1
|
Park YH, Park HP, Kim E, Lee H, Hwang JW, Jeon YT, Lim YJ. The antioxidant effect of preischemic dexmedetomidine in a rat model: increased expression of Nrf2/HO-1 via the PKC pathway. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ELSEVIER) 2023; 73:177-185. [PMID: 34560114 PMCID: PMC10068566 DOI: 10.1016/j.bjane.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 08/03/2021] [Accepted: 08/21/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND The precise underlying mechanism of antioxidant effects of dexmedetomidine-induced neuroprotection against cerebral ischemia has not yet been fully elucidated. Activation of Nuclear factor erythroid 2-related factor (Nrf2) and Heme Oxygenase-1 (HO-1) represents a major antioxidant-defense mechanism. Therefore, we determined whether dexmedetomidine increases Nrf2/HO-1 expression after global transient cerebral ischemia and assessed the involvement of Protein Kinase C (PKC) in the dexmedetomidine-related antioxidant mechanism. METHODS Thirty-eight rats were randomly assigned to five groups: sham (n...=...6), ischemic (n...=...8), chelerythrine (a PKC inhibitor; 5...mg.kg-1 IV administered 30...min before cerebral ischemia) (n...=...8), dexmedetomidine (100.....g.kg-1 IP administered 30...min before cerebral ischemia (n...=...8), and dexmedetomidine...+...chelerythrine (n...=...8). Global transient cerebral ischemia (10...min) was applied in all groups, except the sham group; histopathologic changes and levels of nuclear Nrf2 and cytoplasmic HO-1 were examined 24...hours after ischemia insult. RESULTS We found fewer necrotic and apoptotic cells in the dexmedetomidine group relative to the ischemic group (p...<...0.01) and significantly higher Nrf2 and HO-1 levels in the dexmedetomidine group than in the ischemic group (p...<...0.01). Additionally, chelerythrine co-administration with dexmedetomidine attenuated the dexmedetomidine-induced increases in Nrf2 and HO-1 levels (p...<...0.05 and p...<...0.01, respectively) and diminished its beneficial neuroprotective effects. CONCLUSION Preischemic dexmedetomidine administration elicited neuroprotection against global transient cerebral ischemia in rats by increasing Nrf2/HO-1 expression partly via PKC signaling, suggesting that this is the antioxidant mechanism underlying dexmedetomidine-mediated neuroprotection.
Collapse
Affiliation(s)
- Yong-Hee Park
- Chung-Ang University College of Medicine, Chung-Ang University Hospital, Department of Anesthesiology and Pain Medicine, Seoul, South Korea
| | - Hee-Pyoung Park
- Seoul National University College of Medicine, Seoul National University Hospital, Department of Anesthesiology and Pain Medicine, Seoul, South Korea
| | - Eugene Kim
- Hanyang University Medical Center, College of Medicine, Hanyang University, Department of Anesthesiology and Pain Medicine, Seoul, South Korea
| | - Hannah Lee
- Seoul National University College of Medicine, Seoul National University Hospital, Department of Anesthesiology and Pain Medicine, Seoul, South Korea
| | - Jung-Won Hwang
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Department of Anesthesiology and Pain Medicine, Seongnam, South Korea
| | - Young-Tae Jeon
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Department of Anesthesiology and Pain Medicine, Seongnam, South Korea
| | - Young-Jin Lim
- Seoul National University College of Medicine, Seoul National University Hospital, Department of Anesthesiology and Pain Medicine, Seoul, South Korea.
| |
Collapse
|
2
|
Chen F, Wang D, Jiang Y, Ma H, Li X, Wang H. Dexmedetomidine postconditioning alleviates spinal cord ischemia-reperfusion injury in rats via inhibiting neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation. Int J Neurosci 2023; 133:1-12. [PMID: 33499703 DOI: 10.1080/00207454.2021.1881089] [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] [Indexed: 01/27/2023]
Abstract
PURPOSE Spinal cord ischemia-reperfusion (I/R) injury is an unresolved complication and its mechanisms are still not completely understood. Here, we studied the neuroprotective effects of dexmedetomidine (DEX) postconditioning against spinal cord I/R injury in rats and explored the possible mechanisms. MATERIALS AND METHODS In the study, rats were randomly divided into five groups: sham group, I/R group, DEX0.5 group, DEX2.5 group, and DEX5 group. I/R injury was induced in experimental rats; 0.5 μg/kg, 2.5 μg/kg, 5 μg/kg DEX were intravenously injected upon reperfusion respectively. Neurological function, histological assessment, and the disruption of blood-spinal cord barrier (BSCB) were evaluated via the BBB scoring, hematoxylin and eosin staining, Evans Blue (EB) extravasation and spinal cord edema, respectively. Neutrophil infiltration was evaluated via Myeloperoxidase (MPO) activity. Microglia activation and reactive gliosis was evaluated via ionized calcium-binding adapter molecule-1(IBA-1) and glial fibrillary acidic protein (GFAP) immunofluorescence, respectively. The expression of C-X-C motif ligand 13 (CXCL13), C-X-C chemokine receptor type 5(CXCR5), caspase-3 was determined by western blotting. The expression levels of interleukin 6(IL-6), tumor necrosis factor-α(TNF-α), IL-1β were determined by ELISA assay. RESULTS DEX postconditioning preserved neurological assessment scores, improved histological assessment scores, attenuated BSCB leakage after spinal cord I/R injury. Neutrophil infiltration, microglia activation and reactive gliosis were also inhibited by DEX postconditioning. The expression of CXCL13, CXCR5, caspase-3, IL-6, TNF-α, IL-1β were reduced by DEX postconditioning. CONCLUSIONS DEX postconditioning alleviated spinal cord I/R injury, which might be mediated via inhibition of neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation.
Collapse
Affiliation(s)
- Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yanhua Jiang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hong Ma
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaoqian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - He Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
3
|
Liu T, Liu FC, Xia Y, Luo TJ, Wang F, Chen B, Wang C, Gao GK, Zhai WT, Liu W. Effect of dexmedetomidine on the Montreal Cognitive Assessment in older patients undergoing pulmonary surgery. J Int Med Res 2022; 50:3000605221123680. [PMID: 36151758 PMCID: PMC9513575 DOI: 10.1177/03000605221123680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objective We investigated the effect of dexmedetomidine anesthesia on postoperative
cognitive function after pulmonary surgery. Methods A blinded, prospective, randomized, placebo-controlled study was performed on
60 patients (age range 65–74 years) undergoing lobectomy by video-assisted
thoracoscopic surgery (29 in the dexmedetomidine group; 31 in the placebo
group). Dexmedetomidine group patients received dexmedetomidine, and placebo
group patients received an equal volume of physiological saline 20 minutes
before anesthesia induction. Cognitive function was evaluated using the
Montreal Cognitive Assessment 1 day before surgery and on postoperative day
(POD)1, POD3, and POD7. The regional cerebral oxygen saturation
(rSO2) was monitored continuously by near-infrared
spectroscopy before anesthesia. Results The Montreal Cognitive Assessment score was significantly different between
the two groups on POD1 (dexmedetomidine 26.4 ± 0.73 vs.
placebo 25.5 ± 0.96) and POD3 (dexmedetomidine 27.1 ± 0.79
vs. placebo 26.6 ± 0.80). Specifically, attention and
orientation scores were increased in the dexmedetomidine group on POD1 and
POD3. The rSO2 was not significantly different between the
dexmedetomidine and placebo groups. Conclusion Dexmedetomidine given before induction of anesthesia could reduce the risk of
postoperative cognitive dysfunction and might not decrease rSO2.
Hence, dexmedetomidine could be employed in pulmonary surgical procedures,
especially for older patients with a high risk of cognitive dysfunction.
Collapse
Affiliation(s)
- Tao Liu
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Fang-Chao Liu
- Scientific Research Office, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Xia
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Tai-Jun Luo
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - Fei Wang
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bin Chen
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Chun Wang
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Guang-Kuo Gao
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wen-Ting Zhai
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wei Liu
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Zhu CH, Yu J, Wang BQ, Nie Y, Wang L, Shan SQ. Dexmedetomidine reduces ventilator-induced lung injury via ERK1/2 pathway activation. Mol Med Rep 2020; 22:5378-5384. [PMID: 33173983 PMCID: PMC7647005 DOI: 10.3892/mmr.2020.11612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanical ventilation (MV) can contribute to ventilator-induced lung injury (VILI); dexmedetomidine (Dex) treatment attenuates MV-related pulmonary inflammation, but the mechanisms remain unclear. Therefore, the present study aimed to explore the protective effect and the possible molecular mechanisms of Dex in a VILI rodent model. Adult male Sprague-Dawley rats were randomly assigned to one of seven groups (n=24 rats/group). Rats were euthanized after 4 h of continuous MV, and pathological changes, lung wet/dry (W/D) weight ratio, the levels of inflammatory cytokines (IL-1β, TNF-α and IL-6) in the bronchoalveolar lavage fluid (BALF), and the expression levels of Bcl-2 homologous antagonist/killer (Bak), Bcl-2, pro-caspase-3, cleaved caspase-3 and the phosphorylation of ERK1/2 in the lung tissues were measured. Propidium iodide uptake and TUNEL staining were used to detect epithelial cell death. The Dex pretreatment group exhibited fewer pathological changes, lower W/D ratios and lower expression levels of inflammatory cytokines in BALF compared with the VILI group. Dex significantly attenuated the ratio of Bak/Bcl-2, cleaved caspase-3 expression levels and epithelial cell death, and increased the expression of phosphorylated ERK1/2. The protective effects of Dex could be partially reversed by PD98059, which is a mitogen-activated protein kinase (upstream of ERK1/2) inhibitor. Overall, dexmedetomidine was found to reduce the inflammatory response and epithelial cell death caused by VILI, via the activation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Chun-Hua Zhu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Jian Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Ben-Qing Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yu Nie
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Lei Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Shi-Qiang Shan
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
6
|
Lei S, Lu P, Lu Y, Zheng J, Li W, Wang N, Zhang H, Li R, Wang K, Wen J, Wei H, Zhang Y, Qiu Z, Xu J, Lv H, Chen X, Liu Y, Zhang P. Dexmedetomidine Alleviates Neurogenesis Damage Following Neonatal Midazolam Exposure in Rats through JNK and P38 MAPK Pathways. ACS Chem Neurosci 2020; 11:579-591. [PMID: 31999428 DOI: 10.1021/acschemneuro.9b00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Midazolam, a widely used anesthetic, inhibits proliferation of neural stem cells (NSCs) and induces neuroapoptosis in neonates. Dexmedetomidine, an effective auxiliary medicine in clinical anesthesia, protects the developing brain against volatile anesthetic-induced neuroapoptosis. Whether dexmedetomidine protects against neurogenesis damage induced by midazolam remains unknown. This study aims to clarify the protective effect of dexmedetomidine on midazolam-induced neurogenesis damage and explore its potential mechanism. Postnatal 7-day-old Sprague-Dawley (SD) rats and cultured NSCs were treated with either normal saline, midazolam, or dexmedetomidine combined with midazolam. The rats were sacrificed at 1, 3, and 7 days after treatment. Cell proliferation was assessed by 5-bromodeoxyurdine (BrdU) incorporation. Cell viability was determined using MTT assay. Cell differentiation and apoptosis were detected by immunofluorescent staining and terminal dUTP nick-end labeling (TUNEL), respectively. The protein levels of p-JNK, p-P38, and cleaved caspase-3 were quantified using Western blotting. Midazolam decreased cell proliferation and increased cell apoptosis in the subventricular zone (SVZ), the subgranular zone (SGZ) of the hippocampus, and cultured NSCs. Moreover, midazolam decreased cell viability and increased the expression of p-JNK and p-P38 in cultured NSCs. Co-treatment with dexmedetomidine attenuated midazolam-induced changes in cell proliferation, viability, apoptosis, and protein expression of p-JNK and p-P38 in cultured NSCs. Midazolam and dexmedetomidine did not affect the differentiation of the cultured NSCs. These results indicate that dexmedetomidine alleviated midazolam-induced neurogenesis damage via JNK and P38 MAPK pathways.
Collapse
Affiliation(s)
- Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jing Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
7
|
Gao J, Sun Z, Xiao Z, Du Q, Niu X, Wang G, Chang YW, Sun Y, Sun W, Lin A, Bresnahan JC, Maze M, Beattie MS, Pan JZ. Dexmedetomidine modulates neuroinflammation and improves outcome via alpha2-adrenergic receptor signaling after rat spinal cord injury. Br J Anaesth 2019; 123:827-838. [PMID: 31623841 DOI: 10.1016/j.bja.2019.08.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/17/2019] [Accepted: 08/17/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Spinal cord injury induces inflammatory responses that include the release of cytokines and the recruitment and activation of macrophages and microglia. Neuroinflammation at the lesion site contributes to secondary tissue injury and permanent locomotor dysfunction. Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, is anti-inflammatory and neuroprotective in both preclinical and clinical trials. We investigated the effect of DEX on the microglial response, and histological and neurological outcomes in a rat model of cervical spinal cord injury. METHODS Anaesthetised rats underwent unilateral (right) C5 spinal cord contusion (75 kdyne) using an impactor device. The locomotor function, injury size, and inflammatory responses were assessed. The effect of DEX was also studied in a microglial cell culture model. RESULTS DEX significantly improved the ipsilateral upper-limb motor dysfunction (grooming and paw placement; P<0.0001 and P=0.0012), decreased the injury size (P<0.05), spared white matter (P<0.05), and reduced the number of activated macrophages (P<0.05) at the injury site 4 weeks post-SCI. In DEX-treated rats after injury, tissue RNA expression indicated a significant downregulation of pro-inflammatory markers (e.g. interleukin [IL]-1β, tumour necrosis factor-α, interleukin (IL)-6, and CD11b) and an upregulation of anti-inflammatory and pro-resolving M2 responses (e.g. IL-4, arginase-1, and CD206) (P<0.05). In lipopolysaccharide-stimulated cultured microglia, DEX produced a similar inflammation-modulatory effect as was seen in spinal cord injury. The benefits of DEX on these outcomes were mostly reversed by an α2-adrenergic receptor antagonist. CONCLUSIONS DEX significantly improves neurological outcomes and decreases tissue damage after spinal cord injury, which is associated with modulation of neuroinflammation and is partially mediated via α2-adrenergic receptor signaling.
Collapse
Affiliation(s)
- Jiandong Gao
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhihua Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Xiao
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Qihang Du
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Xinhuan Niu
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Gongming Wang
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yu-Wen Chang
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA
| | - Yongtao Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Qianfoshan Hospital, the First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Wei Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Amity Lin
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jacqueline C Bresnahan
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mervyn Maze
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael S Beattie
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Jonathan Z Pan
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
|
9
|
Zhu MY, Raza MU, Zhan Y, Fan Y. Norepinephrine upregulates the expression of tyrosine hydroxylase and protects dopaminegic neurons against 6-hydrodopamine toxicity. Neurochem Int 2019; 131:104549. [PMID: 31539561 DOI: 10.1016/j.neuint.2019.104549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/27/2019] [Accepted: 09/17/2019] [Indexed: 10/26/2022]
Abstract
As a classic neurotransmitter in the brain, norepinephrine (NE) also is an important modulator to other neuronal systems. Using primary cultures from rat ventral mesencephalon (VM) and dopaminergic cell line MN9D, the present study examined the neuroprotective effects of NE and its effects on the expression of tyrosine hydroxylase (TH). The results showed that NE protected both VM cultures and MN9D cells against 6-hydroxydopamine-caused apoptosis, with possible involvement of adrenal receptors. In addition, treatment with NE upregulated TH protein levels in dose- and time-dependent manner. Further experiments to investigate the potential mechanisms underlying this NE-induced upregulation of TH demonstrated a marked increase in protein levels of the brain-derived neurotrophic factor (BDNF) and the phosphorylated extracellular signal-regulated protein kinase 1 and 2 (pERK1/2) in VM cultures treated with NE. In MN9D cells, a significantly increase of TH and pERK1/2 protein levels were observed after their transfection with BDNF cDNA or exposure to BDNF peptides. Treatment of VM cultures with K252a, an antagonist of the tropomyosin-related kinase B, blocked the upregulatory effects of NE on TH, BDNF and pERK1/2. Administration of MEK1 & MEK2 inhibitors also reversed NE-induced upregulation of TH and pERK1/2. Moreover, ChIP assay showed that treatment with NE or BDNF increased H4 acetylation in the TH promoter. These results suggest that the neuroprotection and modulation of NE on dopaminergic neurons are mediated via BDNF and MAPK/ERK pathways, as well as through epigenetic histone modification, which may have implications for the improvement of therapeutic strategies for Parkinson's disease.
Collapse
Affiliation(s)
- Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Yanqiang Zhan
- Department of Neurology, Remin Hospital of the Wuhan University, Wuhan, China
| | - Yan Fan
- Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| |
Collapse
|
10
|
Goyagi T. Dexmedetomidine reduced sevoflurane-induced neurodegeneration and long-term memory deficits in neonatal rats. Int J Dev Neurosci 2019; 75:19-26. [PMID: 30959098 DOI: 10.1016/j.ijdevneu.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
Exposure to sevoflurane and other inhalational anesthetics can induce neurodegeneration in the developing brain. Although dexmedetomidine (DEX) has provided neuroprotection against hypoxic ischemic injury, relatively little is known about whether it has the neuroprotective effects against anesthetic-induced neurodegeneration. This study examined whether DEX improves the long-term cognitive dysfunction observed after exposure of neonatal rats to 3% sevoflurane. Seven-day-old rats received intraperitoneal saline (DEX 0) or DEX (6.6, 12.5, 25 μg/kg) 30 min before exposure to 3% sevoflurane with 21% oxygen for 4 h (n = 10 per group). The pups in the control group received only DEX 25 μg/kg without anesthesia. The escape latency in the Morris water maze was significantly increased in the DEX 0 group compared with the sham and control group, and the escape latency, but not the swimming path length, was significantly shorter at post-natal day 47 in the DEX 25 than in the DEX 0 group. The percent time spent in the quadrant was significantly decreased in the DEX 0 group compared with the sham and control group, and the percent time spent in the quadrant was significantly increased in the DEX 25 group compared with the DEX 0 groups. The freezing times of the DEX 0 and 6.6 groups were significantly decreased compared with those in the sham, control and DEX 25 groups. The number of NeuN-positive cells in the CA1 region was significantly decreased in the DEX 0 and 6.6 groups compared with the sham, control and DEX 25 groups. These findings indicate pre-treatment with DEX may improve long-term cognitive function and ameliorate the neuronal degeneration induced by sevoflurane exposure in neonatal rats.
Collapse
Affiliation(s)
- Toru Goyagi
- Department of Anesthesia and Intensive Care Medicine, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita 010-8543, Japan.
| |
Collapse
|
11
|
Xia F, Chang X, Zhang Y, Wang L, Xiao F. The effect of intrathecal dexmedetomidine on the dose requirement of hyperbaric bupivacaine in spinal anaesthesia for caesarean section: a prospective, double-blinded, randomized study. BMC Anesthesiol 2018; 18:74. [PMID: 29935528 PMCID: PMC6015661 DOI: 10.1186/s12871-018-0528-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/25/2018] [Indexed: 11/10/2022] Open
Abstract
Background Studies have shown that dexmedetomidine (Dex) can prolong the duration of analgesia when added to local anaesthetic as an adjuvant in a central or peripheral nerve block. We hypothesized that intrathecal Dex can reduce the ED95 of spinal hyperbaric bupivacaine. Therefore, we conducted this prospective, double-blinded, randomized study to verify our hypothesis. Methods Ninety patients were allocated into the Dexmedetomidine group (received bupivacaine + 5 mcg dexmedetomidine) and the Control group (received bupivacaine + the same volume of saline) using a double-blinded and randomized method. The first patient in each group received 5 mg of IT hyperbaric bupivacaine, and the next dose for the following patient was determined by the probability of successful anaesthesia of the previous neighbouring dose. An improved up-down sequence allocated method combined with probit analysis was used to determine the ED95 of intrathecal hyperbaric bupivacaine for the two groups. Results The ED95 and 95% confidence intervals (95% CI) of IT hyperbaric bupivacaine of the Dex group and Control group were 8.4 mg (95% CI, 6.5~ 13.8 mg) and 12.1 mg (95% CI, 8.3~ 312.8 mg), respectively. The duration of sensory block was longer in the Dex group than in the Control group (110.3 ± 35.3 vs 67.5 ± 26.2). The duration of analgesia was also longer in the Dex group than in the Control group (224.9 ± 45.4 vs 155.1 ± 31.6). The consumption of postoperative rescued sufentanil was significantly higher in the Control group than in the Dex group. Conclusion Intrathecal 5 mcg dexmedetomidine potentiated hyperbaric bupivacaine antinociception by 31% in spinal anaesthesia for patients undergoing caesarean section. Trial registration We registered this study in a Chinese Clinical Trial Registry (ChiCTR) centre on Nov 1st 2016 and received the registration number: ChiCTR-IPR-16009699.
Collapse
Affiliation(s)
- Feng Xia
- Department of Anaesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, China
| | - Xiangyang Chang
- Department of Anaesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, China
| | - Yinfa Zhang
- Department of Anaesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, China
| | - Lizhong Wang
- Department of Anaesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, China.
| | - Fei Xiao
- Department of Anaesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, China.
| |
Collapse
|
12
|
Zhang MH, Zhou XM, Cui JZ, Wang KJ, Feng Y, Zhang HA. Neuroprotective effects of dexmedetomidine on traumatic brain injury: Involvement of neuronal apoptosis and HSP70 expression. Mol Med Rep 2018; 17:8079-8086. [PMID: 29693126 PMCID: PMC5983975 DOI: 10.3892/mmr.2018.8898] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of dexmedetomidine (Dex) on traumatic brain injury (TBI), and further evaluate whether the underlying neuroprotective mechanisms are associated with neurological apoptosis and the expression of 70 kDa heat shock protein (HSP70) in the hippocampus. A total of 90 adult male Sprague-Dawley rats were randomly assigned into 3 groups (n=30/group): Sham, TBI and Dex groups. The rat models of TBI were established using a modified weight-drop device and Dex (15 µg/kg) was intravenously administered immediately following TBI. The brain edema and neurological function outcomes of TBI were assessed using wet-dry weight analysis and the Neurological Severity Score method. The expression levels of B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X protein (Bax) in the rat hippocampus were evaluated using immunohistochemical staining and western blot analysis. The protein levels of HSP70 in the hippocampal region were analyzed using western blot analysis. The results of the present study revealed that administration of Dex post-TBI improved brain edema and neurological outcomes, due to the attenuation of the TBI-induced reduction of Bax expression and increase of Bcl-2 and HSP70 expression. In conclusion, the results of the present study suggested that administration of Dex may serve as a neuroprotective agent against brain injury, at least partially via the inhibition of neuronal apoptosis and upregulation of HSP70 expression in the hippocampus.
Collapse
Affiliation(s)
- Man-He Zhang
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xiu-Min Zhou
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jian-Zhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Kai-Jie Wang
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yan Feng
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Hong-Ao Zhang
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
13
|
Ohbuchi M, Kimura T, Nishikawa T, Horiguchi T, Fukuda M, Masaki Y. Neuroprotective Effects of Fasudil, a Rho-Kinase Inhibitor, After Spinal Cord Ischemia and Reperfusion in Rats. Anesth Analg 2018; 126:815-823. [DOI: 10.1213/ane.0000000000002602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|