1
|
Hou Y, Ye W, Tang Z, Li F. Anesthetics in pathological cerebrovascular conditions. J Cereb Blood Flow Metab 2025; 45:32-47. [PMID: 39450477 PMCID: PMC11563546 DOI: 10.1177/0271678x241295857] [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: 05/05/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
The increasing prevalence of pathological cerebrovascular conditions, including stroke, hypertensive encephalopathy, and chronic disorders, underscores the importance of anesthetic considerations for affected patients. Preserving cerebral oxygenation and blood flow during anesthesia is paramount to prevent neurological deterioration. Furthermore, protecting vulnerable neurons from damage is crucial for optimal outcomes. Recent research suggests that anesthetic agents may provide a potentially therapeutic approach for managing pathological cerebrovascular conditions. Anesthetics target neural mechanisms underlying cerebrovascular dysfunction, thereby modulating neuroinflammation, protecting neurons against ischemic injury, and improving cerebral hemodynamics. However, optimal strategies regarding mechanisms, dosage, and indications remain uncertain. This review aims to clarify the physiological effects, mechanisms of action, and reported neuroprotective benefits of anesthetics in patients with various pathological cerebrovascular conditions. Investigating anesthetic effects in cerebrovascular disease holds promise for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuhui Hou
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Ye
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Ziyuan Tang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
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
|
3
|
Cheng S, Sun X, Li Y, Dong Y. Evaluation of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α in preterm infants with brain injury. Ital J Pediatr 2024; 50:172. [PMID: 39256844 PMCID: PMC11389426 DOI: 10.1186/s13052-024-01744-4] [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: 12/27/2023] [Accepted: 08/31/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Prematurity-related brain injury is a common and serious complication that has long-term effects on the survival and development of affected infants. Currently, the roles of certain biomarkers such as the protein hydrolysis product SBDP145, melatonin, soluble lectin-like oxidized low-density lipoprotein receptor-1 (sLOX-1), high mobility group box 1 protein (HMGB1), and hypoxia-inducible factor 1-alpha (HIF-1α) in prematurity-related brain injury remain not fully elucidated. Our study aims to assess the significance of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α in preterm infants with brain injury. METHODS 135 preterm infants admitted to our hospital from January 2020 to February 2022 were selected and divided into 78 cases in a prematurity-associated brain injury group, and 57 cases in another group of preterm infants without brain injury or other diseases according to the magnetic resonance imaging results. The levels of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α in the two groups were analyzed. The serum concentrations of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α in newborns with different severity of ventricular hemorrhage were observed, and the levels of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α in those with different severity of white matter brain injury were compared. RESULTS The levels of SBDP145, sLOX-1, HMGB1 and HIF-1α were significantly higher in the preterm combined brain injury group than in the preterm group, and melatonin levels were significantly lower than in the preterm group(P < 0.05). The levels of SBDP145, sLOX-1, HMGB1 and HIF-1α were higher in the moderate to severe group and melatonin levels were lower in the mild group of newborns with ventricular hemorrhage (P < 0.05). The levels of SBDP145, sLOX-1, HMGB1 and HIF-1α were higher in the moderate-severe group and melatonin levels were lower in the mild group in newborns with cerebral white matter injury (P < 0.05). The independent variables were SBDP145, melatonin, sLOX-1, HMGB1, HIF-1α, and the dependent variable was the prognosis of neonates with brain injury. Univariate logistic regression analysis and multivariate logistic regression analysis were performed. The results showed that the influencing factors of newborns with brain injury were SBDP145, melatonin, sLOX-1, HMGB1, HIF-1α. CONCLUSION The levels of SBDP145, melatonin, sLOX-1, HMGB1 and HIF-1α were highly expressed in preterm newborns with brain injury, and the levels were higher when the condition of the newborns was more severe. These findings suggest the potential clinical utility of these biomarkers in predicting and monitoring brain injury in preterm infants, which could aid in early intervention and improve long-term outcomes.
Collapse
Affiliation(s)
- Sisi Cheng
- Department of Pediatrics, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shijiazhuang, Hebei, 050000, China
| | - Xiao Sun
- Department of Pediatrics, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shijiazhuang, Hebei, 050000, China
| | - Yanyan Li
- Department of Pediatrics, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shijiazhuang, Hebei, 050000, China
| | - Yan Dong
- Department of Pediatrics, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
4
|
Yang LN, Sun Y, Wang YZ, Wang J, Qi YS, Mu SS, Liu YP, Zhang ZQ, Chen ZM, Wang XJ, Xie WX, Wei CW, Wang Y, Wu AS. Effect of Postoperative Prolonged sedation with Dexmedetomidine after successful reperfusion with Endovascular Thrombectomy on long-term prognosis in patients with acute ischemic stroke (PPDET): study protocol for a randomized controlled trial. Trials 2024; 25:166. [PMID: 38439027 PMCID: PMC10913237 DOI: 10.1186/s13063-024-08015-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Endovascular thrombectomy (EVT) is a standard treatment for acute ischemic stroke (AIS) with large vessel occlusion. Hypertension and increased blood pressure variability within the first 24 h after successful reperfusion are related to a higher risk of symptomatic intracerebral hemorrhage and higher mortality. AIS patients might suffer from ischemia-reperfusion injury following reperfusion, especially within 24 h. Dexmedetomidine (DEX), a sedative commonly used in EVT, can stabilize hemodynamics by inhibiting the sympathetic nervous system and alleviate ischemia-reperfusion injury through anti-inflammatory and antioxidative properties. Postoperative prolonged sedation for 24 h with DEX might be a potential pharmacological approach to improve long-term prognosis after EVT. METHODS This single-center, open-label, prospective, randomized controlled trial will include 368 patients. The ethics committee has approved the protocol. After successful reperfusion (modified thrombolysis in cerebral infarction scores 2b-3, indicating reperfusion of at least 50% of the affected vascular territory), participants are randomly assigned to the intervention or control group. In the intervention group, participants will receive 0.1~1.0 μg/kg/h DEX for 24 h. In the control group, participants will receive an equal dose of saline for 24 h. The primary outcome is the functional outcome at 90 days, measured with the categorical scale of the modified Rankin Scale, ranging from 0 (no symptoms) to 6 (death). The secondary outcome includes (1) the changes in stroke severity between admission and 24 h and 7 days after EVT, measured by the National Institute of Health Stroke Scale (ranging from 0 to 42, with higher scores indicating greater severity); (2) the changes in ischemic penumbra volume/infarct volume between admission and 7 days after EVT, measured by neuroimaging scan; (3) the length of ICU/hospital stay; and (4) adverse events and the all-cause mortality rate at 90 days. DISCUSSION This randomized clinical trial is expected to verify the hypothesis that postoperative prolonged sedation with DEX after successful reperfusion may promote the long-term prognosis of patients with AIS and may reduce the related socio-economic burden. TRIAL REGISTRATION ClinicalTrials.gov NCT04916197. Prospectively registered on 7 June 2021.
Collapse
Affiliation(s)
- Li-Na Yang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yu-Zhu Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Jing Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yi-Sha Qi
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Shan-Shan Mu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yun-Peng Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Zi-Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Zi-Mo Chen
- Department of Neurology, Beijing Tian-tan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xiao-Jie Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Wu-Xiang Xie
- Peking University Clinical Research Institute, Peking University Health Science Center, Beijing, 101125, People's Republic of China
| | - Chang-Wei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
| | - An-Shi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
| |
Collapse
|
5
|
Chen B, Di B. Endogenous Ligands of TLR4 in Microglia: Potential Targets for Related Neurological Diseases. Curr Drug Targets 2024; 25:953-970. [PMID: 39234911 DOI: 10.2174/0113894501316051240821060249] [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: 04/08/2024] [Revised: 07/09/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Chronic inflammation mediated by microglia is a cause of some neuroinflammatory diseases. TLR4, a natural immune receptor on microglia, plays an important role in the occurrence of inflammation and the process of diseases. TLR4 can be activated by a variety of ligands to trigger inflammatory responses, including endogenous ligands HMGB1, S100A8/9, Heme, and Fetuin-A. As ligands derived from the body itself, they have the ability to bind directly to TLR4 and can be used as inducers of aseptic inflammation. In the past 20 years, targeting ligands rather than receptors has become an emerging therapeutic strategy for the treatment of diseases, so understanding the relationship between microglia, TLR4, TLR4 ligands, and corresponding diseases may have new implications for the treatment of diseases. In the article, we will discuss the TLR4 and the endogenous substances that can activate the TLR4 signaling pathway and present literature support for their role in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Bo Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P.R. China
- Office of China National Narcotics Control Commission, China Pharmaceutical University, Joint Laboratory on Key Technologies of Narcotics Control, Nanjing, 210009, P.R. China
| | - Bin Di
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P.R. China
- Office of China National Narcotics Control Commission, China Pharmaceutical University, Joint Laboratory on Key Technologies of Narcotics Control, Nanjing, 210009, P.R. China
| |
Collapse
|
6
|
Wang F, Ma Q, Dong X, Wang T, Ma C. circ-Gucy1a2 Protects Mice from Cerebral Ischemia-Reperfusion Injury by Attenuating Neuronal Apoptosis and Mitochondrial Membrane Potential Loss. J INVEST SURG 2023; 36:2152509. [PMID: 37277119 DOI: 10.1080/08941939.2022.2152509] [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: 07/15/2022] [Accepted: 11/22/2022] [Indexed: 06/07/2023]
Abstract
Cerebral ischemia-reperfusion (I/R) injury (CI/RI) is a severe problem in patients with cerebral ischemia. The current study explored the influences of circular (circ)-Gucy1a2 on neuronal apoptosis and mitochondrial membrane potential (MMP) in the brain tissue of CI/RI mice. Forty-eight mice were randomized into the sham group, transient middle cerebral artery occlusion (tMCAO) group, lentivirus negative control (LV-NC) group, and LV-Gucy1a2 group. Mice were first injected with lentivirus loaded with LV-Gucy1a2 or LV-NC via lateral ventricle, followed by the establishment of CI/RI models 2 weeks later. Twenty-four hours after CI/RI, the neurological impairment of mice was assessed using a 6-point scoring system. The cerebral infarct volume and brain histopathological changes were determined in CI/RI mice through histological staining. In vitro, pcDNA3.1-NC and pcDNA3.1-Gucy1a2 were transfected into mouse primary cortical neurons for 48 hours, followed by the establishment of oxygen-glucose deprivation/reoxygenation (OGD/R) models. The levels of circ-Gucy1a2 in mouse brain tissues and neurons were examined using RT-qPCR. Neuronal proliferation and apoptosis, MMP loss, and oxidative stress (OS)-related indexes in neurons were detected using CCK-8 assay, flow cytometry, JC-1 staining, and H2DFFDA staining. CI/RI mouse models and OGD/R cell models were successfully established. After CI/RI, neurons in mice were impaired and the cerebral infarction volume was increased. circ-Gucy1a2 was poorly expressed in CI/RI mouse brain tissues. Overexpression of circ-Gucy1a2 increased OGD/R-induced neuronal proliferation and mitigated apoptosis, MMP loss, and OS. Overall, circ-Gucy1a2 was down-regulated in brain tissues of CI/RI mice, and overexpression of circ-Gucy1a2 can protect mice from CI/RI.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang Province, China
| | - Qian Ma
- Department of Neurology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang Province, China
| | - Xinglu Dong
- Department of Neurology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang Province, China
| | - Tinghuan Wang
- Department of Neurology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang Province, China
| | - Chao Ma
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
7
|
Hassan FE, Aboulhoda BE, Ali IH, Elwi HM, Matter LM, Abdallah HA, Khalifa MM, Selmy A, Alghamdi MA, Morsy SA, Al Dreny BA. Evaluating the protective role of trimetazidine versus nano-trimetazidine in amelioration of bilateral renal ischemia/reperfusion induced neuro-degeneration: Implications of ERK1/2, JNK and Galectin-3 /NF-κB/TNF-α/HMGB-1 signaling. Tissue Cell 2023; 85:102241. [PMID: 37865040 DOI: 10.1016/j.tice.2023.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/24/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) is a primary culprit of acute kidney injury. Neurodegeneration can result from I/R, but the mechanisms are still challenging. We studied the implications of bilateral renal I/R on brain and potential involvement of the oxidative stress (OS) driven extracellular signal-regulated kinase1/2, c-Jun N-terminal kinase (ERK1/2, JNK) and Galectin-3 (Gal-3)/nuclear factor Kappa B (NF-қB)/tumor necrosis factor-alpha (TNF-α), high mobility group box-1 (HMGB-1), and caspase-3 paths upregulation. We tested the impact of Nano-trimetazidine (Nano-TMZ) on these pathways being a target of its neuroprotective effects. METHODS Study groups; Sham, I/R, TMZ+I/R, and Nano-TMZ+I/R. Kidney functions, cognition, hippocampal OS markers, Gal-3, NF-қB, p65 and HMGB-1 gene expression, TNF-α level, t-JNK/p-JNK and t-ERK/p-ERK proteins, caspase-3, glial fibrillary acidic protein (GFAP) and ionized calcium binding protein-1 (Iba-1) were assessed. RESULTS Nano-TMZ averted renal I/R-induced hippocampal impairment by virtue of its anti: oxidative, inflammatory, and apoptotic properties. CONCLUSION Nano-TMZ is more than anti-ischemic.
Collapse
Affiliation(s)
- Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt; General Medicine Practice Program, Department of Physiology, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Basma Emad Aboulhoda
- Anatomy and Embryology Department, Faculty of Medicine, Cairo University, Egypt.
| | - Isra H Ali
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sadat City, P.O. Box 32897, Sadat City, Egypt; Nanomedicine Laboratory, Faculty of Pharmacy, University of Sadat City, P.O. Box 32897, Sadat City, Egypt
| | - Heba M Elwi
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Lamiaa M Matter
- Medical pharmacology, Kasr Alainy, Faculty of Medicine, Cairo University, Egypt
| | - Hend Ahmed Abdallah
- Anatomy and Embryology Department, Faculty of Medicine, Cairo University, Egypt
| | - Mohamed Mansour Khalifa
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt; Department of Human Physiology, College of Medicine, King Saud University, Saudi Arabia
| | - Asmaa Selmy
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia; Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Suzan Awad Morsy
- Fakeeh College For Medical Sciences, Jeddah, Saudi Arabia; Faculty of Medicine, Alexandria University, Egypt
| | - Basant A Al Dreny
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt
| |
Collapse
|
8
|
Li X, Yang X, Lu H, Wang W, Cai L, Chen J, Wang Y. Calycosin attenuates the inflammatory damage of microglia induced by oxygen and glucose deprivation through the HMGB1/TLR4/NF-κB signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1415-1424. [PMID: 37528661 PMCID: PMC10520471 DOI: 10.3724/abbs.2023125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/07/2023] [Indexed: 08/03/2023] Open
Abstract
Stroke seriously threatens human life and health worldwide, but only very few effective stroke medicines are currently available. Our previous studies have indicated that the phytoestrogen calycosin exerts neuroprotective effects in cerebral ischemia and reperfusion injury rats. Therefore, the objective of this study is to further explore the protective effect of calycosin on inflammatory injury in microglia after oxygen-glucose deprivation/reoxygenation (OGD/R) and to clarify whether its protective effect is related to the HMGB1/TLR4/NF-κB signaling pathway. Here, the OGD/R model of rodent microglia is established in vitro to simulate cerebral ischemia-reperfusion injury. Through the CCK-8 test, ELISA, qRT-PCR, and western blot analysis, we find that the activity of microglia is decreased, the expressions of HMGB1 and TLR4 and the phosphorylation of NF-κB (p-NF-κB) are increased, and the releases of the inflammatory factors IL-6, IL-1β, and TNF-α are increased after OGD/R. Pretreatment with calycosin could ameliorate these states, increase cell viability, reduce HMGB1, TLR4 and p-NF-κB expression, and reduce inflammatory cytokine production. In addition, the effect of calycosin is similar to that of TAK-242 (an inhibitor of TLR4), and the effect of the combined treatment is better than that of the single treatment. The results indicate that calycosin protects microglia from OGD/R injury and reduces the inflammatory response. Calycosin might alleviate cerebral ischemia-reperfusion injury by inhibiting the HMGB1/TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Xiang Li
- Key Laboratory of Tumor Immunology and Microenvironmental RegulationGuilin Medical UniversityGuilin541199China
| | - Xin Yang
- Key Laboratory of Tumor Immunology and Microenvironmental RegulationGuilin Medical UniversityGuilin541199China
| | - Huiling Lu
- Department of Pathology and PhysiopathologyGuilin Medical UniversityGuilin541199China
| | - Wenbo Wang
- Department of NeurosurgeryNanxishan Hospital of Guangxi Zhuang Autonomous RegionGuilin541002China
| | - Le Cai
- Key Laboratory of Tumor Immunology and Microenvironmental RegulationGuilin Medical UniversityGuilin541199China
| | - Jian Chen
- Key Laboratory of Tumor Immunology and Microenvironmental RegulationGuilin Medical UniversityGuilin541199China
| | - Yong Wang
- Key Laboratory of Tumor Immunology and Microenvironmental RegulationGuilin Medical UniversityGuilin541199China
- Department of PhysiologyGuilin Medical UniversityGuilin541199China
| |
Collapse
|
9
|
Liao YQ, Min J, Wu ZX, Hu Z. Comparison of the effects of remimazolam and dexmedetomidine on early postoperative cognitive function in elderly patients with gastric cancer. Front Aging Neurosci 2023; 15:1123089. [PMID: 37342357 PMCID: PMC10277633 DOI: 10.3389/fnagi.2023.1123089] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
Purpose To compare the effects of remimazolam and dexmedetomidine on early postoperative cognitive dysfunction (POCD) in aged gastric cancer patients. Methods From June to December 2022, 104 elderly patients (aged 65-80 years) received laparoscopic radical resection of gastric cancer at the First Affiliated Hospital of Nanchang University. Using the random number table approach, the patients were separated into three groups: remimazolam (Group R), dexmedetomidine (Group D), and saline (Group C). The primary outcome was the incidence of POCD, and secondary outcomes included TNF-α and S-100β protein concentrations, hemodynamics, VAS scores, anesthesia recovery indicators, and the occurrence of adverse events within 48 h postoperatively. Results At 3 and 7 days after surgery, there were no statistically significant differences in the incidence of POCD, the MMSE and MoCA scores between groups R and D (p > 0.05). However, compared to the saline group, both groups had higher MMSE and MoCA scores and decreased incidences of POCD. These differences were statistically significant (p < 0.05). Between group R and group D, there were no statistically significant changes (p > 0.05) in the levels of TNF-α and S-100β protein at the three time points (at the end of the surgery, 1 day later, and 3 days later). Even though neither group's concentration of the two factors was as high as that of the saline group, the differences were statistically significant (p < 0.05). At all three time points-following induction (T2), 30 min into the operation (T3), and at the conclusion of the surgery (T4)-the heart rate and blood pressure in group R were greater than those in groups D and C. Statistics showed that the differences were significant (p < 0.05). The incidence of intraoperative hypotension was highest in group D and lowest in group R (p < 0.05). The dose of propofol and remifentanil, group C > group R > group D. Extubation and PACU residence times did not differ statistically significantly (p > 0.05) between the three groups. There was no significant difference in VAS scores between groups R and D after 24 h postoperatively (p > 0.05), although both had lower scores than group C, and the difference was statistically significant (p < 0.05). The VAS scores between the three groups at 72 h (T6) and 7 days (T7) were not statistically significant (p > 0.05). Adverse reactions such as respiratory depression, hypotension, bradycardia, agitation, drowsiness, and nausea and vomiting had the lowest incidence in group R and the highest incidence in group C (p < 0.05). Conclusion Remimazolam is similarly beneficial as dexmedetomidine in lowering the incidence of early POCD in aged patients after radical gastric cancer resection, probably due to reduced inflammatory response.
Collapse
Affiliation(s)
| | - Jia Min
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | | | | |
Collapse
|
10
|
Dai M, Zhu X, Zeng S, Liu Q, Hu R, Huang L, Wang Y, Deng J, Yu Q. Dexmedetomidine protects cells from Angiotensin II-induced smooth muscle cell phenotype switch and endothelial cell dysfunction. Cell Cycle 2023; 22:450-463. [PMID: 36196460 PMCID: PMC9879174 DOI: 10.1080/15384101.2022.2124489] [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: 06/07/2022] [Revised: 08/07/2022] [Accepted: 09/10/2022] [Indexed: 01/29/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disorder greatly threatening life of the elderly population. Dexmedetomidine (DEX), an α2-adrenergic receptor agonist, has been shown to suppress AAA development. Nevertheless, the signaling pathways that might be mediated by DEX in AAA has not been clarified. Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) were treated with Angiotensin II (Ang II) to mimic AAA in vitro. BrdU, wound healing, and Transwell assays were utilized for measuring VSMC proliferation and migration. Western blotting was used for evaluating protein levels of contractile VSMC markers, collagens and matrix metalloproteinases (MMPs) in VSMCs as well as apoptosis- and HMGB1/TLR4/NF-κB signaling-related markers in ECs. Cell adhesion molecule expression and monocyte-endothelial adhesion were assessed by immunofluorescence staining and adhesion assays. Flow cytometry was implemented for analyzing EC apoptosis. Hematoxylin-eosin staining and ELISA were used to detect the effect of DEX in vivo. In this study, DEX inhibited Ang II-evoked VSMC phenotype switch and extracellular matrix degradation. DEX suppressed the inflammatory response and apoptosis of ECs induced by Ang II. DEX inhibited HMGB1/TLR4/NF-κB signaling pathway in Ang II-treated ECs. DEX attenuated Ang II-induced AAA and inflammation in mice. Overall, DEX ameliorates Ang II-induced VSMC phenotype switch, and inactivates HMGB1/TLR4/NF-κB signaling pathway to alleviate Ang II-induced EC dysfunction.
Collapse
Affiliation(s)
- Min Dai
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaohong Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Simin Zeng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qiang Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ruilin Hu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lianghui Huang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun Deng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qi Yu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Cong D, Yu Y, Meng Y, Qi X. Dexmedetomidine (Dex) exerts protective effects on rat neuronal cells injured by cerebral ischemia/reperfusion via regulating the Sphk1/S1P signaling pathway. J Stroke Cerebrovasc Dis 2023; 32:106896. [PMID: 36395661 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106896] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
AIM To investigate the influence of dexmedetomidine (Dex) on cerebral ischemia/reperfusion (I/R)-injured rat neuronal cells by regulating the Sphk1/S1P pathway. METHODS The rats were divided into the following groups, with 18 rats in each group categorized on the basis of random number tables: sham (Sham), I/R (I/R), Dex, Sphk1 inhibitor (PF-543), and Dex together with the Sphk1 agonist phorbol-12-myristate-13-acetate (Dex+PMA). The neurological functions of the rats were assessed by the Longa scoring system at 24 h post reperfusion. The area of brain infarction was inspected using 2,3,5-triphenyltetrazolium chloride staining, and the water content of brain tissue was determined by the dry-wet weight method. The morphology of neurons in the CA1 region of the rat hippocampus was inspected using Nissl staining, while the apoptosis of neurons in this region was detected by terminal-deoxynucleotidyl transferase mediated nick end labeling staining. The Sphk1 and S1P protein levels were determined by immunofluorescence and western blotting, respectively. RESULTS Compared to the I/R group, rats in the Dex, PF-543, and Dex+PMA groups had a significantly lower neurological function score, as well as lower brain water content and a decreased infarction area. Moreover, the apoptotic index of the neurons and the Sphk1 and S1P levels in the hippocampal CA1 region were significantly lower in these groups (p<0.05). PMA, an agonist of Sphk1, was able to reverse the protective effects of Dex on I/R-induced neuronal cell injury. CONCLUSION Dex could protect cerebral I/R-induced neuronal cell injury by suppressing the Sphk1/S1P signaling pathway.
Collapse
Affiliation(s)
- Dawei Cong
- Department of Neurosurgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, China
| | - Yunlong Yu
- Department of Neurosurgery, Yantai Harbour Hospital, Yantai 264000, China.
| | - Yan Meng
- Yantai Comprehensive Health Service Center, Yantai 264000, China
| | - Xia Qi
- Yantai Comprehensive Health Service Center, Yantai 264000, China
| |
Collapse
|
12
|
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
|
13
|
Jiang Y, Liu Z, Liao Y, Sun S, Dai Y, Tang Y. Ischemic stroke: From pathological mechanisms to neuroprotective strategies. Front Neurol 2022; 13:1013083. [PMID: 36438975 PMCID: PMC9681807 DOI: 10.3389/fneur.2022.1013083] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
Ischemic stroke (IS) has complex pathological mechanisms, and is extremely difficult to treat. At present, the treatment of IS is mainly based on intravenous thrombolysis and mechanical thrombectomy, but they are limited by a strict time window. In addition, after intravenous thrombolysis or mechanical thrombectomy, damaged neurons often fail to make ideal improvements due to microcirculation disorders. Therefore, finding suitable pathways and targets from the pathological mechanism is crucial for the development of neuroprotective agents against IS. With the hope of making contributions to the development of IS treatments, this review will introduce (1) how related targets are found in pathological mechanisms such as inflammation, excitotoxicity, oxidative stress, and complement system activation; and (2) the current status and challenges in drug development.
Collapse
Affiliation(s)
- Yang Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenquan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuyong Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yajie Dai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yibo Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Ren W, Zhao F, Han Y, Liu Z, Zhai J, Jia K. Muscone improves hypoxia/reoxygenation (H/R)-induced neuronal injury by blocking HMGB1/TLR4/NF-κB pathway via modulating microRNA-142. PeerJ 2022; 10:e13523. [PMID: 35860039 PMCID: PMC9290999 DOI: 10.7717/peerj.13523] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 05/10/2022] [Indexed: 01/25/2023] Open
Abstract
Previous reports have indicated that natural muscone has neuroprotective effects against cerebral hypoxia injury; however, little is known in regards to its pharmacological mechanism. In this study, we tried to evaluate the neuroprotective effects and mechanisms of muscone against cerebral hypoxia injury using an in vitro model. The cerebral hypoxia injury cell model was produced by hypoxia/reoxygenation (H/R). The cell viability and apoptosis were measured using the cell counting Kit-8 and the Annexin V-FITC/PI Apoptosis Detection kit, respectively. To screen microRNAs regulated by muscone, we analyzed the gene expression datasets of GSE84216 retrieved from gene expression omnibus (GEO). Here, it was demonstrated that muscone treatment significantly alleviated the cell apoptosis, oxidative stress and inflammation in H/R-exposed neurons. Subsequently, through analyzing GSE84216 from the GEO database, miR-142-5p was markedly upregulated by treatment of muscone in this cell model of cerebral hypoxia injury. Further experiments revealed that downregulation of miR-142-5p eliminated the neuroprotective effects of muscone against H/R induced neuronal injury. Additionally, high mobility group box 1 (HMGB1), an important inflammatory factor, was identified as a direct target of miR-142-5p in neurons. Meanwhile, we further demonstrated that muscone could reduce the expression of HMGB1 by upregulating miR-142-5p expression, which subsequently resulted in the inactivation of TLR4/NF-κB pathway, finally leading to the improvement of cell injury in H/R-exposed neurons. Overall, we demonstrate for the first time that muscone treatment alleviates cerebral hypoxia injury in in vitro experiments through blocking activation of the TLR4/NF-κB signaling pathway by targeting HMGB1, suggesting that muscone may serve as a potential therapeutic drug for treating cerebral hypoxia injury.
Collapse
|
15
|
Abd El-Aal SA, AbdElrahman M, Reda AM, Afify H, Ragab GM, El-Gazar AA, Ibrahim SSA. Galangin Mitigates DOX-induced Cognitive Impairment in Rats: Implication of NOX-1/Nrf-2/HMGB1/TLR4 and TNF-α/MAPKs/RIPK/MLKL/BDNF. Neurotoxicology 2022; 92:77-90. [PMID: 35843304 DOI: 10.1016/j.neuro.2022.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022]
Abstract
The cognitive and behavioral decline observed in cancer survivors who underwent doxorubicin (DOX)-based treatment raises the need for therapeutic interventions to counteract these complications. Galangin (GAL) is a flavonoid-based phytochemical with pronounced protective effects in various neurological disorders. However, its impact on DOX-provoked neurotoxicity has not been clarified. Hence, the current investigation aimed to explore the ability of GAL to ameliorate DOX-provoked chemo-brain in rats. DOX (2mg/kg, once/week, i.p.) and GAL (50mg/kg, 5 times/week., via gavage) were administered for four successive weeks. The MWM and EPM tests were used to evaluate memory disruption and anxiety-like behavior, respectively. Meanwhile, targeted biochemical markers and molecular signals were examined by the aid of ELISA, Western blotting, and immune-histochemistry. In contrast to DOX-impaired rats, GAL effectively preserved hippocampal neurons, improved cognitive/behavioral functions, and enhanced the expression of the cell repair/growth index and BDNF. The antioxidant feature of GAL was confirmed by the amelioration of MDA, NO and NOX-1, along with restoring the Nrf-2/HO-1/GSH cue. In addition, GAL displayed marked anti-inflammatory properties as verified by the suppression of the HMGB1/TLR4 nexus and p-NF-κB p65 to inhibit TNF-α, IL-6, IL-1β, and iNOS. This inhibitory impact extended to entail astrocyte activation, as evidenced by the diminution of GFAP. These beneficial effects were associated with a notable reduction in p-p38MAPK, p-JNK1/2, and p-ERK1/2, as well as the necroptosis cascade p-RIPK1/p-RIPK3/p-MLKL. Together, these pleiotropic protective impacts advocate the concurrent use of GAL as an adjuvant agent for managing DOX-driven neurodegeneration and cognitive/behavioral deficits. DATA AVAILABILITY: The authors confirm that all relevant data are included in the supplementary materials.
Collapse
Affiliation(s)
- Sarah A Abd El-Aal
- Department of Pharmacy, Kut University College, Al Kut, Wasit 52001, Iraq.
| | - Mohamed AbdElrahman
- Department of Pharmacy, Al-Mustaqbal University College, Babylon 51001, Iraq; Department of Clinical Pharmacy, Badr University Hospital, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
| | - Ahmed M Reda
- Department of Pharmacy, Kut University College, Al Kut, Wasit 52001, Iraq; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11562, Egypt
| | - Hassan Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11562, Egypt
| | - Ghada M Ragab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12585, Egypt
| | - Amira A El-Gazar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | | |
Collapse
|
16
|
Qi Z, Yan Z, Wang Y, Ji N, Yang X, Zhang A, Li M, Xu F, Zhang J. Ginsenoside Rh2 Inhibits NLRP3 Inflammasome Activation and Improves Exosomes to Alleviate Hypoxia-Induced Myocardial Injury. Front Immunol 2022; 13:883946. [PMID: 35865525 PMCID: PMC9294352 DOI: 10.3389/fimmu.2022.883946] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022] Open
Abstract
The inflammatory microenvironment after acute myocardial infarction (MI) is a key limiting factor in the clinical application of stem cell transplantation and paracrine exosome therapy. Qishen Yiqi Pills contain a saponin ingredient called Ginsenoside Rh2 (Rh2) which exhibits a certain therapeutic effect on MI. However, the mechanism by which Rh2 alleviates the inflammatory microenvironment and improves the therapeutic efficiency of exosomes remains enigmatic. Here, we found that Rh2 attenuated the adverse effect of oxygen-glucose deprivation (OGD)-induced cellular injury, an in vitro pathological model of MI. Confocal microscopy revealed that DiI-labeled BMSCs-derived exosomes exhibited an increased homing ability of cardiomyocytes, which, in turn, inhibited the nuclear translocation of NF-κB p65 and NLRP3 inflammasome activation, thereby alleviating the inflammatory microenvironment and further facilitating the homing of exosomes to cardiomyocytes by forming a feed-forward enhancement loop. Additionally, we found that Rh2 could regulate the HMGB1/NF-κB signaling pathway to improve the OGD environment of cardiomyocytes, increasing the efficiency of the feed-forward loop. In conclusion, we found that Rh2 can improve the inflammatory microenvironment by enhancing the protection of exosomes against myocardial injury, providing new insights into the indirect modification of exosomes by Rh2 in MI treatment.
Collapse
Affiliation(s)
- Zhongwen Qi
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Institute of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhipeng Yan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueyao Wang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nan Ji
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoya Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ao Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meng Li
- Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Fengqin Xu
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Institute of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
17
|
Clinical Observation of Salvianolic Acid Combined with Panax Notoginseng Saponins Combined with Basic Nursing Intervention on Cerebral Ischemia-Reperfusion Injury in Rats. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8706730. [PMID: 35136538 PMCID: PMC8818411 DOI: 10.1155/2022/8706730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 12/30/2022]
Abstract
Objective To analyze the clinical observation of salvianolic acid combined with panax notoginseng saponins combined with basic nursing intervention on cerebral ischemia-reperfusion injury in rats and its effects on the expression of apoptosis-related proteins Bcl-2, Bax and caspase-3. Methods A total of 72 male Wistar rats were randomly divided into sham, ischemia/reperfusion (I/R), edaravone (Eda), salvianolic acid (SA), panax notoginseng saponins (PNS), and SA+PNS group. After administration for 5 days, the neurological function, cerebral infarction volume, brain index, and brain water content of rats were observed. ELISA kit assay was applied to measure the levels of IL-1β, IL-6, IL-8, TNF-α, MDA, SOD, GSH-Px, and T-AOC activity. Western blotting assay was used to detect the protein levels of p-53, NF-κB, Bcl-2, Bax, and Caspase-3 in the brain tissues surrounding infarction lesion. Results Compared with sham group, the mNSS score, brain index, brain water content, infarction volumes, MDA activity, and the levels of IL-6, IL-8, TNF-α and IL-1β as well as the protein levels of p-53, NF-κB, Bax and Caspase-3 were significantly increased, while the levels of Bcl-2 protein, SOD, GSH-Px and T-AOC were significantly decreased in I/R group. However, these levels were reversed in SA group, PNS group and SA + PNS group. Moreover, these changes in SA + PNS group were more obvious than those in SA and PNS group, and the differences were statistically significant. Conclusions SA, PNS and they combined with basic nursing have protective effects on cerebral I/R injury, and the combination with basic nursing has better effects than that used alone. The mechanism may be to regulate the expression of downstream apoptotic proteins by inhibiting the TLR4/NF-κB signaling pathway, thereby reducing neurological damage in rats.
Collapse
|
18
|
Liu Y, Fu H, Wang T. Neuroinflammation in perioperative neurocognitive disorders: From bench to the bedside. CNS Neurosci Ther 2022; 28:484-496. [PMID: 34990087 PMCID: PMC8928922 DOI: 10.1111/cns.13794] [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: 08/02/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
The perioperative neurocognitive disorders (PNDs) are one of the most common complications in elderly patients characterized by various forms of cognitive decline after anesthesia and surgery. Although the etiology for PNDs remained unclear, neuroinflammation has been characterized as one of the major causes, especially in the elderly patients. The activation of glial cells including microglia and astrocytes plays a significant role in the inflammatory responses in central nerve system (CNS). Although carefully designed, clinical studies on PNDs showed controversial results. Meanwhile, preclinical studies provided evidence from various levels, including behavior performance, protein levels, and gene expression. In this review, we summarize high‐quality studies and recent advances from both clinical and preclinical studies and provide a broad view from the onset of PNDs to its potential therapeutic targets. Future studies are needed to investigate the signaling pathways in PNDs for prevention and treatment, as well as the relationship of PNDs and future neurocognitive dysfunction.
Collapse
Affiliation(s)
- Yang Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Huiqun Fu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianlong Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Zhang Y, Lei Y, Yao X, Yi J, Feng G. Pinoresinol diglucoside alleviates ischemia/reperfusion-induced brain injury by modulating neuroinflammation and oxidative stress. Chem Biol Drug Des 2021; 98:986-996. [PMID: 34546621 PMCID: PMC9293043 DOI: 10.1111/cbdd.13956] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/11/2021] [Accepted: 09/18/2021] [Indexed: 01/03/2023]
Abstract
Brain ischemia/reperfusion (I/R) injury is a common pathological process after ischemic stroke. Pinoresinol diglucoside (PDG) has antioxidation and anti-inflammation activities. However, whether PDG ameliorates brain I/R injury is still unclear. In this study, middle cerebral artery occlusion (MCAO) model was established with male C57BL/6 mice, and the mice were treated with 5 and 10 mg/kg PDG via intravenous injection, respectively. The neurological deficit, infarct volume, and brain water content were then evaluated. HE staining and Nissl staining were used to analyze neuron injury. Besides, enzyme-linked immunosorbent assay and colorimetry assay were used to examine the level of inflammatory markers and oxidative stress markers, and Western blot was used to detect the expressions of p-p65, Nrf2, and HO-1. It was revealed that PDG could significantly alleviate the MCAO-induced neurological dysfunction of the mice and reduce the infarct volume, brain water content, and neuron injury. PDG treatment decreased the levels of TNF-α, IL-1β, IL-6, NO, ROS, and MDA, and significantly increased the activities of SOD, GSH, and GSH-Px in the brain tissue of the mice. Additionally, PDG could repress the activation of p65 and promote Nrf2 and HO-1 expressions. In conclusion, PDG exerts anti-inflammatory and antioxidation effects via regulating the NF-κB pathway and Nrf2/HO-1 pathway, thereby reducing the I/R-induced brain injury of mice.
Collapse
Affiliation(s)
- Yi Zhang
- Department of NeurologyChenzhou No. 1 People’s HospitalChenzhouChina
| | - Yuanbiao Lei
- Department of NeurologyChenzhou No. 1 People’s HospitalChenzhouChina
| | - Xiaoxi Yao
- Department of NeurologyChenzhou No. 1 People’s HospitalChenzhouChina
| | - Jiping Yi
- Department of NeurologyChenzhou No. 1 People’s HospitalChenzhouChina
| | - Ganghua Feng
- Department of NeurologyChenzhou No. 1 People’s HospitalChenzhouChina
| |
Collapse
|
20
|
Wang J, Wang Y. Circular RNA cerebellar degeneration-related protein 1 antisense RNA (Circ-CDR1as) downregulation induced by dexmedetomidine treatment protects hippocampal neurons against hypoxia/reoxygenation injury through the microRNA-28-3p (miR-28-3p)/tumor necrosis factor receptor-associated factor-3 (TRAF3) axis. Bioengineered 2021; 12:10512-10524. [PMID: 34787053 PMCID: PMC8810102 DOI: 10.1080/21655979.2021.1999369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cerebral ischemia/reperfusion (CI/R) injury results in serious brain tissue damage, thereby leading to long-term disability and mortality. It has been reported that dexmedetomidine (DEX) exerted neuroprotective effects in CI/R injury. Herein, we intended to investigate whether and how circular RNA (circRNA) cerebellar degeneration-related protein 1 antisense RNA (circ-CDR1as) was involved in the DEX-mediated protection on hippocampal neurons. In our work, the mouse hippocampal neuronal cells (HT-22) were used to construct a hypoxia/reperfusion (H/R) model for CI/R injury. Cell proliferation and apoptosis were evaluated by CCK-8 and flow cytometry. Gene expressions were detected by RT-qPCR. Levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) were measured by ELISA. The association between miR-28-3p and circ-CDR1as or TRAF3 was verified by dual-luciferase assay. The results indicated that DEX alleviated HT-22 cell dysfunction induced by H/R treatment. In addition, circ-CDR1as was downregulated after DEX treatment and reversed the effects of DEX on the proliferation, apoptosis, and inflammatory responses of H/R-treated HT-22 cells. Circ-CDR1as positively regulated TRAF3 expression via interaction with miR-28-3p in HT-22 cells. Circ-CDR1as aggravated H/R-treated HT-22 cell dysfunction through targeting miR-28-3p. Furthermore, TRAF3 inhibition partly abolished the effect of circ-CDR1as overexpression on cellular activities of H/R-treated HT-22 cells. To sum up, our findings, for the first time, demonstrated that DEX exerted neuroprotective effects on hippocampal neurons against H/R treatment via the circ-CDR1as/miR-28-3p/TRAF3 regulatory network, providing novel therapeutic targets for DEX administration in CI/R treatment.
Collapse
Affiliation(s)
- Junhua Wang
- Department of Anesthesia, Liyang People's Hospital, Changzhou, P.R. China
| | - Ying Wang
- Department of Anesthesia, Liyang People's Hospital, Changzhou, P.R. China
| |
Collapse
|
21
|
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
|
22
|
Kamdee K, Panadsako N, Mueangson O, Nuinoon M, Janwan P, Poonsawat W, Pongpanitanont P, Kitkumthorn N, Thongsroy J, Chunglok W. Promoter polymorphism of TNF-α (rs1800629) is associated with ischemic stroke susceptibility in a southern Thai population. Biomed Rep 2021; 15:78. [PMID: 34405050 PMCID: PMC8329996 DOI: 10.3892/br.2021.1454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/29/2021] [Indexed: 01/19/2023] Open
Abstract
Stroke represents the leading cause of disability and mortality amongst the elderly worldwide. Multiple risk factors, including both genetic and non-genetic components, as well as their interactions, are proposed as etiological factors involved in the development of ischemic stroke (IS). Promoter polymorphisms of the IL-6-174G/C (rs1800795) and TNF-α-308G/A (rs1800629) genes have been considered as predictive risk factors of IS; however, these have not yet been evaluated in a Thai population. The aims of this study were to investigate the association of IL-6-174G/C and TNF-α-308G/A polymorphisms with IS. Genomic DNA from 200 patients with IS and 200 controls were genotyped for IL-6-174G/C and TNF-α-308G/A polymorphisms using TaqMan™ SNP genotyping and quantitative PCR-high resolution melting analysis, respectively. It was found that the TNF-α-308 A allele was significantly associated with an increased risk of IS development compared with the G allele [odds ratio (OR)=2.044; 95% CI=1.154-3.620; P=0.014]. Moreover, the IS risk was significantly higher in the presence of TNF-α-308 GA or AA genotypes compared with that in the presence of GG genotypes with a dominant inheritance (OR=1.971; 95% CI=1.080-3.599; P=0.027). However, there was no association between IL-6-174G/C and the risk of IS development. The interaction study demonstrated that IL-6-174 GG and TNF-α-308 GG genotypes enhanced IS susceptibility when combined with hypertension, hyperlipidemia and alcohol consumption. Hypertensive and hyperlipidemic subjects with the TNF-α-308 GA and AA genotypes were more likely to develop IS compared with those who did not have these two conditions and had the GG genotype. In a matched study design (1:1), the IL-6-174 GC genotype was associated with higher IL-6 levels in the control group. Collectively, the present results highlight the utility of the TNF-α-308G/A polymorphism as a predictive genetic risk factor for development of IS.
Collapse
Affiliation(s)
- Kornyok Kamdee
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Nitirat Panadsako
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Onchuma Mueangson
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Manit Nuinoon
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Penchom Janwan
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wasinee Poonsawat
- Research Institute for Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | | | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Jirapan Thongsroy
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Warangkana Chunglok
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand.,Food Technology and Innovation Research Center of Excellence, Institute of Research and Innovation, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
23
|
Liu W, Shao C, Zang C, Sun J, Xu M, Wang Y. Protective effects of dexmedetomidine on cerebral ischemia/reperfusion injury via the microRNA-214/ROCK1/NF-κB axis. BMC Anesthesiol 2021; 21:203. [PMID: 34399695 PMCID: PMC8365892 DOI: 10.1186/s12871-021-01423-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 07/27/2021] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Cerebral ischemia/reperfusion injury (CIRI) is a complication of surgical procedure associated with high mortality. The protective effect of dexmedetomidine (DEX) on CIRI has been explored in previous works, yet the underlying molecular mechanism remains unclear. Our study explored the protective effect of DEX and its regulatory mechanism on CIRI. METHODS A CIRI rat model was established using middle cerebral artery occlusion (MCAO). Neurological deficit scores for rats received MCAO modeling or DEX treatment were measured. Cerebral infarction area of rats was detected by TTC staining, while damage of neurons in hippocampal regions of rats was determined by hematoxylin-eosin (HE) staining. Apoptosis rate of neurons in hippocampal regions was examined by TUNEL staining. The dual-luciferase assay was performed to detect the binding of microRNA-214 (miR-214) to Rho-associated kinase 1 (ROCK1). RESULTS DEX treatment significantly reduced infarction area of MCAO rats and elevated miR-214 expression. Injection of miR-214 inhibitor attenuated the effect of DEX in MCAO rats by increasing the area of cerebral infarction in rats and apoptosis rate of hippocampal neurons. ROCK1 was targeted and negatively regulated by miR-214. The overexpression of ROCK1 led to activation of NF-κB to aggravate CIRI. CONCLUSION Therapeutic effects of DEX on CIRI was elicited by overexpressing miR-214 and impairing ROCK1 expression and NF-κB activation. Our finding might provide novel insights into the molecular mechanism of DEX in rats with CIRI.
Collapse
Affiliation(s)
- Wenyi Liu
- Department of Anesthesiology|, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Laoshan District, Qingdao, 266003, Shandong, PR China
| | - Cuihua Shao
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, PR China
| | - Chuanshan Zang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, PR China
| | - Jian Sun
- Department of Anesthesiology|, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Laoshan District, Qingdao, 266003, Shandong, PR China
| | - Min Xu
- Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, PR China
| | - Yuna Wang
- Department of Anesthesiology|, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Laoshan District, Qingdao, 266003, Shandong, PR China.
| |
Collapse
|
24
|
Yang JJ, Zhao YH, Yin KW, Zhang XQ, Liu J. Dexmedetomidine inhibits inflammatory response and oxidative stress through regulating miR-205-5p by targeting HMGB1 in cerebral ischemic/reperfusion. Immunopharmacol Immunotoxicol 2021; 43:478-486. [PMID: 34196265 DOI: 10.1080/08923973.2021.1942901] [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] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate effects of dexmedetomidine (DEX) on miR-205-5p/HMGB1 axis in cerebral ischemic/reperfusion (I/R) injury. METHODS Both in vivo I/R rat model and in vitro hypoxia/reoxygenation (H/R) cell model using rat hippocampal neurons cells were established. miR-205-5p was overexpressed or inhibited by transfection of miR-205-5p mimics or inhibitor. HMGB1 was overexpressed by transfection overexpression plasmids (OE-HMGB1). TTC staining was used for measurement of infraction volume. Oxidative stress was evaluated by measurement of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) and inflammation was evaluated by measurement of IL-1β, IL-6 and TNF-α. Dual luciferase reporter assay was performed to confirm binding between miR-205-5p and HMGB1. The expression levels of miR-205-5p, and HMGB1 were measured using RT-qPCR. Western blotting was used to test the protein expression levels of HMGB1, nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione peroxidase (GPx), glutathione reductase (GR), heme oxygenase 1 (HO-1) and catalase (CAT). RESULTS Treatment of DEX significantly reduced brain infraction volume, decreased Longa's neurological function score and inhibited oxidative stress and inflammation in brain tissues of I/R rats, which were all reversed by inhibition of miR-205-5p. Both treatment of DEX or overexpression of miR-205-5p restricted oxidative stress and inflammation in H/R rat hippocampal neurons cells. The inhibition of miR-205-5p reversed the effects of DEX, while the overexpression of HMGB1 reversed the effects of miR-205-5p overexpression in H/R rat hippocampal neurons cells. Dual luciferase reporter assay showed miR-205-5p directly targeted HMGB1. CONCLUSION DEX improved I/R injury by suppressing brain oxidative stress and inflammation DEX improved I/R injury by suppressing brain oxidative stress and inflammation through activating miR-205-5p/HMGB1 axis through activating miR-205-5p/HMGB1 axis.
Collapse
Affiliation(s)
- Jun-Jun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Yan-Hong Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Ke-Wen Yin
- Department of Anesthesiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Xiao-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Jianhui Liu
- Department of Anesthesiology, Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
25
|
Ashayeri Ahmadabad R, Mirzaasgari Z, Gorji A, Khaleghi Ghadiri M. Toll-Like Receptor Signaling Pathways: Novel Therapeutic Targets for Cerebrovascular Disorders. Int J Mol Sci 2021; 22:ijms22116153. [PMID: 34200356 PMCID: PMC8201279 DOI: 10.3390/ijms22116153] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptors (TLRs), a class of pattern recognition proteins, play an integral role in the modulation of systemic inflammatory responses. Cerebrovascular diseases (CVDs) are a group of pathological conditions that temporarily or permanently affect the brain tissue mostly via the decrease of oxygen and glucose supply. TLRs have a critical role in the activation of inflammatory cascades following hypoxic-ischemic events and subsequently contribute to neuroprotective or detrimental effects of CVD-induced neuroinflammation. The TLR signaling pathway and downstream cascades trigger immune responses via the production and release of various inflammatory mediators. The present review describes the modulatory role of the TLR signaling pathway in the inflammatory responses developed following various CVDs and discusses the potential benefits of the modulation of different TLRs in the improvement of functional outcomes after brain ischemia.
Collapse
Affiliation(s)
- Rezan Ashayeri Ahmadabad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
| | - Zahra Mirzaasgari
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
- Department of Neurology, Iran University of Medical Sciences, Tehran 1593747811, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
- Epilepsy Research Center, Westfälische Wilhelms-Universität, 48149 Münster, Germany
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität, 48149 Münster, Germany;
- Department of Neurology, Westfälische Wilhelms-Universität Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-8355564; Fax: +49-251-8347479
| | | |
Collapse
|
26
|
Huang Y, Wang Y, Duan Z, Liang J, Xu Y, Zhang S, Tang T. Restored microRNA-326-5p Inhibits Neuronal Apoptosis and Attenuates Mitochondrial Damage via Suppressing STAT3 in Cerebral Ischemia/Reperfusion Injury. NANOSCALE RESEARCH LETTERS 2021; 16:63. [PMID: 33877455 PMCID: PMC8058131 DOI: 10.1186/s11671-021-03520-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/30/2021] [Indexed: 05/10/2023]
Abstract
Studies have greatly explored the role of microRNAs (miRNAs) in cerebral ischemia/reperfusion injury (CI/RI). But the specific mechanism of miR-326-5p in CI/RI is still elusive. Hence, this study was to unmask the mechanism of miR-326-5p/signal transducer and activator of transcription-3 (STAT3) axis in CI/RI. Two models (oxygen and glucose deprivation [OGD] in primary rat cortical neurons and middle cerebral artery occlusion [MCAO] in Sprague-Dawley rats) were established to mimic CI/RI in vitro and in vivo, respectively. Loss- and gain-of function assays were performed with OGD-treated neurons and with MCAO rats. Afterward, viability, apoptosis, oxidative stress and mitochondrial membrane potential in OGD-treated neurons were tested, as well as pathological changes, apoptosis and mitochondrial membrane potential in brain tissues of MCAO rats. Mitofusin-2 (Mfn2), miR-326-5p and STAT3 expression in OGD-treated neurons and in brain tissues of MCAO rats were detected. Mfn2 and miR-326-5p were reduced, and STAT3 was elevated in OGD-treated neurons and brain tissues of MCAO rats. miR-326-5p targeted and negatively regulated STAT3 expression. Restoring miR-326-5p or reducing STAT3 reinforced viability, inhibited apoptosis and oxidative stress, increased mitochondrial membrane potential and increased Mfn2 expression in OGD-treated neurons. Up-regulating miR-326-5p or down-regulating STAT3 relieved pathological changes, inhibited apoptosis and elevated mitochondrial membrane potential and Mfn2 expression in brain tissues of rats with MCAO. This study elucidates that up-regulated miR-326-5p or down-regulated STAT3 protects against CI/RI by elevating Mfn2 expression.
Collapse
Affiliation(s)
- Yumin Huang
- Department of Respiratory and Critical Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yingge Wang
- Department of Neurology, Affiliated Hospital of Yangzhou University; Yangzhou University, 45 Taizhou Road, Yangzhou, 225001, Jiangsu, People's Republic of China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Zuowei Duan
- Department of Neurology, Affiliated Hospital of Yangzhou University; Yangzhou University, 45 Taizhou Road, Yangzhou, 225001, Jiangsu, People's Republic of China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Department of Jiangsu Key Laboratory of Experimental, Translational Non‑coding RNA Research, Yangzhou, Jiangsu, 225001, People's Republic of China
| | - Yijun Xu
- Medical College, Yangzhou University; Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Shuai Zhang
- Department of Neurology, Affiliated Hospital of Yangzhou University; Yangzhou University, 45 Taizhou Road, Yangzhou, 225001, Jiangsu, People's Republic of China
| | - Tieyu Tang
- Department of Neurology, Affiliated Hospital of Yangzhou University; Yangzhou University, 45 Taizhou Road, Yangzhou, 225001, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
MiR-22-3p suppresses sepsis-induced acute kidney injury by targeting PTEN. Biosci Rep 2021; 40:224157. [PMID: 32412059 PMCID: PMC7268257 DOI: 10.1042/bsr20200527] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Septic acute kidney injury is considered as a severe and frequent complication that occurs during sepsis. The present study was performed to understand the role of miR-22-3p and its underlying mechanism in sepsis-induced acute kidney injury. Methods: Rats were injected with adenovirus carrying miR-22-3p or miR-NC in the caudal vein before cecal ligation. Meanwhile, HK-2 cells were transfected with the above adenovirus following LPS stimulation. We measured the markers of renal injury (blood urea nitrogen (BUN), serum creatinine (SCR)). Histological changes in kidney tissues were examined by hematoxylin and eosin (H&E), Masson staining, periodic acid Schiff staining and TUNEL staining. The levels of IL-1β, IL-6, TNF-α and NO were determined by ELISA assay. Using TargetScan prediction and luciferase reporter assay, we predicted and validated the association between PTEN and miR-22-3p. Results: Our data showed that miR-22-3p was significantly down-regulated in a rat model of sepsis-induced acute kidney injury, in vivo and LPS-induced sepsis model in HK-2 cells, in vitro. Overexpression of miR-22-3p remarkably suppressed the inflammatory response and apoptosis via down-regulating HMGB1, p-p65, TLR4 and pro-inflammatory factors (IL-1β, IL-6, TNF-α and NO), both in vivo and in vitro. Moreover, PTEN was identified as a target of miR-22-3p. Furthermore, PTEN knockdown augmented, while overexpression reversed the suppressive role of miR-22-3p in LPS-induced inflammatory response. Conclusions: Our results showed that miR-22-3p induced protective role in sepsis-induced acute kidney injury may rely on the repression of PTEN.
Collapse
|
28
|
Youssef ME, Abdelrazek HM, Moustafa YM. Cardioprotective role of GTS-21 by attenuating the TLR4/NF-κB pathway in streptozotocin-induced diabetic cardiomyopathy in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:11-31. [PMID: 32776158 DOI: 10.1007/s00210-020-01957-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
The cholinergic anti-inflammatory pathway (CAP) was investigated in a variety of inflammatory conditions and constitutes a valuable line in their treatment. In the current study, we investigated the anti-inflammatory effect of GTS-21 (GTS) as a partial selective α7 nicotinic acetylcholine receptor (α7-nAchR) agonist in diabetic cardiomyopathy model in rats. This mechanism was elaborated to study whether it could alleviate the electrocardiographic, histopathological, and molecular levels of Toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB) pathway proteins. Diabetes was induced by the injection of streptozotocin (STZ) (50 mg/kg). Diabetic rats were treated with GTS (1 or 2 mg/kg/day), methyllycaconitine (MLA), a selective α7-nAchR antagonist (2 mg/kg/day) plus GTS (2 mg/kg/day), or the vehicle. All treatments were given by the intraperitoneal route. Ventricular rate and different electrocardiograph (ECG) anomalies were detected. Plasma levels of cardiac troponin T (cTnT) and creatine kinase MB (CK-MB) were measured by ELISA. Additionally, we elucidated the levels of several proteins involved in the TLR4/NF-κB pathway. Cardiac levels of TLR4 and phosphorylated protein kinase B (p-Akt) were detected by ELISA. The cardiac expression of myeloid differentiation primary response 88 (Myd88), tumor necrosis factor receptor-associated factor 6 (TRAF6), NF-κB, interleukin 1β (IL-1β), and active caspase-1 were evaluated by immunohistochemical staining. Finally, the cardiac levels of interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) were determined by ELISA. Diabetic rats showed (i) ECG signs of cardiomyopathy such as significant ST segment elevations, prolonged QRS, QT intervals, and ventricular tachycardia; (ii) increased plasma levels of cTnT and CK-MB; (iii) increased expression of cardiac TLR4; (iv) elevated immunohistochemical expression of cardiac, Myd88, TRAF6, and NF-κB; (v) diminution in the cardiac expression of p-Akt; and (vi) adaptive increases in cardiac expression of TNF-α and IL-6. These effects were ameliorated in diabetic rats treated with both doses of GTS. Pretreatment with MLA did not completely reverse the ameliorative effect of GTS on cTnT, TRAF6, TNF-α, and IL-6, thereby reinforcing the presence of possible α7-nAchR-independent mechanisms. The activation of α7-nAchR with GTS offers a promising prophylactic strategy for diabetic cardiomyopathy by attenuating the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Mahmoud E Youssef
- Department of pharmacology and biochemistry, Faculty of pharmacy, Delta University for Science and Technology, Mansoura, Egypt.
| | - Heba M Abdelrazek
- Department of Physiology, Faculty of veterinary medicine, Suez Canal University, Ismailia, Egypt
| | - Yasser M Moustafa
- Department of Pharmacology and Toxicology, Dean of the Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
29
|
Ocak U, Eser Ocak P, Huang L, Xu W, Zuo Y, Li P, Gamdzyk M, Zuo G, Mo J, Zhang G, Zhang JH. Inhibition of mast cell tryptase attenuates neuroinflammation via PAR-2/p38/NFκB pathway following asphyxial cardiac arrest in rats. J Neuroinflammation 2020; 17:144. [PMID: 32366312 PMCID: PMC7199326 DOI: 10.1186/s12974-020-01808-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cardiac arrest survivors suffer from neurological dysfunction including cognitive impairment. Cerebral mast cells, the key regulators of neuroinflammation contribute to neuroinflammation-associated cognitive dysfunction. Mast cell tryptase was demonstrated to have a proinflammatory effect on microglia via the activation of microglial protease-activated receptor-2 (PAR-2). This study investigated the potential anti-neuroinflammatory effect of mast cell tryptase inhibition and the underlying mechanism of PAR-2/p-p38/NFκB signaling following asphyxia-induced cardiac arrest in rats. Methods Adult male Sprague-Dawley rats resuscitated from 10 min of asphyxia-induced cardiac arrest were randomized to four separate experiments including time-course, short-term outcomes, long-term outcomes and mechanism studies. The effect of mast cell tryptase inhibition on asphyxial cardiac arrest outcomes was examined after intranasal administration of selective mast cell tryptase inhibitor (APC366; 50 μg/rat or 150 μg/rat). AC55541 (selective PAR-2 activator; 30 μg/rat) and SB203580 (selective p38 inhibitor; 300 μg/rat) were used for intervention. Short-term neurocognitive functions were evaluated using the neurological deficit score, number of seizures, adhesive tape removal test, and T-maze test, while long-term cognitive functions were evaluated using the Morris water maze test. Hippocampal neuronal degeneration was evaluated by Fluoro-Jade C staining. Results Mast cell tryptase and PAR-2 were dramatically increased in the brain following asphyxia-induced cardiac arrest. The inhibition of mast cell tryptase by APC366 improved both short- and long-term neurological outcomes in resuscitated rats. Such behavioral benefits were associated with reduced expressions of PAR-2, p-p38, NFκB, TNF-α, and IL-6 in the brain as well as less hippocampal neuronal degeneration. The anti-neuroinflammatory effect of APC366 was abolished by AC55541, which when used alone, indeed further exacerbated neuroinflammation, hippocampal neuronal degeneration, and neurologic deficits following cardiac arrest. The deleterious effects aggregated by AC55541 were minimized by p38 inhibitor. Conclusions The inhibition of mast cell tryptase attenuated neuroinflammation, led to less hippocampal neuronal death and improved neurological deficits following cardiac arrest. This effect was at least partly mediated via inhibiting the PAR-2/p-p38/NFκB signaling pathway. Thus, mast cell tryptase might be a novel therapeutic target in the management of neurological impairment following cardiac arrest.
Collapse
Affiliation(s)
- Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Emergency Medicine, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, 16310, Bursa, Turkey.,Department of Emergency Medicine, Bursa City Hospital, 16110, Bursa, Turkey
| | - Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Uludag University School of Medicine, 16069, Bursa, Turkey
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Weilin Xu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China
| | - Yuchun Zuo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Peng Li
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Suzhou, Taicang, 215400, Jiangsu, China
| | - Jun Mo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Guangyu Zhang
- Mass Spectrometry Core Facility, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
30
|
Yang H, Wang H, Andersson U. Targeting Inflammation Driven by HMGB1. Front Immunol 2020; 11:484. [PMID: 32265930 PMCID: PMC7099994 DOI: 10.3389/fimmu.2020.00484] [Citation(s) in RCA: 394] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/02/2020] [Indexed: 12/22/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a highly conserved, nuclear protein present in all cell types. It is a multi-facet protein exerting functions both inside and outside of cells. Extracellular HMGB1 has been extensively studied for its prototypical alarmin functions activating innate immunity, after being actively released from cells or passively released upon cell death. TLR4 and RAGE operate as the main HMGB1 receptors. Disulfide HMGB1 activates the TLR4 complex by binding to MD-2. The binding site is separate from that of LPS and it is now feasible to specifically interrupt HMGB1/TLR4 activation without compromising protective LPS/TLR4-dependent functions. Another important therapeutic strategy is established on the administration of HMGB1 antagonists precluding RAGE-mediated endocytosis of HMGB1 and HMGB1-bound molecules capable of activating intracellular cognate receptors. Here we summarize the role of HMGB1 in inflammation, with a focus on recent findings on its mission as a damage-associated molecular pattern molecule and as a therapeutic target in inflammatory diseases. Recently generated HMGB1-specific inhibitors for treatment of inflammatory conditions are discussed.
Collapse
Affiliation(s)
- Huan Yang
- Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Haichao Wang
- Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|