1
|
Park JH, Oh JE, Kim N, Kwak YL. Dexmedetomidine alleviates CoCl2-induced hypoxic cellular damage in INS-1 cells by regulating autophagy. Korean J Anesthesiol 2024; 77:623-634. [PMID: 39355897 PMCID: PMC11637589 DOI: 10.4097/kja.24457] [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: 07/04/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion (I/R) injury is inevitable during the perioperative period. The pancreas is susceptible to I/R injury. Autophagy, a self-digestion process, is upregulated during I/R injury and strongly induced by hypoxia. This study aims to determine whether dexmedetomidine can decrease pancreatic β-cell damage by regulating autophagy under hypoxia. METHODS INS-1 rat insulinoma cells were cultured in dexmedetomidine before being exposed to cobalt chloride (CoCl2)-induced hypoxia. Cell viability and the expression of autophagy-related proteins (light chain 3B [LC3B]-II, p62, and ATGs) were assessed. The expression of apoptosis-related proteins (BCL-2 and P-BAD) were also evaluated. CoCl2-treated INS-1 cells were pretreated with the autophagosome formation inhibitor, 3-methyladenine (3-MA), to compare its effects with those of dexmedetomidine. Bafilomycin-A1 (Baf-A1) that inhibits autophagosome degradation was used to confirm the changes in autophagosome formation induced by dexmedetomidine. RESULTS Dexmedetomidine attenuated the increased expression of autophagic proteins (LC3B-II, p62, and ATGs) and reversed the CoCl2-induced reduction in the proliferation of INS-1 cells after hypoxia. Dexmedetomidine also alleviated the decreased expression of the anti-apoptotic protein (BCL-2) and the increased expression of apoptotic protein (BAX). Dexmedetomidine reduces the activation of autophagy through inhibiting autophagosome formation, as confirmed by a decrease in LC3B-II/I ratio, a marker of autophagosome formation, in LC3B turnover assay combined with Baf-A1. CONCLUSIONS Dexmedetomidine alleviates the degree of cellular damage in INS-1 cells against CoCl2-induced hypoxia by regulating autophagosome formation. These results provide a basis for further studies to confirm these effects in clinical practice.
Collapse
Affiliation(s)
- Jin Ha Park
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ju Eun Oh
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Namo Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Lan Kwak
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
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
|
Hou Y, Ye W, Tang Z, Li F. Anesthetics in pathological cerebrovascular conditions. J Cereb Blood Flow Metab 2024:271678X241295857. [PMID: 39450477 PMCID: PMC11563546 DOI: 10.1177/0271678x241295857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
4
|
Xu Y, Teng X, Wei M, Liu Y. TREK-1 channel as a therapeutic target for dexmedetomidine-mediated neuroprotection in cerebral ischemia. Neurogenetics 2024; 25:367-375. [PMID: 38976083 DOI: 10.1007/s10048-024-00772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Our objective is to explore the protective effect of Dexmedetomidine on brain apoptosis and its mechanism through TREK-1 pathway. Forty male Sprague-Dawley rats were allocated into four groups: Sham, Cerebral Ischemia/Reperfusion Injury (CIRI), 50 µg/kg Dex, and 100 µg/kg Dex. A rat model of middle cerebral artery occlusion (MCAO) was employed to simulate cerebral embolism. Primary cortical neurons were exposed to Dex for 48 h, with some receiving additional treatment with 100 µM yohimbine hydrochloride (YOH) or TREK-1 small interfering RNA (siRNA). Neuronal damage was assessed using hematoxylin and eosin (HE) staining. Cell viability and apoptosis were measured by Cell Counting Kit-8 (CCK8) and flow cytometry, respectively. Protein and gene expression levels of Bcl-2, Bax, and TREK-1 were determined by Western blot and real-time polymerase chain reaction (PCR). Histopathological changes revealed that Dex treatment at both 50 µg/kg and 100 µg/kg significantly mitigated neuronal damage compared to the CIRI group. YOH treatment and Trek1 siRNA significantly reduced cell viability (p < 0.05). The mRNA expression and protein levels of TREK-1 and Bax were remarkably increased, while mRNA expression and protein levels of Bcl-2 was seriously decreased after CIRI modeling. In contrast, Dex treatment at both concentrations led to decreased TREK-1 and Bax expression and increased Bcl-2 expression in primary cortical neurons. Addition of 100 µM YOH and Trek1 siRNA reversed the effects of Dex on apoptosis-related genes (p < 0.05). Dex exerts neuroprotective effects through the TREK-1 pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Yang Xu
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - XiaoDan Teng
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - Ming Wei
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - Yang Liu
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China.
| |
Collapse
|
5
|
Zhang ZZ, Nasir A, Li D, Khan S, Bai Q, Yuan F. Effect of dexmedetomidine on ncRNA and mRNA profiles of cerebral ischemia-reperfusion injury in transient middle cerebral artery occlusion rats model. Front Pharmacol 2024; 15:1437445. [PMID: 39170713 PMCID: PMC11335533 DOI: 10.3389/fphar.2024.1437445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Ischemic stroke poses a significant global health burden, with rapid revascularization treatments being crucial but often insufficient to mitigate ischemia-reperfusion (I/R) injury. Dexmedetomidine (DEX) has shown promise in reducing cerebral I/R injury, but its potential molecular mechanism, particularly its interaction with non-coding RNAs (ncRNAs), remains unclear. This study investigates DEX's therapeutic effect and potential molecular mechanisms in reducing cerebral I/R injury. A transient middle cerebral artery obstruction (tMACO) model was established to simulate cerebral I/R injury in adult rats. DEX was administered pre-ischemia and post-reperfusion. RNA sequencing and bioinformatic analyses were performed on the ischemic cerebral cortex to identify differentially expressed non-coding RNAs (ncRNAs) and mRNAs. The sequencing results showed 6,494 differentially expressed (DE) mRNA and 2698 DE circRNA between the sham and tMCAO (I/R) groups. Additionally, 1809 DE lncRNA, 763 DE mRNA, and 2795 DE circRNA were identified between the I/R group and tMCAO + DEX (I/R + DEX) groups. Gene ontology (GO) analysis indicated significant enrichment in multicellular biogenesis, plasma membrane components, and protein binding. KEGG analysis further highlighted the potential mechanism of DEX action in reducing cerebral I/R injury, with hub genes involved in inflammatory pathways. This study demonstrates DEX's efficacy in reducing cerebral I/R injury and offers insights into its brain-protective effects, especially in ischemic stroke. Further research is warranted to fully understand DEX's neuroprotective mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Zhen Zhen Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Abdul Nasir
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dong Li
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Suliman Khan
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Bai
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Yuan
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Selvanathan T, Miller SP. Effects of pain, sedation and analgesia on neonatal brain injury and brain development. Semin Perinatol 2024; 48:151928. [PMID: 38937163 DOI: 10.1016/j.semperi.2024.151928] [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] [Indexed: 06/29/2024]
Abstract
Critically ill newborns experience numerous painful procedures as part of lifesaving care in the Neonatal Intensive Care Unit. However, painful exposures in the neonatal period have been associated with alterations in brain maturation and poorer neurodevelopmental outcomes in childhood. The most frequently used medications for pain and sedation in the NICU are opioids, benzodiazepines and sucrose; these have also been associated with abnormalities in brain maturation and neurodevelopment making it challenging to know what the best approach is to treat neonatal pain. This article provides clinicians with an overview of how neonatal exposure to pain as well as analgesic and sedative medications impact brain maturation and neurodevelopmental outcomes in critically ill infants. We also highlight areas in need of future research to develop standardized neonatal pain monitoring and management strategies.
Collapse
Affiliation(s)
- Thiviya Selvanathan
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, BC, Canada; Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Steven P Miller
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, BC, Canada; Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada; Neurosciences and Mental Health, SickKids Research Institute, Toronto, ON, Canada.
| |
Collapse
|
7
|
Liu S, Jia X, Liu B, Liu Y, Yin H. Suppression of cerebral ischemia injury induced blood brain barrier breakdown by dexmedetomidine via promoting CCN1. Aging (Albany NY) 2024; 16:3750-3762. [PMID: 38364236 PMCID: PMC10929797 DOI: 10.18632/aging.205557] [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: 10/12/2023] [Accepted: 01/03/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Blood-brain barrier (BBB) could aggravate cerebral ischemia injury. Dexmedetomidine (Dex) has been believed to play a protective role in cerebral ischemia injury-induced BBB injury. METHODS Middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation (OGD) models were established to simulate cerebral ischemia injury. Animal experiments included 4 groups, Sham, MCAO, MCAO+Dex, MCAO+Dex+sh-CCN1. Generally applicable gene set enrichment analysis was performed to analyze gene expression difference. Total collagen content and Evans blue staining were performed to measure infarct ratio and BBB breakdown, respectively. The cell apoptosis, mRNA and protein expression were measured through flow cytometry, PCR, and western blotting, respectively. The levels of IL-1β, TNF-α, and IL-6 in serum were measured with commercial ELISA kits. RESULTS Dex greatly promoted the expression level of CCN1. Dex suppressed cerebral ischemia injury, increased tight junction protein expression, improved the memory ability and neurological function of MCAO rats through targeting CCN1. The significant increase of inflammatory factors in the serum of MCAO rats were suppressed by Dex. Dex suppressed OGD induced increase of HRP permeability and promoting tight junction protein expression in vitro through regulating CCN1. The neurological function evaluation was performed with Neurological Severity Score (NSS) and Longa Score Scale. CONCLUSIONS Dex could remarkably alleviate cerebral ischemia injury by inhibiting BBB breakdown, inflammatory response, and promoting neurological function and tight junction protein expression via up-regulating CCN1. This study might provide a novel therapeutic target for the prevention and treatment of cerebral ischemia injury-induced BBB.
Collapse
Affiliation(s)
- Shuangmei Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Xuepeng Jia
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Bo Liu
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shenyang 110004, Liaoning, China
| | - Yue Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Hong Yin
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| |
Collapse
|
8
|
Dexmedetomidine: An Alternative to Pain Treatment in Neonatology. CHILDREN 2023; 10:children10030454. [PMID: 36980013 PMCID: PMC10047358 DOI: 10.3390/children10030454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Infants might be exposed to pain during their admissions in the neonatal intensive care unit [NICU], both from their underlying conditions and several invasive procedures required during their stay. Considering the particularities of this population, recognition and adequate management of pain continues to be a challenge for neonatologists and investigators. Diverse therapies are available for treatment, including non-pharmacological pain management measures and pharmacological agents (sucrose, opioids, midazolam, acetaminophen, topical agents…) and research continues. In recent years one of the most promising drugs for analgesia has been dexmedetomidine, an alpha-2 adrenergic receptor agonist. It has shown a promising efficacy and safety profile as it produces anxiolysis, sedation and analgesia without respiratory depression. Moreover, studies have shown a neuroprotective role in animal models which could be beneficial to neonatal population, especially in preterm newborns. Side effects of this therapy are mainly cardiovascular, but in most studies published, those were not severe and did not require specific therapeutic measures for their resolution. The main objective of this article is to summarize the existing literature on neonatal pain management strategies available and review the efficacy of dexmedetomidine as a new therapy with increasing use in the NICU.
Collapse
|
9
|
Dexmedetomidine Attenuates LPS-Stimulated Alveolar Type II Cells’ Injury through Upregulation of miR-140-3p and Partial Suppression of PD-L1 Involving Inactivating JNK-Bnip3 Pathway. Can Respir J 2022; 2022:8433960. [PMID: 35958433 PMCID: PMC9357803 DOI: 10.1155/2022/8433960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/10/2022] [Accepted: 07/14/2022] [Indexed: 11/24/2022] Open
Abstract
Dexmedetomidine (DEX), which is reported to be a newly discovered, novel α-2 adrenoceptor agonist, is known to exhibit anti-inflammatory properties in several diseases. DEX regulates inflammation-related signaling pathways and genes through interactions with several miRNAs. This study verified that expression levels of miR-140-3p were diminished when alveolar type II cells were exposed to LPS. However, the levels of miR-140-3p were confirmed as showing an increase with DEX treatment. These observations revealed that the expression of miR-140-3p was related to the beneficial effects that accompanied the DEX treatment of LPS-induced ALI. In addition, PD-1/PD-L1 expression increased extensively when RLE-6TN cells were induced by LPS. The increased expression was reduced after treatment with DEX. Thus, it appears that the PD-L1 expression was targeted directly by miR-140-3p, resulting in the partial repression of PD-L1 levels, which involved the inhibition of p-JNK and Bnip3 expression. Therefore, DEX was shown to inhibit the PD-L1 expression by promoting partially increased miR-140-3p levels in RLE-6TN cells. DEX also inactivated the JNK-Bnip3 pathway, resulting in the inhibition of inflammation and alleviating alveolar type II cell injury.
Collapse
|
10
|
Ji SH, Kang P, Song IS, Jang YE, Lee JH, Kim JT, Kim HS, Kim EH. The effect of dexmedetomidine on neuroprotection in pediatric cardiac surgery patients: study protocol for a prospective randomized controlled trial. Trials 2022; 23:271. [PMID: 35395776 PMCID: PMC8991922 DOI: 10.1186/s13063-022-06217-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/26/2022] [Indexed: 12/03/2022] Open
Abstract
Background Infants undergoing cardiac surgery under cardiopulmonary bypass are vulnerable to postoperative neurodevelopmental delays. Dexmedetomidine has been shown to have protective effects on the heart, kidneys, and brain in animals and adults undergoing cardiac surgery with cardiopulmonary bypass. We hypothesized that dexmedetomidine would have a neuroprotective effect on infants undergoing cardiopulmonary bypass and planned a prospective randomized controlled trial with postoperative neurodevelopment measurements. Methods This is a single-center, prospective, double-blinded, randomized controlled trial with 1:1 allocation. A cohort of 160 infants undergoing cardiac surgery with cardiopulmonary bypass will be enrolled. After induction, dexmedetomidine will be infused with a loading dose of 1 μg/kg and a maintenance dose of 0.5 μg/kg/h or the same amount of normal saline will be administered. Upon initiation of cardiopulmonary bypass, an additional dose of dexmedetomidine (0.01 μg/cardiopulmonary priming volume) will be mixed with the cardiopulmonary bypass circuit. The primary outcome will be the proportion of infants who score lower than 85 in any of the cognitive, language, or motor Bayley scales of infant development-III tests 1 year after the surgery. Other feasible outcome measures will include differences in plasma glial fibrillary acidic protein, troponin I, interleukin-6, urinary neutrophil gelatinase-associated lipocalin, and perioperative major adverse events. The results of the Bayley scales of infant development-III test from the study group and the control group will be compared using a chi-squared test under intention-to-treat analysis. A generalized estimating equation will be used to analyze repeated measurements over time. Discussion This study will enable us to assess whether the use of dexmedetomidine can alter the early neurodevelopmental outcome in infants undergoing cardiac surgery with cardiopulmonary bypass and also estimate effects of dexmedetomidine on other organs. Trial registration ClinicalTrials.gov NCT04484922. Registered on 24 July 2020 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06217-9.
Collapse
Affiliation(s)
- Sang-Hwan Ji
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Pyoyoon Kang
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - In-Sun Song
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Young-Eun Jang
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ji-Hyun Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jin-Tae Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hee-Soo Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Eun-Hee Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
11
|
Chen S, Zhang S, Wu H, Zhang D, You G, You J, Zheng N. Protective effect of phillyrin against cerebral ischemia/reperfusion injury in rats and oxidative stress-induced cell apoptosis and autophagy in neurons. Bioengineered 2022; 13:7940-7950. [PMID: 35291908 PMCID: PMC9278963 DOI: 10.1080/21655979.2022.2042142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
This study explored the role and potential molecular mechanism of phillyrin in cerebral ischemia/reperfusion (I/R) injury. The rat middle cerebral artery occlusion (MCAO)/R model was constructed, and cerebral infarction volume, brain water content, and neurological score were measured. Neuron morphological structures in brain tissues and primary neuron apoptosis were detected using hematoxylin and eosin (H&E) staining and Hoechst 33258 staining, respectively. In MCAO/R rats, phillyrin markedly reduced cerebral infarction volume, neurological score, and brain water content and inhibited neuron apoptosis. In vitro experiments showed that phillyrin remarkably increased viability and decreased lactate dehydrogenase (LDH) release of H2O2-injured neurons. Moreover, phillyrin remarkably downregulated the proportion of apoptosis-related protein B-associated X (Bax)/B-cell lymphoma protein 2 (Bcl-2) and reduced procaspase-3, phospho-Akt (p-Akt-1), and phosphorylation-mammalian target of rapamycin (p-mTOR) levels in H2O2-injured neurons. Furthermore, phosphatidylinositol-3 kinase (PI3K) inhibitor ZSTK474 weakened the effects of phillyrin on p-mTOR, p-Akt-1, characteristic proteins of autophagy 3-II (LC3-II) and beclin-1 levels, and H2O2-induced neuronal apoptosis and autophagy. Taken together, phillyrin alleviates I/R injury by inhibiting neuronal cell apoptosis and autophagy pathway, which may provide a new treatment strategy for cerebral I/R injury.
Collapse
Affiliation(s)
- Shu Chen
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Shan Zhang
- Department of Neurology, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Honggang Wu
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Daobao Zhang
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Guoliang You
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Jing You
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Niandong Zheng
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, Sichuan, China
| |
Collapse
|
12
|
Unchiti K, Leurcharusmee P, Samerchua A, Pipanmekaporn T, Chattipakorn N, Chattipakorn SC. The potential role of dexmedetomidine on neuroprotection and its possible mechanisms: Evidence from in vitro and in vivo studies. Eur J Neurosci 2021; 54:7006-7047. [PMID: 34561931 DOI: 10.1111/ejn.15474] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
Neurological disorders following brain injuries and neurodegeneration are on the rise worldwide and cause disability and suffering in patients. It is crucial to explore novel neuroprotectants. Dexmedetomidine, a selective α2-adrenoceptor agonist, is commonly used for anxiolysis, sedation and analgesia in clinical anaesthesia and critical care. Recent studies have shown that dexmedetomidine exerts protective effects on multiple organs. This review summarized and discussed the current neuroprotective effects of dexmedetomidine, as well as the underlying mechanisms. In preclinical studies, dexmedetomidine reduced neuronal injury and improved functional outcomes in several models, including hypoxia-induced neuronal injury, ischaemic-reperfusion injury, intracerebral haemorrhage, post-traumatic brain injury, anaesthetic-induced neuronal injury, substance-induced neuronal injury, neuroinflammation, epilepsy and neurodegeneration. Several mechanisms are associated with the neuroprotective function of dexmedetomidine, including neurotransmitter regulation, inflammatory response, oxidative stress, apoptotic pathway, autophagy, mitochondrial function and other cell signalling pathways. In summary, dexmedetomidine has the potential to be a novel neuroprotective agent for a wide range of neurological disorders.
Collapse
Affiliation(s)
- Kantarakorn Unchiti
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prangmalee Leurcharusmee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Artid Samerchua
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Tanyong Pipanmekaporn
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
13
|
Zhu Y, Zhao H, Zhang W, Ma X, Liu Y. Dexmedetomidine attenuates neuronal injury induced by cerebral ischemia‑reperfusion by regulating miR‑199a. Mol Med Rep 2021; 24:574. [PMID: 34109426 PMCID: PMC8201450 DOI: 10.3892/mmr.2021.12213] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/21/2021] [Indexed: 12/30/2022] Open
Abstract
As is well known, dexmedetomidine (DEX) serves a neuroprotective role in cerebral ischemia‑reperfusion (CIR) injury, and microRNA (miR)‑199a has been re‑ported to be associated with IR injury. However, the association between DEX and miR‑199a in CIR injury remains unknown. Thus, the aim of the present study was to verify whether the neuroprotective effect of DEX on cerebral ischemia‑reperfusion rats is associated with miR‑199a. A rat model of CIR was established, and the modified neurological severity score (mNSS) was evaluated. The effect of DEX on the patholog‑ical structure of the cerebral cortex in CIR rats was observed by hematoxylin and eosin and Nissl staining. Reverse transcription‑quantitative PCR was used to analyze the expression levels of miR‑199a in brain tissue following intracerebroventricular injection of miR‑199a antagomir. The co‑expression of NeuN and microtubule‑associated proteins 1A/1B light chain 3B in the cerebral cortex was analyzed by immunofluorescence staining. Western blotting and immunohistochemistry were performed to analyze the expression of autophagy‑associated proteins in the brain tissue. DEX inhibited the expression of miR‑199a, decreased the mNSS and improved pathological damage to the cerebral cortex. DEX also inhibited autophagy and expression levels of associated proteins and decreased nerve cell injury. In conclusion, DEX inhibited expression of miR‑199a and improved neurocyte injury induced by CIR.
Collapse
Affiliation(s)
- Yulin Zhu
- Department of Anesthesiology, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Huatang Zhao
- Department of Anesthesiology, The Second Hospital of Liaocheng Affiliated to Shan-dong First Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Wenshan Zhang
- Department of Anesthesiology, Laixi People's Hospital, Qingdao, Shandong 266600, P.R. China
| | - Xingang Ma
- Department of Anesthesiology, Zibo Maternal and Child Health Hospital, Zibo, Shandong 255000, P.R. China
| | - Ye Liu
- Department of Anesthesiology, Zibo Maternal and Child Health Hospital, Zibo, Shandong 255000, P.R. China
| |
Collapse
|
14
|
Zhang Q, Dai J, Song Z, Guo Y, Deng S, Yu Y, Li T, Zhang Y. Anti-Inflammatory Dipeptide, a Metabolite from Ambioba Secretion, Protects Cerebral Ischemia Injury by Blocking Apoptosis Via p-JNK/Bax Pathway. Front Pharmacol 2021; 12:689007. [PMID: 34220513 PMCID: PMC8249563 DOI: 10.3389/fphar.2021.689007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/03/2021] [Indexed: 12/03/2022] Open
Abstract
MQ (l-methionyl-l-glutamic acid), anti-inflammatory dipeptide, is one of the metabolites of monocyte locomotion inhibitory factor, a thermostable pentapeptide secreted by Entamoeba histolytica. Monocyte locomotion inhibitory factor injection has been approved as an investigational drug for the potential neural protection in acute ischemic stroke. This study further investigated the neuroprotective effect of MQ in ischemic brain damage. Ischemia-reperfusion injury of the brain was induced in the rat model by middle cerebral artery occlusion. 2,3,5-triphenyltetrazolium chloride staining assay was used to measure cerebral infarction areas in rats. Laser Doppler measurement instrument was used to detect blood flow changes in the rat model. Nissl staining and NeuN staining were utilized to observe the numbers and structures of neuron cells, and the pathological changes in the brain tissues were examined by hematoxylin–eosin staining. Terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) staining was used to assess cell apoptosis. The changes in oxidative stress indexes, superoxide dismutase and malondialdehyde (MDA), were measured in serum. Methyl thiazolyl tetrazolium was used to measure the survival rates of PC12 cells. Flow cytometry assessed the apoptosis rates and the levels of reactive oxygen species. Real-time PCR was used to evaluate the mRNA expression levels, and Western blotting was used to analyze the changes in protein levels of p-JNK, Bax, cleaved Caspase3. We revealed that MQ improved neurobehavior, decreased cerebral infarction areas, altered blood flow volume, and the morphology of the cortex and hippocampus. On the other hand, it decreased the apoptosis of cortical neurons and the levels of MDA, and increased the levels of superoxide dismutase. In vitro studies demonstrated that MQ enhanced the cell survival rates and decreased the levels of reactive oxygen species. Compared to the oxygen-glucose deprivation/reperfusion group, the protein and mRNA expressions of p-JNK, Bax, cleaved Caspase3 was decreased significantly. These findings suggested that MQ exerts a neuroprotective effect in cerebral ischemia by blocking apoptosis via the p-JNK/Bax pathway.
Collapse
Affiliation(s)
- Qian Zhang
- School of Medicine, Shanghai University, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Jinwei Dai
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhibing Song
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Yuchen Guo
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Shanshan Deng
- School of Medicine, Shanghai University, Shanghai, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, China
| | - Tiejun Li
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
15
|
Tan L, Liu L, Yao J, Piao C. miR-145-5p attenuates inflammatory response and apoptosis in myocardial ischemia-reperfusion injury by inhibiting (NADPH) oxidase homolog 1. Exp Anim 2021; 70:311-321. [PMID: 33658472 PMCID: PMC8390312 DOI: 10.1538/expanim.20-0160] [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: 02/05/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a common complication following reperfusion therapy that involves a series of immune or apoptotic reactions. Studies have revealed the potential roles of miRNAs in I/R injury. Herein, we established a myocardial I/R model in rats and a hypoxia/reoxygenation (H/R) model in H9c2 cells and investigated the effect of miR-145-5p on myocardial I/R injury. After 3 h or 24 h of reperfusion, left ventricular end-systolic pressure (LVESP), ejection fraction (EF), and fractional shortening (FS) were obviously decreased, and left ventricular end-diastolic pressure (LVEDP) was increased. Meanwhile, I/R induced an increase in myocardial infarction area. Moreover, a decrease in miR-145-5p and increase in (NADPH) oxidase homolog 1 (NOH-1) were observed following I/R injury. With this in mind, we performed a luciferase reporter assay and demonstrated that miR-145-5p directly bound to NOH-1 3' untranslated region (UTR). Furthermore, miR-145-5p mimics decreased the levels of tumor necrosis factor (TNF)-α, IL-1β, and IL-6 via oxygen and glucose deprivation/reperfusion (OGD/R) stimulation. Upregulation of miR-145-5p increased cell viability and reduced apoptosis accompanied by downregulation of Bax, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP) and upregulation of Bcl2. In addition, miR-145-5p overexpression increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) content under OGD/R stress. Notably, NOH-1 could significantly abrogate the above effects, suggesting that it is involved in miR-145-5p-regulated I/R injury. In summary, our findings indicated that miR-145-5p/NOH-1 has a protective effect on myocardial I/R injury by inhibiting the inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Lili Tan
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Limin Liu
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Jian Yao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Chenghao Piao
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| |
Collapse
|
16
|
Li YQ, Hui ZR, Tao T, Shao KY, Liu Z, Li M, Gu LL. Protective effect of hypoxia inducible factor-1α gene therapy using recombinant adenovirus in cerebral ischaemia-reperfusion injuries in rats. PHARMACEUTICAL BIOLOGY 2020; 58:438-446. [PMID: 32432963 PMCID: PMC7301712 DOI: 10.1080/13880209.2020.1762667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 03/24/2020] [Accepted: 04/26/2020] [Indexed: 06/01/2023]
Abstract
Context: Hypoxia-inducible factor-1α (HIF-1α)-induced genes can improve blood circulation.Objective: To investigate brain protective effect of recombinant adenovirus-mediated HIF-1α (AdHIF-1α) expression and its mechanism.Materials and methods: Male SD rats were used to establish focal cerebral ischaemia-reperfusion (CIR) injury models and randomly divided into normal, sham, CIR, Ad and AdHIF-1α groups. Ad or AdHIF-1α (108 pfu/10 µL) were administered into lateral ventricle of rats in Ad and AdHIF-1α groups. Modified neurological severity score (mNSS), brain water content (BWC) and cerebral infarct volumes (CIVs) were analyzed, and HE staining was performed using the brain tissues. Furthermore, the expression of caspase-3 and HSP90 was analyzed using qRT-PCR and Western blotting.Results: Compared to CIR (mNSS, 8.52 ± 0.52; CIV, 0.22 ± 0.01) and Ad groups (mNSS, 8.83 ± 0.41; CIV, 0.22 ± 0.02), mNSS and CIV were significantly decreased in AdHIF-1α group (mNSS, 6.03 ± 0.61; CIV, 0.11 ± 0.01) at 72 h (p < 0.05). With prolonged reperfusion time (6 h to 72 h), BWC of all rats increased gradually, although the increase was markedly less in AdHIF-1α group (78.15 ± 0.16 to 87.01 ± 0.31) compared to that in CIR (78.77 ± 0.60 to 89.74 ± 0.34) and Ad groups (78.77 ± 0.35 to 89.71 ± 0.27) (p < 0.01). There were significantly greater pathological changes in the neurons in AdHIF-1α group at 72 h following CIR. Furthermore, expression of caspase-3 (p < 0.01) down-regulated and HSP90 up-regulated (p < 0.05) at mRNA and protein levels in AdHIF-1α group.Discussion and conclusions: HIF‑1α gene therapy is neuroprotective towards the CIR rat model. HIF-1α may be a candidate gene for the treatment of ischaemic brain injury.
Collapse
Affiliation(s)
- Ya-Qi Li
- Department of Emergency, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| | - Zhi-Rong Hui
- Department of Rehabilitation Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| | - Tao Tao
- Department of Rehabilitation Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| | - Kang-Yu Shao
- Department of Neurology, Chengdu Aerospace Hospital, Chengdu, Sichuan Province, China
| | - Zhi Liu
- Department of Pharmacy, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Min Li
- Department of Rehabilitation Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| | - Li-Ling Gu
- Department of Rehabilitation Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| |
Collapse
|
17
|
Huang Y, Pan L, Wu T. Improvement of cerebral ischemia-reperfusion injury by L-3-n-butylphthalide through promoting angiogenesis. Exp Brain Res 2020; 239:341-350. [PMID: 33180154 DOI: 10.1007/s00221-020-05978-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury may lead to a poor prognosis for ischemic stroke patients after reperfusion therapy, and currently, lacks effective therapeutic intervention. This study aimed to investigate the effects of L-3-n-butylphthalide (L-NBP) on cerebral I/R injury in rats. Rat models of cerebral I/R injury were established using the middle cerebral artery occlusion/refusion (MACO/R) surgery and were administrated intragastrically with L-NBP or vehicle. We found that L-NBP attenuated the histological damages and reduced the brain hematoma in MACO/R rats. L-NBP also significantly improved the neurological function, alleviated the brain edema, and reduced the permeability of blood-brain barrier of MACO/R rats. Moreover, we detected that L-NBP considerably facilitated microvessel formation in the lesion area of brain in MACO/R rats. Finally, we found that L-NBP significantly increased the protein and mRNA expression levels of Nrf2, HIF-1α, and VEGF in the brain of MACO/R rats. In conclusion, our results demonstrated that L-NBP exerted significant beneficial effects on cerebral I/R injury in rats through promoting angiogenesis, which may be associated with the activation of Nrf2/HIF-1α/VEGF signaling pathway. Our results suggested that L-NBP could be a potential therapeutic drug for cerebral I/R injury.
Collapse
Affiliation(s)
- Ying Huang
- Department of Pharmacy, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China
| | - Lishou Pan
- Department of Neurology, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China
| | - Ting Wu
- Department of Pharmacy, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China.
| |
Collapse
|
18
|
Rao Z, Shen D, Chen J, Jin L, Wu X, Chen M, Li L, Chu M, Lin J. Basic Fibroblast Growth Factor Attenuates Injury in Myocardial Infarction by Enhancing Hypoxia-Inducible Factor-1 Alpha Accumulation. Front Pharmacol 2020; 11:1193. [PMID: 32848793 PMCID: PMC7427464 DOI: 10.3389/fphar.2020.01193] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 11/13/2022] Open
Abstract
Background The combination of antiapoptotic and angiogenic actions may represent a pharmacotherapeutic strategy for the treatment of myocardial infarction. Fibroblast growth factor (FGF) is expressed in various cell types including endothelial and muscle cells and promotes their survival, migration, and proliferation. Methods and Results Myocardial microvascular endothelial cells were divided into four treatment groups, the sham, hypoxia, basic FGF (bFGF), and bFGF plus 2-methoxyestradiol groups, and subjected to in vitro apoptotic analysis and Matrigel assays. An in vivo model of myocardial infarction was established by ligaturing the left coronary artery of mice in the four treatment groups. Cardiac performance, myocardial injury, endothelial cell angiogenesis, and myocardial apoptosis were assessed. bFGF administration after myocardial infarction improved cardiac function and cell viability, attenuated myocardial injury and apoptosis, and enhanced angiogenesis. Western blotting of HIF-1α, p-AKT, VEGF, p53, BAX, and Bcl-2 showed that bFGF increased HIF-1α, p-AKT, VEGF, and Bcl-2 and decreased BAX protein levels. Conclusion The results of the present study indicated that bFGF attenuates myocardial injury by inhibiting apoptosis and promoting angiogenesis via a novel HIF-1α-mediated mechanism and a potential utility of bFGF in protecting against myocardial infarction.
Collapse
Affiliation(s)
- Zhiheng Rao
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Danping Shen
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiahui Chen
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Cardiology, Taishun People's Hospital, Wenzhou, China
| | - Lushen Jin
- Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueping Wu
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Ming Chen
- Department of Clinical Medicine, The First School of Wenzhou Medical University, Wenzhou, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Maoping Chu
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiafeng Lin
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Meng Q, Guo P, Jiang Z, Bo L, Bian J. Dexmedetomidine inhibits LPS-induced proinflammatory responses via suppressing HIF1α-dependent glycolysis in macrophages. Aging (Albany NY) 2020; 12:9534-9548. [PMID: 32433037 PMCID: PMC7288940 DOI: 10.18632/aging.103226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has been reported to exert an anti-inflammatory effect in several animal models, but the mechanism remains unclear. Previous studies have shown that hypoxia inducible factor 1α-induced glycolysis is essential for the activation of inflammatory macrophages. However, whether dexmedetomidine influences hypoxia inducible factor 1α-induced glycolysis and thus exerts an anti-inflammatory effect has been poorly investigated. This study aims to elucidate the anti-inflammatory mechanism of dexmedetomidine involving the hypoxia inducible factor 1α-dependent glycolytic pathway. We showed that dexmedetomidine could suppress lipopolysaccharide-induced inflammatory cytokine production; inhibit the extracellular acidification rate, glucose consumption and lactate production; and decrease the expression of glycolytic genes in macrophages. The enhancement of glycolysis by the granulocyte-macrophage colony-stimulating factor or higher concentration of glucose could reverse the anti-inflammatory effect of dexmedetomidine on lipopolysaccharide-treated macrophages. Moreover, dexmedetomidine significantly inhibited the upregulation of hypoxia inducible factor 1α at the mRNA and protein levels. Genetic inhibition of hypoxia inducible factor 1α expression could reverse the anti-inflammatory effect of dexmedetomidine. Taken together, our results indicate that dexmedetomidine attenuates lipopolysaccharide-induced proinflammatory responses partially by suppressing hypoxia inducible factor 1α-dependent glycolysis in macrophages.
Collapse
Affiliation(s)
- Qingyuan Meng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Pinhao Guo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
20
|
König KS, Verhaar N, Hopster K, Pfarrer C, Neudeck S, Rohn K, Kästner SBR. Ischaemic preconditioning and pharmacological preconditioning with dexmedetomidine in an equine model of small intestinal ischaemia-reperfusion. PLoS One 2020; 15:e0224720. [PMID: 32348301 PMCID: PMC7190151 DOI: 10.1371/journal.pone.0224720] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Small intestinal strangulation associated with ischaemia-reperfusion injury (IRI) is common in horses. In laboratory animals IRI can be ameliorated by ischaemic preconditioning (IPC) and pharmacological preconditioning (PPC) with dexmedetomidine. The aim of this study was to determine the effect of PPC with dexmedetomidine or IPC in an equine model of small intestinal ischaemia-reperfusion (IR). In a randomized controlled experimental trial, 15 horses were assigned to three groups: control (C), IPC, and PPC with dexmedetomidine (DEX). All horses were placed under general anaesthesia and 90% jejunal ischaemia was induced for 90 minutes, followed 30 minutes of reperfusion. In group IPC, three short bouts of ischaemia and reperfusion were implemented, and group DEX received a continuous rate infusion of dexmedetomidine prior to the main ischaemia. Jejunal biopsies were collected before ischaemia (P), and at the end of ischaemia (I) and reperfusion (R). Mucosal injury was assessed by the Chiu-Score, inflammatory cells were stained by cytosolic calprotectin. The degree of apoptosis and cell necrosis was assessed by cleaved-caspase-3 and TUNEL. Parametric data were analyzed by two-way ANOVA for repeated measurements followed by Dunnetts t-test. Non parametric data were compared between groups at the different time points by a Kruskal-Wallis-Test and a Wilcoxon-2-Sample-test. The mucosal injury score increased during I in all groups. After reperfusion, IRI further progressed in group C, but not in IPC and DEX. In all groups the number of cleaved caspase-3 and TUNEL positive cells increased from P to I. The number of TUNEL positive cells were lower in group DEX compared to group C after I and R. Infiltration with calprotectin positive cells was less pronounced in group DEX compared to group C, whereas in group IPC more calprotectin positive cells were seen. In conclusion, IPC and DEX exert protective effects in experimental small intestinal ischaemia in horses.
Collapse
Affiliation(s)
- Kathrin S. König
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Verhaar
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus Hopster
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christiane Pfarrer
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Stephan Neudeck
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Karl Rohn
- Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sabine B. R. Kästner
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
- Clinic for Small Animals, University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
21
|
Zhai M, Liu C, Li Y, Zhang P, Yu Z, Zhu H, Zhang L, Zhang Q, Wang J, Wang J. Dexmedetomidine inhibits neuronal apoptosis by inducing Sigma-1 receptor signaling in cerebral ischemia-reperfusion injury. Aging (Albany NY) 2019; 11:9556-9568. [PMID: 31682592 PMCID: PMC6874446 DOI: 10.18632/aging.102404] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022]
Abstract
Dexmedetomidine is known to alleviate cerebral ischemia-reperfusion injury (CIRI). We established a rat model of CIRI, which exhibited higher neurological deficit scores and a greater number of apoptotic cells in the cerebral ischemic penumbra than controls. Dexmedetomidine reversed the neuronal apoptosis and improved neurological function in this model. We then examined Sigma-1 receptor (Sig-1R) expression on the endoplasmic reticulum (ER) in brain tissues at different reperfusion time points. Sig-1R expression increased with CIRI and decreased with increasing reperfusion times. After 24 hours of reperfusion, dexmedetomidine upregulated Sig-1R expression, and ER stress proteins (GRP78, CHOP, JNK and Caspase-3) were detected in brain tissues with Western blotting. Moreover, GRP78 expression followed a pattern similar to Sig-1R. Dexmedetomidine induced GRP78 expression but inhibited CHOP, Caspase-3 and phosphorylated-JNK expression in brain tissues. A Sig-1R-specific inhibitor reduced GRP78 expression and partially inhibited the upregulation of GRP78 by dexmedetomidine. The inhibitor also increased CHOP and Caspase-3 expression and partially reversed the inhibitory effects of dexmedetomidine on these pro-apoptotic ER stress proteins. These results suggest that dexmedetomidine at least partially inhibits ER stress-induced apoptosis by activating Sig-1R, thereby attenuating brain damage after 24 hours of ischemia-reperfusion.
Collapse
Affiliation(s)
- Meili Zhai
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Chong Liu
- Department of Anesthesiology, Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, Tianjin 300140, China
| | - Yuexiang Li
- Department of Anesthesiology, Tianjin Xiqing Hospital, Tianjin 300380, China
| | - Peijun Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Zhiqiang Yu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - He Zhu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Li Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Qian Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jianbo Wang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jinhua Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province 318000, China
| |
Collapse
|
22
|
Anti-Neuroinflammatory Effect of Alantolactone through the Suppression of the NF-κB and MAPK Signaling Pathways. Cells 2019; 8:cells8070739. [PMID: 31323885 PMCID: PMC6678480 DOI: 10.3390/cells8070739] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 01/06/2023] Open
Abstract
Neuroinflammation is a major cause of central nervous system (CNS) damage and can result in long-term disability and mortality. Therefore, the development of effective anti-neuroinflammatory agents for neuroprotection is vital. To our surprise, the naturally occurring molecule alantolactone (Ala) was reported to significantly inhibit tumor growth and metastasis as a result of its excellent anti-inflammatory effects. Thus, we proposed that it could also act as an anti-neuroinflammatory agent. Thus, in this study, a coculture system of BV2 cells and PC12 cells were used as an in vitro neuroinflammatory model to investigate the anti-neuroinflammatory mechanism of Ala. The results indicated that Ala downregulated the expression of proinflammatory factors by suppressing the nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. Further evaluation using a middle cerebral artery occlusion and reperfusion (MCAO/R) rat model supported the conclusion that Ala could (1) alleviate cerebral ischemia-reperfusion injury; (2) reduce neurological deficits, cerebral infarct volume, and brain edema; and (3) attenuate the apoptosis and necrosis of neurons. In sum, Ala demonstrates anti-neuroinflammatory properties that contribute to the amelioration of CNS damage, and it could be a promising candidate for future applications in CNS injury treatment.
Collapse
|
23
|
Sun Y, Zhu Y, Zhong X, Chen X, Wang J, Ying G. Crosstalk Between Autophagy and Cerebral Ischemia. Front Neurosci 2019; 12:1022. [PMID: 30692904 PMCID: PMC6339887 DOI: 10.3389/fnins.2018.01022] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
With the use of advanced electron microscopy and molecular biology tools, several studies have shown that autophagy is involved in the development of ischemic stroke. A series of molecular mechanisms are involved in the regulation of autophagy. In this work, the possible molecular mechanisms involved in autophagy during ischemic stroke were reviewed and new potential targets for the study and treatment of ischemic stroke were provided.
Collapse
Affiliation(s)
- Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yuanhan Zhu
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xinle Chen
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Jun Wang
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Guozheng Ying
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|