase of F4/80+Ly6G+ macrophages, provokes further secretion of TGF-β1, leading to the attenuated cytokine storm and accelerated inflammation resolving.
Collapse
Affiliation(s)
- Lin-Chao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yang Tian
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Jin-Ni Wu
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China
| | - Yan Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Fang Gao-Smith
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| | - Jian-Guang Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China.
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| |
Collapse
|