1
|
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
|
2
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [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: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
3
|
Tao Z, Li P, Zhao X. Progress on the Mechanisms and Neuroprotective Benefits of Dexmedetomidine in Brain Diseases. Brain Behav 2024; 14:e70116. [PMID: 39482839 PMCID: PMC11527817 DOI: 10.1002/brb3.70116] [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: 07/14/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION Dexmedetomidine, a highly specific α2 agonist, has been extensively utilized in clinical sedation and surgical anesthesia since its introduction in 2000 due to its excellent sympatholytic, sedative, and analgesic effects. This review aimed to identify new approaches for the treatment of patients with brain disorders by thoroughly describing the mechanism of action of dexmedetomidine and examining its neuroprotective effects from the standpoints of basic and clinical research. METHODS The PubMed and Web of Science databases were searched using the keywords dexmedetomidine and related brain diseases, although relevant articles from the last decade were included for detailed summarization and analysis. RESULTS Dexmedetomidine has shown strong neuroprotective effects, such as protection of the blood-brain barrier, decreased neuronal death, maintained hemodynamic stability, and reduced postoperative agitation and cognitive dysfunction. Furthermore, dexmedetomidine has been shown to exert various neuroprotective effects, including anti-inflammatory and antioxidative stress effects, modulation of autophagy, and reduction of apoptosis in cerebral diseases. CONCLUSIONS Dexmedetomidine acts as a neuroprotective agent against brain diseases during all phases of treatment. However, clinical trials with larger sample sizes are required to optimize dosage and dosing strategies.
Collapse
Affiliation(s)
- Zhenxing Tao
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Pengpeng Li
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Xudong Zhao
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
- Wuxi Neurosurgical InstituteWuxiChina
| |
Collapse
|
4
|
Lin Y, Wang D, Li B, Wang J, Xu L, Sun X, Ji K, Yan C, Liu F, Zhao Y. Targeting DRP1 with Mdivi-1 to correct mitochondrial abnormalities in ADOA+ syndrome. JCI Insight 2024; 9:e180582. [PMID: 38916953 PMCID: PMC11383607 DOI: 10.1172/jci.insight.180582] [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: 03/01/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Autosomal dominant optic atrophy plus (ADOA+) is characterized by primary optic nerve atrophy accompanied by a spectrum of degenerative neurological symptoms. Despite ongoing research, no effective treatments are currently available for this condition. Our study provided evidence for the pathogenicity of an unreported c.1780T>C variant in the OPA1 gene through patient-derived skin fibroblasts and an engineered HEK293T cell line with OPA1 downregulation. We demonstrate that OPA1 insufficiency promoted mitochondrial fragmentation and increased DRP1 expression, disrupting mitochondrial dynamics. Consequently, this disruption enhanced mitophagy and caused mitochondrial dysfunction, contributing to the ADOA+ phenotype. Notably, the Drp1 inhibitor, mitochondrial division inhibitor-1 (Mdivi-1), effectively mitigated the adverse effects of OPA1 impairment. These effects included reduced Drp1 phosphorylation, decreased mitochondrial fragmentation, and balanced mitophagy. Thus, we propose that intervening in DRP1 with Mdivi-1 could correct mitochondrial abnormalities, offering a promising therapeutic approach for managing ADOA+.
Collapse
Affiliation(s)
- Yan Lin
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dongdong Wang
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Busu Li
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiayin Wang
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ling Xu
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaohan Sun
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Kunqian Ji
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Shandong University, Qingdao, Shandong, China
- Brain Science Research Institute, Shandong University, Jinan, Shandong, China
| | - Fuchen Liu
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
5
|
Portelli K, Kandraju H, Ryu M, Shah PS. Efficacy and safety of dexmedetomidine for analgesia and sedation in neonates: a systematic review. J Perinatol 2024; 44:164-172. [PMID: 37845426 DOI: 10.1038/s41372-023-01802-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Opioids and benzodiazepines have historically been employed for pain relief; however, they are associated with detrimental long-term neurodevelopmental consequences. Dexmedetomidine, a highly selective alpha-2-adrenoreceptor agonist, has piqued interest as a viable alternative for neonates, owing to its potential analgesic and neuroprotective attributes. We conducted a systematic review to assess the efficacy and safety of dexmedetomidine utilization in neonates. We conducted a comprehensive search of Ovid, MEDLINE, EMBASE, PubMed, Cochrane, and CINAHL, spanning from January 2010 to September 2022. Our review encompassed six studies involving 252 neonates. Overall, dexmedetomidine may be effective in achieving sedation and analgesia. Furthermore, it may reduce the need for adjunctive sedation or analgesia, shorten the time to extubation, decrease the duration of mechanical ventilation, and accelerate the attainment of full enteral feeds. Notably, no significant adverse effects associated with dexmedetomidine were reported. Nevertheless, additional well-designed studies to establish both the efficacy and safety of dexmedetomidine in neonatal care are needed.
Collapse
Affiliation(s)
- Katelyn Portelli
- Department of Pediatrics, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Pediatrics, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Hemasree Kandraju
- Department of Pediatrics, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Michelle Ryu
- Library and knowledge services, Trillium Health Partners, Ontario, Canada
| | - Prakesh S Shah
- Department of Pediatrics, Mount Sinai Hospital, Toronto, ON, Canada.
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Zhang H, Tian Y, Yu W, Tong D, Ji Y, Qu X, Deng T, Li X, Xu Y. TMEM175 downregulation participates in impairment of the autophagy related lysosomal dynamics following neonatal hypoxic-ischemic brain injury. J Cell Physiol 2023; 238:2512-2527. [PMID: 37566721 DOI: 10.1002/jcp.31096] [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: 05/19/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023]
Abstract
The mechanism underlying long-term cognitive impairment caused by neonatal hypoxic-ischemic brain injury (HIBI) remains unclear. Autophagy is a closely related mechanism and may play a role in this process. We aimed to investigate the role of lysosomal transmembrane protein 175 (TMEM175) in the autophagy-lysosome pathway in neonatal rats with HIBI. A neonatal rat model of HIBI was established, hypoxia was induced, followed by left common carotid artery ligation. Expression levels of TMEM175 and the corresponding proteins involved in autophagy flux and the endolysosomal system fusion process were measured. Rats were administered TMEM175 plasmid via intracerebroventricular injection to induce overexpression. Brain damage and cognitive function were then assessed. TMEM175 was downregulated in the hippocampal tissue, and the autophagy-lysosome pathway was impaired following HIBI in neonatal rats. Overexpression of TMEM175 significantly mitigated neuronal injury and improved long-term cognitive and memory function in neonatal rats with HIBI. We found that improvement in the autophagy-lysosome pathway and endolysosomal system homeostasis, which are TMEM175 related, occurred via regulation of lysosomal membrane dynamic fusion. TMEM175 plays a critical role in maintaining the autophagy-lysosome pathway and endolysosomal homeostasis, contributing to the amelioration of neuronal injury and impaired long-term cognitive function following neonatal HIBI.
Collapse
Affiliation(s)
- Huiyi Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ye Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weiwei Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dongyi Tong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yichen Ji
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinrui Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianjiao Deng
- The First Clinical College, China Medical University, Shenyang, China
| | - Xinsheng Li
- The First Clinical College, China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Puls R, von Haefen C, Bührer C, Endesfelder S. Dexmedetomidine Protects Cerebellar Neurons against Hyperoxia-Induced Oxidative Stress and Apoptosis in the Juvenile Rat. Int J Mol Sci 2023; 24:ijms24097804. [PMID: 37175511 PMCID: PMC10178601 DOI: 10.3390/ijms24097804] [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: 04/03/2023] [Revised: 04/13/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
The risk of oxidative stress is unavoidable in preterm infants and increases the risk of neonatal morbidities. Premature infants often require sedation and analgesia, and the commonly used opioids and benzodiazepines are associated with adverse effects. Impairment of cerebellar functions during cognitive development could be a crucial factor in neurodevelopmental disorders of prematurity. Recent studies have focused on dexmedetomidine (DEX), which has been associated with potential neuroprotective properties and is used as an off-label application in neonatal units. Wistar rats (P6) were exposed to 80% hyperoxia for 24 h and received as pretreatment a single dose of DEX (5µg/kg, i.p.). Analyses in the immature rat cerebellum immediately after hyperoxia (P7) and after recovery to room air (P9, P11, and P14) included examinations for cell death and inflammatory and oxidative responses. Acute exposure to high oxygen concentrations caused a significant oxidative stress response, with a return to normal levels by P14. A marked reduction of hyperoxia-mediated damage was demonstrated after DEX pretreatment. DEX produced a much earlier recovery than in controls, confirming a neuroprotective effect of DEX on alterations elicited by oxygen stress on the developing cerebellum.
Collapse
Affiliation(s)
- Robert Puls
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
8
|
Puls R, von Haefen C, Bührer C, Endesfelder S. Protective Effect of Dexmedetomidine against Hyperoxia-Damaged Cerebellar Neurodevelopment in the Juvenile Rat. Antioxidants (Basel) 2023; 12:antiox12040980. [PMID: 37107355 PMCID: PMC10136028 DOI: 10.3390/antiox12040980] [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: 03/10/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Impaired cerebellar development of premature infants and the associated impairment of cerebellar functions in cognitive development could be crucial factors for neurodevelopmental disorders. Anesthetic- and hyperoxia-induced neurotoxicity of the immature brain can lead to learning and behavioral disorders. Dexmedetomidine (DEX), which is associated with neuroprotective properties, is increasingly being studied for off-label use in the NICU. For this purpose, six-day-old Wistar rats (P6) were exposed to hyperoxia (80% O2) or normoxia (21% O2) for 24 h after DEX (5 µg/kg, i.p.) or vehicle (0.9% NaCl) application. An initial detection in the immature rat cerebellum was performed after the termination of hyperoxia at P7 and then after recovery in room air at P9, P11, and P14. Hyperoxia reduced the proportion of Calb1+-Purkinje cells and affected the dendrite length at P7 and/or P9/P11. Proliferating Pax6+-granule progenitors remained reduced after hyperoxia and until P14. The expression of neurotrophins and neuronal transcription factors/markers of proliferation, migration, and survival were also reduced by oxidative stress in different manners. DEX demonstrated protective effects on hyperoxia-injured Purkinje cells, and DEX without hyperoxia modulated neuronal transcription in the short term without any effects at the cellular level. DEX protects hyperoxia-damaged Purkinje cells and appears to differentially affect cerebellar granular cell neurogenesis following oxidative stress.
Collapse
Affiliation(s)
- Robert Puls
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
9
|
Gatica S, Aravena C, Prado Y, Aravena D, Echeverría C, Santibanez JF, Riedel CA, Stehberg J, Simon F. Appraisal of the Neuroprotective Effect of Dexmedetomidine: A Meta-Analysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:163-181. [PMID: 37093427 DOI: 10.1007/978-3-031-26163-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Dexmedetomidine is an adrenergic receptor agonist that has been regarded as neuroprotective in several studies without an objective measure to it. Thus, the aim of this meta-analysis was to analyze and quantify the current evidence for the neuroprotective effects of dexmedetomidine in animals. The search was performed by querying the National Library of Medicine. Studies were included based on their language, significancy of their results, and complete availability of data on animal characteristics and interventions. Risk of bias was assessed using SYRCLE's risk of bias tool and certainty was assessed using the ARRIVE Guidelines 2.0. Synthesis was performed by calculating pooled standardized mean difference and presented in forest plots and tables. The number of eligible records included per outcome is the following: 22 for IL-1β, 13 for IL-6, 19 for apoptosis, 7 for oxidative stress, 7 for Escape Latency, and 4 for Platform Crossings. At the cellular level, dexmedetomidine was found protective against production of IL-1β (standardized mean difference (SMD) = - 4.3 [- 4.8; - 3.7]) and IL-6 (SMD = - 5.6 [- 6.7; - 4.6]), apoptosis (measured through TUNEL, SMD = - 6.0 [- 6.8; - 4.6]), and oxidative stress (measured as MDA production, SMD = - 2.0 [- 2.4; - 1.4]) exclusively in the central nervous system. At the organism level, dexmedetomidine improved behavioral outcomes measuring escape latency (SMD = - 2.4 [- 3.3; - 1.6]) and number of platform crossings (SMD = 9.1 [- 6.8; - 11.5]). No eligible study had high risk of bias and certainty was satisfactory for reproducibility in all cases. This meta-analysis highlights the complexity of adrenergic stimulation and sheds light into the mechanisms potentiated by dexmedetomidine, which could be exploited for improving current neuroprotective formulations.
Collapse
Affiliation(s)
- Sebastian Gatica
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| | - Cristobal Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Yolanda Prado
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cesar Echeverría
- Laboratory of Molecular Biology, Nanomedicine and Genomics, Faculty of Medicine, University of Atacama, Copiapo, Chile
| | - Juan F Santibanez
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
- Integrative Center for Biology and Applied Chemistry (CIBQA), Bernardo O'Higgins University, Santiago, Chile
| | - Claudia A Riedel
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Jimmy Stehberg
- Laboratory of Neurobiology, Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Chen X, Chen D, Chen P, Chen A, Deng J, Wei J, Zeng W, Zheng X. Dexmedetomidine Attenuates Apoptosis and Neurological Deficits by Modulating Neuronal NADPH Oxidase 2-Derived Oxidative Stress in Neonates Following Hypoxic Brain Injury. Antioxidants (Basel) 2022; 11:2199. [PMID: 36358571 PMCID: PMC9686745 DOI: 10.3390/antiox11112199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 08/31/2023] Open
Abstract
Hypoxic-ischemic brain injury is an important cause of neonatal neurological deficits. Our previous study demonstrated that dexmedetomidine (Dex) provided neuroprotection against neonatal hypoxic brain injury; however, the underlying mechanisms remain incompletely elucidated. Overactivation of NADPH oxidase 2 (NOX2) can cause neuronal apoptosis and neurological deficits. Hence, we aimed to investigate the role of neuronal NOX2 in Dex-mediated neuroprotection and to explore its potential mechanisms. Hypoxic injury was modeled in neonatal rodents in vivo and in cultured hippocampal neurons in vitro. Our results showed that pre- or post-treatment with Dex improved the neurological deficits and alleviated the hippocampal neuronal damage and apoptosis caused by neonatal hypoxia. In addition, Dex treatment significantly suppressed hypoxia-induced neuronal NOX2 activation; it also reduced oxidative stress, as evidenced by decreases in intracellular reactive oxygen species (ROS) production, malondialdehyde, and 8-hydroxy-2-deoxyguanosine, as well as increases in the antioxidant enzymatic activity of superoxide dismutase and glutathione peroxidase in neonatal rat hippocampi and in hippocampal neurons. Lastly, the posthypoxicneuroprotective action of Dex was almost completely abolished in NOX2-deficient neonatal mice and NOX2-knockdown neurons. In conclusion, our data demonstrated that neuronal NOX2-mediated oxidative stress is involved in the neuroprotection that Dex provides against apoptosis and neurological deficits in neonates following hypoxia.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Dongtai Chen
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Pinzhong Chen
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Andi Chen
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianhui Deng
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianjie Wei
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Co-Constructed Laboratory of “Belt and Road”, Fuzhou 350001, China
| |
Collapse
|
11
|
Yang W, Li H, Cheng Z, Lu Y, Li W, Feng J, Wang L, Cheng J. Dex modulates the balance of water-electrolyte metabolism by depressing the expression of AVP in PVN. Front Pharmacol 2022; 13:919032. [PMID: 36081946 PMCID: PMC9445239 DOI: 10.3389/fphar.2022.919032] [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: 04/17/2022] [Accepted: 07/11/2022] [Indexed: 12/01/2022] Open
Abstract
Dexmedetomidine (Dex) is a highly selective α2 adrenergic agonist used in clinical anesthesia. Studies have shown that Dex can act on the collecting duct and reduce the body’s water reabsorption, thereby increasing water discharge. However, the specific mechanism of Dex on water homeostasis remains unclear. The hypothalamus is the regulatory center of water and salt balance and secretes related neurochemical hormones, such as arginine vasopressin (AVP), to regulate the discharge of water and salt. The paraventricular nucleus (PVN) and supraoptic nucleus (SON) in the hypothalamus are also considered to be the key targets of the thirst loop. They are responsible for the secretion of AVP. The suprachiasmatic nucleus (SCN) is also one of the brain regions where AVP neurons are densely distributed in the hypothalamus. This study used C57BL/6J mice for behavior, immunofluorescence, and blood analysis experiments. Our results showed that Dex could not only depress the expression of AVP in the PVN but also reduce serum AVP concentration. The animal water intake was decreased without impairing the difference in food consumption and the urine excretion was enhanced after the intraperitoneal injection of Dex, while AVP supplementation restored the water intake and inhibited the urine excretion of mice in the Dex group. In addition, the renin-angiotensin-aldosterone system is vital to maintaining serum sodium concentration and extracellular volume. We found that serum sodium, serum chloride, serum aldosterone (ALD) concentration, and plasma osmolality were decreased in the Dex group, which inhibited water reabsorption, and the plasma osmolarity of mice in the Dex group supplemented with AVP was significantly higher than that in Dex group. We also found that Dex significantly increased the concentration of blood urea nitrogen and decreased the concentration of creatinine within the normal range of clinical indicators, indicating that there was no substantive lesion in the renal parenchyma. These results showed that Dex could modulate the balance of water-electrolyte metabolism by depressing the expression of AVP in PVN without impairing renal function.
Collapse
Affiliation(s)
- Wenzhi Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hao Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Zhongle Cheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - You Lu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wuli Li
- College and Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Jun Feng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liecheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Juan Cheng, ; Liecheng Wang,
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Juan Cheng, ; Liecheng Wang,
| |
Collapse
|
12
|
Zhao S, Wu W, Lin X, Shen M, Yang Z, Yu S, Luo Y. Protective effects of dexmedetomidine in vital organ injury: crucial roles of autophagy. Cell Mol Biol Lett 2022; 27:34. [PMID: 35508984 PMCID: PMC9066865 DOI: 10.1186/s11658-022-00335-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
Vital organ injury is one of the leading causes of global deaths. Accumulating studies have demonstrated that dexmedetomidine (DEX) has an outstanding protective effect on multiple organs for its antiinflammatory and antiapoptotic properties, while the underlying molecular mechanism is not clearly understood. Autophagy, an adaptive catabolic process, has been found to play a crucial role in the organ-protective effects of DEX. Herein, we present a first attempt to summarize all the evidence on the proposed roles of autophagy in the action of DEX protecting against vital organ injuries via a comprehensive review. We found that most of the relevant studies (17/24, 71%) demonstrated that the modulation of autophagy was inhibited under the treatment of DEX on vital organ injuries (e.g. brain, heart, kidney, and lung), but several studies suggested that the level of autophagy was dramatically increased after administration of DEX. Albeit not fully elucidated, the underlying mechanisms governing the roles of autophagy involve the antiapoptotic properties, inhibiting inflammatory response, removing damaged mitochondria, and reducing oxidative stress, which might be facilitated by the interaction with multiple associated genes (i.e., hypoxia inducible factor-1α, p62, caspase-3, heat shock 70 kDa protein, and microRNAs) and signaling cascades (i.e., mammalian target of rapamycin, nuclear factor-kappa B, and c-Jun N-terminal kinases pathway). The authors conclude that DEX hints at a promising strategy in the management of vital organ injuries, while autophagy is crucially involved in the protective effect of DEX.
Collapse
Affiliation(s)
- Shankun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Weizhou Wu
- Department of Urology, Maoming People's Hospital, Maoming, 525000, Guangdong, China
| | - Xuezheng Lin
- Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Maolei Shen
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Zhenyu Yang
- Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Sicong Yu
- Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Yu Luo
- Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China.
| |
Collapse
|
13
|
Ajoolabady A, Wang S, Kroemer G, Penninger JM, Uversky VN, Pratico D, Henninger N, Reiter RJ, Bruno A, Joshipura K, Aslkhodapasandhokmabad H, Klionsky DJ, Ren J. Targeting autophagy in ischemic stroke: From molecular mechanisms to clinical therapeutics. Pharmacol Ther 2021; 225:107848. [PMID: 33823204 PMCID: PMC8263472 DOI: 10.1016/j.pharmthera.2021.107848] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 01/18/2023]
Abstract
Stroke constitutes the second leading cause of death and a major cause of disability worldwide. Stroke is normally classified as either ischemic or hemorrhagic stroke (HS) although 87% of cases belong to ischemic nature. Approximately 700,000 individuals suffer an ischemic stroke (IS) in the US each year. Recent evidence has denoted a rather pivotal role for defective macroautophagy/autophagy in the pathogenesis of IS. Cellular response to stroke includes autophagy as an adaptive mechanism that alleviates cellular stresses by removing long-lived or damaged organelles, protein aggregates, and surplus cellular components via the autophagosome-lysosomal degradation process. In this context, autophagy functions as an essential cellular process to maintain cellular homeostasis and organismal survival. However, unchecked or excessive induction of autophagy has been perceived to be detrimental and its contribution to neuronal cell death remains largely unknown. In this review, we will summarize the role of autophagy in IS, and discuss potential strategies, particularly, employment of natural compounds for IS treatment through manipulation of autophagy.
Collapse
Affiliation(s)
- Amir Ajoolabady
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Shuyi Wang
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; School of Medicine Shanghai University, Shanghai 200444, China
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria; Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts, Worcester, Massachusetts, USA; Department of Psychiatry, University of Massachusetts, Worcester, Massachusetts, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Askiel Bruno
- Department of Neurology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Kaumudi Joshipura
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Center for Clinical Research and Health Promotion, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936-5067, Puerto Rico
| | | | - Daniel J Klionsky
- Life Sciences Institute and Departments of Molecular, Cellular and Developmental Biology and Biological Chemistry, University of Michigan, Ann Arbor 48109, USA.
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington Seattle, Seattle, WA 98195, USA; Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
14
|
Feng X, Ma W, Zhu J, Jiao W, Wang Y. Dexmedetomidine alleviates early brain injury following traumatic brain injury by inhibiting autophagy and neuroinflammation through the ROS/Nrf2 signaling pathway. Mol Med Rep 2021; 24:661. [PMID: 34278508 PMCID: PMC8335733 DOI: 10.3892/mmr.2021.12300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem and a major cause of mortality and disability that imposes a substantial economic burden worldwide. Dexmedetomidine (DEX), a highly selective α-2-adrenergic receptor agonist that functions as a sedative and analgesic with minimal respiratory depression, has been reported to alleviate early brain injury (EBI) following traumatic brain injury by reducing reactive oxygen species (ROS) production, apoptosis and autophagy. Autophagy is a programmed cell death mechanism that serves a vital role in neuronal cell death following TBI. However, the precise role of autophagy in DEX-mediated neuroprotection following TBI has not been confirmed. The present study aimed to investigate the neuroprotective effects and potential molecular mechanisms of DEX in TBI-induced EBI by regulating neural autophagy in a C57BL/6 mouse model. Mortality, the neurological score, brain water content, neuroinflammatory cytokine levels, ROS production, malondialdehyde levels and neuronal death were evaluated by TUNEL staining, Evans blue extravasation, ELISA, analysis of ROS/lipid peroxidation and western blotting. The results showed that DEX treatment markedly increased the survival rate and neurological score, increased neuron survival, decreased the expression of the LC3, Beclin-1 and NF-κB proteins, as well as the cytokines IL-1β, IL-6 and TNF-α, which indicated that DEX-mediated inhibition of autophagy and neuroinflammation ameliorated neuronal death following TBI. The neuroprotective capacity of DEX is partly dependent on the ROS/nuclear factor erythroid 2-related factor 2 signaling pathway. Taken together, the results of the present study indicated that DEX improves neurological outcomes in mice and reduces neuronal death by protecting against neural autophagy and neuroinflammation.
Collapse
Affiliation(s)
- Xiaoyan Feng
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Weiwei Ma
- Clinical Medicine Five‑Year Program, 11 Class, 2019 Grade, Wannan Medical College, Wuhu, Jiangsu 241002, P.R. China
| | - Jie Zhu
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Wei Jiao
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Yuhai Wang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu 214044, P.R. China
| |
Collapse
|