1
|
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
|
2
|
Seong H, Jeong D, Kim EH, Yoon KS, Na D, Yoon SZ, Cho JE. MicroRNA-323-5p Involved in Dexmedetomidine Preconditioning Impart Neuroprotection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1518. [PMID: 37763638 PMCID: PMC10532972 DOI: 10.3390/medicina59091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives: Cerebral ischemia is one of the major preoperative complications. Dexmedetomidine is a well-known sedative-hypnotic agent that has potential organ-protective effects. We examine the miRNAs associated with preconditioning effects of dexmedetomidine in cerebral ischemia. Materials and Methods: Transient infarcts were induced in mice via reperfusion after temporary occlusion of one side of the middle cerebral artery. A subset of these mice was exposed to dexmedetomidine prior to cerebral infarction and miRNA profiling of the whole brain was performed. We administered dexmedetomidine and miRNA-323-5p mimic/inhibitor to oxygen-glucose deprivation/reoxygenation astrocytes. Additionally, we administered miR-323-5p mimic and inhibitor to mice via intracerebroventricular injection 2 h prior to induction of middle cerebral artery occlusion. Results: The infarct volume was significantly lower in the dexmedetomidine-preconditioned mice. Analysis of brain samples revealed an increased expression of five miRNAs and decreased expression of three miRNAs in the dexmedetomidine-pretreated group. The viability of cells significantly increased and expression of miR-323-5p was attenuated in the dexmedetomidine-treated oxygen-glucose deprivation/reoxygenation groups. Transfection with anti-miR-323-5p contributed to increased astrocyte viability. When miRNA-323-5p was injected intraventricularly, infarct volume was significantly reduced when preconditioned with the miR-323-5p inhibitor compared with mimic and negative control. Conclusions: Dexmedetomidine has a protective effect against transient neuronal ischemia-reperfusion injury and eight specific miRNAs were profiled. Also, miRNA-323-5p downregulation has a cell protective effect under ischemic conditions both in vivo and in vitro. Our findings suggest the potential of the miR-323-5p inhibitor as a therapeutic agent against cerebral infarction.
Collapse
Affiliation(s)
- Hyunyoung Seong
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Daun Jeong
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Eung Hwi Kim
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Seob Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Donghyun Na
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Zhoo Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jang Eun Cho
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
3
|
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
|
4
|
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; 24:7804. [PMID: 37175511 PMCID: PMC10178601 DOI: 10.3390/ijms24097804] [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: 04/03/2023] [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; (R.P.); (C.B.)
| | - 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; (R.P.); (C.B.)
| | - Stefanie Endesfelder
- Department of Neonatology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (R.P.); (C.B.)
| |
Collapse
|
5
|
Hu Y, Zhou H, Zhang H, Sui Y, Zhang Z, Zou Y, Li K, Zhao Y, Xie J, Zhang L. The neuroprotective effect of dexmedetomidine and its mechanism. Front Pharmacol 2022; 13:965661. [PMID: 36204225 PMCID: PMC9531148 DOI: 10.3389/fphar.2022.965661] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective α2 receptor agonist that is routinely used in the clinic for sedation and anesthesia. Recently, an increasing number of studies have shown that DEX has a protective effect against brain injury caused by traumatic brain injury (TBI), subarachnoid hemorrhage (SAH), cerebral ischemia and ischemia–reperfusion (I/R), suggesting its potential as a neuroprotective agent. Here, we summarized the neuroprotective effects of DEX in several models of neurological damage and examined its mechanism based on the current literature. Ultimately, we found that the neuroprotective effect of DEX mainly involved inhibition of inflammatory reactions, reduction of apoptosis and autophagy, and protection of the blood–brain barrier and enhancement of stable cell structures in five way. Therefore, DEX can provide a crucial advantage in neurological recovery for patients with brain injury. The purpose of this study was to further clarify the neuroprotective mechanisms of DEX therefore suggesting its potential in the clinical management of the neurological injuries.
Collapse
Affiliation(s)
- Yijun Hu
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
- Graduate School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong Zhou
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Huanxin Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yunlong Sui
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhen Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yuntao Zou
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Kunquan Li
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yunyi Zhao
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Jiangbo Xie
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Lunzhong Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
- *Correspondence: Lunzhong Zhang,
| |
Collapse
|
6
|
Burlacu CC, Neag MA, Mitre AO, Sirbu AC, Badulescu AV, Buzoianu AD. The Role of miRNAs in Dexmedetomidine's Neuroprotective Effects against Brain Disorders. Int J Mol Sci 2022; 23:5452. [PMID: 35628263 PMCID: PMC9141783 DOI: 10.3390/ijms23105452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
There are limited neuroprotective strategies for various central nervous system conditions in which fast and sustained management is essential. Neuroprotection-based therapeutics have become an intensively researched topic in the neuroscience field, with multiple novel promising agents, from natural products to mesenchymal stem cells, homing peptides, and nanoparticles-mediated agents, all aiming to significantly provide neuroprotection in experimental and clinical studies. Dexmedetomidine (DEX), an α2 agonist commonly used as an anesthetic adjuvant for sedation and as an opioid-sparing medication, stands out in this context due to its well-established neuroprotective effects. Emerging evidence from preclinical and clinical studies suggested that DEX could be used to protect against cerebral ischemia, traumatic brain injury (TBI), spinal cord injury, neurodegenerative diseases, and postoperative cognitive disorders. MicroRNAs (miRNAs) regulate gene expression at a post-transcriptional level, inhibiting the translation of mRNA into functional proteins. In vivo and in vitro studies deciphered brain-related miRNAs and dysregulated miRNA profiles after several brain disorders, including TBI, ischemic stroke, Alzheimer's disease, and multiple sclerosis, providing emerging new perspectives in neuroprotective therapy by modulating these miRNAs. Experimental studies revealed that some of the neuroprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, such as lipopolysaccharides induced neuroinflammation, β-amyloid induced dysfunction, brain ischemic-reperfusion injury, and anesthesia-induced neurotoxicity models. This review aims to outline the neuroprotective mechanisms of DEX in brain disorders by modulating miRNAs. We address the neuroprotective effects of DEX by targeting miRNAs in modulating ischemic brain injury, ameliorating the neurotoxicity of anesthetics, reducing postoperative cognitive dysfunction, and improving the effects of neurodegenerative diseases.
Collapse
Affiliation(s)
- Codrin-Constantin Burlacu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandru-Constantin Sirbu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Vlad Badulescu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
7
|
Unchiti K, Leurcharusmee P, Samerchua A, Pipanmekaporn T, Chattipakorn N, Chattipakorn SC. The potential role of dexmedetomidine on neuroprotection and its possible mechanisms: Evidence from in vitro and in vivo studies. Eur J Neurosci 2021; 54:7006-7047. [PMID: 34561931 DOI: 10.1111/ejn.15474] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
Neurological disorders following brain injuries and neurodegeneration are on the rise worldwide and cause disability and suffering in patients. It is crucial to explore novel neuroprotectants. Dexmedetomidine, a selective α2-adrenoceptor agonist, is commonly used for anxiolysis, sedation and analgesia in clinical anaesthesia and critical care. Recent studies have shown that dexmedetomidine exerts protective effects on multiple organs. This review summarized and discussed the current neuroprotective effects of dexmedetomidine, as well as the underlying mechanisms. In preclinical studies, dexmedetomidine reduced neuronal injury and improved functional outcomes in several models, including hypoxia-induced neuronal injury, ischaemic-reperfusion injury, intracerebral haemorrhage, post-traumatic brain injury, anaesthetic-induced neuronal injury, substance-induced neuronal injury, neuroinflammation, epilepsy and neurodegeneration. Several mechanisms are associated with the neuroprotective function of dexmedetomidine, including neurotransmitter regulation, inflammatory response, oxidative stress, apoptotic pathway, autophagy, mitochondrial function and other cell signalling pathways. In summary, dexmedetomidine has the potential to be a novel neuroprotective agent for a wide range of neurological disorders.
Collapse
Affiliation(s)
- Kantarakorn Unchiti
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prangmalee Leurcharusmee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Artid Samerchua
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Tanyong Pipanmekaporn
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
8
|
Sun K, Zhang J, Yang Q, Zhu J, Zhang X, Wu K, Li Z, Xie W, Luo X. Dexmedetomidine exerts a protective effect on ischemic brain injury by inhibiting the P2X7R/NLRP3/Caspase-1 signaling pathway. Brain Res Bull 2021; 174:11-21. [PMID: 33991606 DOI: 10.1016/j.brainresbull.2021.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/29/2022]
Abstract
Dexmedetomidine (Dex) has been suggested to exert a protective function in ischemic brain injury. In this study, we aimed to elucidate the intrinsic mechanisms of Dex in regulating microglia pyroptosis in ischemic brain injury via the purinergic 2X7 receptor (P2X7R)/NLRP3/Caspase-1 signaling pathway. First, permanent middle cerebral artery occlusion (p-MCAO) rat model was established, followed by the measurement of behavioral deficit, neuronal injury, the volume of brain edema and the infarct size. Dex treatment was suggested to alleviate the neurological deficits in p-MCAO rats and reduce the brain water content and infarct size. Additionally, rat microglia were cultured in vitro and a model of oxygen and glucose (OGD) was established. Microglia cell activity and ultrastructure were detected. Dex could increase cell activity and reduce LDH activity, partially reversing the changes in cell morphology. Furthermore, the activation of P2X7R/NLRP3/Caspase-1 pathway was tested. The obtained findings indicated Dex suppressed microglial pyroptosis by inhibiting the P2X7R/NLRP3/Caspase-1 pathway. Inhibition of P2X7R or NLRP3 could inhibit Caspase-1 p10 expression, improve cell activity, and reduce LDH activity. The same result was verified in vivo experiments. This study indicated that Dex inhibited microglia pyroptosis by blocking the P2X7R/NLRP3/Caspase-1 pathway, thus playing a protective role against ischemic brain injury.
Collapse
Affiliation(s)
- Ke Sun
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Jiangang Zhang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Qingcheng Yang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China.
| | - Jinzhao Zhu
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Xiangdong Zhang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Kun Wu
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Zhenhua Li
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Weizheng Xie
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Xue Luo
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| |
Collapse
|
9
|
Mechanism of dexmedetomidine regulating osteogenesis-angiogenesis coupling through the miR-361-5p/VEGFA axis in postmenopausal osteoporosis. Life Sci 2021; 275:119273. [PMID: 33631172 DOI: 10.1016/j.lfs.2021.119273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022]
Abstract
AIMS Postmenopausal osteoporosis (PMOP) is a growing health problem affecting many postmenopausal women. This study intended to identify the role of dexmedetomidine (Dex) in osteoporosis (OP). MAIN METHODS Microarray analysis was performed for the gene expression profiles of PMOP patients and postmenopausal healthy volunteers, and the most differentially expressed microRNA (miR)-361-5p was verified in clinic, and its diagnostic value in PMOP patients was analyzed. After establishment of OP model by ovariectomy, Dex treatment and overexpression of miR-361-5p or vascular endothelial growth factor A (VEGFA) were performed in OP rats or isolated bone marrow mesenchymal stem cells (BMSCs). Bone mineral density (BMD) related indexes and levels of osteogenesis-angiogenesis related genes were measured. The apoptosis and osteogenic differentiation of BMSCs were detected. After human umbilical vein endothelial cells (HUVECs) and BMSCs were cocultured, the angiogenesis of BMSCs was detected by Matrigel-based angiogenesis experiment. KEY FINDINGS miR-361-5p was highly expressed in PMOP patients and OP rats, with good diagnostic effect on PMOP. After Dex treatment, the expressions of miR-361-5p, VEGFA, BMD related indexes were increased in OP rats. In BMSCs, level of osteogenesis-angiogenesis related genes were increased after adding Dex, and the apoptosis was decreased after coculture of HUVECs and BMSCs. miR-361-5p could target VEGFA. After miR-361-5p overexpression + Dex treatment, the indexes related to osteogenesis and angiogenesis in OP rats and BMSCs were decreased, which were reversed after further overexpressing VEGFA. SIGNIFICANCE Dex can enhance VEGFA by inhibiting miR-361-5p, and then promote osteogenesis-angiogenesis, thus providing potential targets for PMOP treatment.
Collapse
|
10
|
Dexmedetomidine post-conditioning ameliorates long-term neurological outcomes after neonatal hypoxic ischemia: The role of autophagy. Life Sci 2021; 270:118980. [PMID: 33428879 DOI: 10.1016/j.lfs.2020.118980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypoxic-ischemic brain injury (HIBI) is a major cause of mortality in neonates and can cause long-term neurological sequelae. Excessive autophagy caused by HI may cause neuronal death. Dexmedetomidine was reported neuroprotective against HIBI. Therefore, in the present study, the autophagy-related mechanisms underlying the protective effects of dexmedetomidine against cerebral HI in neonatal rats were investigated. METHODS In the present study, the expression of autophagy-related proteins microtubule-associated protein 1 light chain 3 (LC3) B-II and Beclin1, neuronal and microglia autophagy levels, the myelin basic protein (MBP) expression, long-term neuronal density ratio, and long-term behavioral prognosis in HIBI model were investigated by ligating the left common carotid artery in neonatal rats, followed by 2-h hypoxia. RESULTS Dexmedetomidine inhibited the overactivated autophagy of hippocampal neurons and microglia after HI. In addition, dexmedetomidine inhibited neuronal density decrease and axon demyelination after HI-induced overactivated autophagy. Lastly, dexmedetomidine improved the long-term neurological prognosis and was reversed by the autophagy agonist rapamycin. CONCLUSION The protective effects of dexmedetomidine on HI neonatal rats were evidenced by inhibition of excessive autophagy of neurons and microglia, thereby reducing the decline of long-term neuronal density and axon demyelination as well as improving long-term learning cognitive function.
Collapse
|
11
|
Fang H, Li HF, Yan JY, Yang M, Zhang JP. Dexmedetomidine-up-regulated microRNA-381 exerts anti-inflammatory effects in rats with cerebral ischaemic injury via the transcriptional factor IRF4. J Cell Mol Med 2020; 25:2098-2109. [PMID: 33314611 PMCID: PMC7882963 DOI: 10.1111/jcmm.16153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Dexmedetomidine (Dex) possesses analgesic and anaesthetic values and reported being used in cerebral ischaemic injury therapeutics. Accumulating studies have determined the effect of microRNAs (miRNAs) on the cerebral ischaemic injury. Thus, the present study aimed to unravel the molecular mechanism of miR-381 and Dex in cerebral ischaemic injury. For this purpose, the cerebral ischaemic injury rat model was established by induction of middle cerebral artery occlusion (MCAO) and expression of miR-381 and IRF4 was determined. Thereafter, MCAO rats were treated with Dex, miR-381 mimic, miR-381 inhibitor and oe-IRF4 respectively, followed by evaluation of neurological function. Furthermore, neuron cells were isolated from the hippocampus of rats and subjected to oxygen-glucose deprivation (OGD). Then, OGD-treated neuron cells and primary neuron cells were examined by gain- and loss-of-function assay. Neuron cell apoptosis was detected using TUNEL staining and flow cytometry. The correlation between interferon regulatory factor 4 (IRF4) and interleukin (IL)-9 was detected. Our results showed down-regulated miR-38 and up-regulated IRF4 in MCAO rats. Besides, IRF4 was targeted by miR-381 in neuron cells. Dex and overexpressed miR-381, or silenced IRF4 improved the neurological function and inhibited neuron cell apoptosis in MCAO rats. Additionally, in MCAO rats, Dex was found to increase the miR-381 expression and reduced IRF4 expression to decrease the IL-9 expression, which suppressed the inflammatory response and cell apoptosis both in vivo and in vitro. Importantly, our study demonstrated that Dex elevated the expression of miR-381, which ultimately results in the inhibition of inflammation response in rats with cerebral ischaemic injury.
Collapse
Affiliation(s)
- Hua Fang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Hua-Feng Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jian-Yong Yan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Miao Yang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Jian-Ping Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| |
Collapse
|
12
|
Zhong Y, Li YP, Yin YQ, Hu BL, Gao H. Dexmedetomidine inhibits pyroptosis by down-regulating miR-29b in myocardial ischemia reperfusion injury in rats. Int Immunopharmacol 2020; 86:106768. [PMID: 32679539 DOI: 10.1016/j.intimp.2020.106768] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Dexmedetomidine (DEX) was reported to protect heart against ischemic-reperfusion (IR) but the mechanism herein remains elusive. This study aims to explore the mechanism of DEX on pyroptosis induced by myocardial ischemic reperfusion (MIR). METHODS MIR rat models were established and injected DEX or miR-29b agomir/antagomir separately. The possible effect of DEX or miR-29b on myocardial cells was assessed according to measurement on creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), interleukin-1β (IL-1β) and interleukin-18 (IL-18), myocardial infarction size, myocardial injury and apoptosis. Western blot determined the expression levels of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC) and cleaved-caspase-1. Hypoxia/reoxygenation (H/R) cell model was established. The lactate dehydrogenase (LDH) content released by myocardial cells was examined. The relation between miR-29b and FoxO3a was confirmed by dual luciferase reporter gene assay. FoxO3a or ARC level was elevated in H/R myocardial cells to detect its effect on pyroptosis. RESULTS MIR rat models were successfully established, in which cell pyroptosis was triggered as evidenced by increased expression levels of NLRP3, ASC and cleaved-caspase-1. Rats with DEX precondition had attenuated cell pyroptosis and ameliorated inflammatory response. FoxO3a was a target of miR-29b. MiR-29b agomir or miR-29b antagomir could inhibit or promote the protective effect of DEX on MIR. Overexpression of FoxO3a/ARC axis could suppress myocardial pyroptosis induced by H/R. CONCLUSION DEX could ameliorate MIR injury (MIRI) and H/R injury in rats and inhibit H/R induced pyroptosis in myocardial cells via down-regulating miR-29b to activate FoxO3a/ARC axis.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yi-Ping Li
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yong-Qiang Yin
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Bai-Long Hu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Hong Gao
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| |
Collapse
|
13
|
MicroRNA-29b-3p aggravates 1,2-dichloroethane-induced brain edema by targeting aquaporin 4 in Sprague-Dawley rats and CD-1 mice. Toxicol Lett 2020; 319:160-167. [DOI: 10.1016/j.toxlet.2019.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022]
|
14
|
Fang H, Li HF, Yang M, Wang RR, Wang QY, Zheng PC, Zhang FX, Zhang JP. microRNA-128 enhances neuroprotective effects of dexmedetomidine on neonatal mice with hypoxic-ischemic brain damage by targeting WNT1. Biomed Pharmacother 2019; 113:108671. [PMID: 30875657 DOI: 10.1016/j.biopha.2019.108671] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Hypoxic-ischemic brain damage (HIBD) is a major cause of acute mortality and chronic neurological morbidity in infants and children. Dexmedetomidine (DEX) is an effective choice in HIBD treatment. Recent findings have revealed that microRNA-128 (miR-128) is implicated in cerebral ischemia reperfusion. Hence, this study aimed to investigate the role of miR-128 in HIBD. METHODS HIBD models of neonatal mice were established. HIBD mice were treated with DEX, and injected with agomir (ago)-miR-128 or antagomir (anti)-miR-128 into the lateral ventricles to explore the influence of miR-128 on the neuroprotective effects of DEX on HIBD. Subsequently, the mice body weight, left/right (L/R) brain weight ratio, left-brain water content as well as learning and memory abilities were measured. Furthermore, the pathological changes of brain tissues and apoptosis rate of nerve cells were determined. The potential relationship between miR-128 and WNT1 was analyzed. RESULTS Over-expression of miR-128 caused an increase in mouse body weight, L/R brain weight ratio, and learning and memory abilities, while led to a decline in left-brain water content, brain tissue injury and apoptosis rate of nerve cells in DEX-treated HIBD mice. WNT1 was targeted and negatively regulated by miR-128. Silencing of WNT1 exerted the same effect as miR-128 on enhancing the neuroprotective effect of DEX on HIBD mice. CONCLUSION Collectively, miR-128 enhanced neuroprotective effect of DEX on HIBD neonatal mice by inhibiting WNT1.
Collapse
Affiliation(s)
- Hua Fang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, 550002, PR China; Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, PR China
| | - Hua-Feng Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Miao Yang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, 550002, PR China; Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, PR China
| | - Ru-Rong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Quan-Yun Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Peng-Cheng Zheng
- Guizhou University Research Center for Analysis of Drugs and Metabolites, Guizhou University, Guiyang, 550025, PR China
| | - Fang-Xiang Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, 550002, PR China; Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, PR China
| | - Jian-Ping Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, 550002, PR China; Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, PR China.
| |
Collapse
|
15
|
Identification of Candidate Genes and Pathways in Dexmedetomidine-Induced Cardioprotection in the Rat Heart by Bioinformatics Analysis. Int J Mol Sci 2019; 20:ijms20071614. [PMID: 30939728 PMCID: PMC6480577 DOI: 10.3390/ijms20071614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022] Open
Abstract
Dexmedetomidine (DEX), a highly selective alpha2 adrenergic receptor agonist, directly protects hearts against ischemia/reperfusion (I/R) injury. However, the detailed mechanism has not been fully elucidated. We studied differentially expressed mRNAs and miRNAs after DEX administration in rat hearts by comprehensive analysis. Additionally, bioinformatics analysis was applied to explore candidate genes and pathways that might play important roles in DEX-induced cardioprotection. The results of microarray analysis showed that 165 mRNAs and 6 miRNAs were differentially expressed after DEX administration. Through bioinformatics analysis using differentially expressed mRNAs, gene ontology (GO) terms including MAP kinase tyrosine/serine/threonine phosphatase activity and pathways including the p53 pathway were significantly enriched in the down-regulated mRNAs. Dusp1 and Atm were associated with the GO term of MAP kinase tyrosine/serine/threonine phosphatase activity and the p53 pathway, respectively. On the other hand, no significant pathway was found in the target mRNAs of deregulated miRNAs. The results indicated some possible key genes and pathways that seem to be of significance in DEX-induced cardioprotection, although miRNAs seem to be unlikely to contribute to cardioprotection induced by DEX.
Collapse
|
16
|
Alam A, Hana Z, Jin Z, Suen KC, Ma D. Surgery, neuroinflammation and cognitive impairment. EBioMedicine 2018; 37:547-556. [PMID: 30348620 PMCID: PMC6284418 DOI: 10.1016/j.ebiom.2018.10.021] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/25/2022] Open
Abstract
Trauma experienced during surgery can contribute to the development of a systemic inflammatory response that can cause multi-organ dysfunction or even failure. Post-surgical neuroinflammation is a documented phenomenon that results in synaptic impairment, neuronal dysfunction and death, and impaired neurogenesis. Various pro-inflammatory cytokines, such as TNFα, maintain a state of chronic neuroinflammation, manifesting as post-operative cognitive dysfunction and post-operative delirium. Furthermore, elderly patients with post-operative cognitive dysfunction or delirium are three times more likely to experience permanent cognitive impairment or dementia. We conducted a narrative review, considering evidence extracted from various databases including Pubmed, MEDLINE and EMBASE, as well as journals and book reference lists. We found that further pre-clinical and well-powered clinical studies are required to delineate the precise pathogenesis of post-operative delirium and cognitive dysfunction. Despite the burden of post-operative neurological sequelae, clinical studies investigating therapeutic agents, such as dexmedetomidine, ibuprofen and statins, have yielded conflicting results. In addition, evidence supporting novel therapeutic avenues, such as nicotinic and HMGB-1 targeting and remote ischaemic pre-conditioning, is limited and necessitates further investigation.
Collapse
Affiliation(s)
- Azeem Alam
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Zac Hana
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Zhaosheng Jin
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Ka Chun Suen
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|