1
|
Jiang L, Xiong W, Yang Y, Qian J. Insight into Cardioprotective Effects and Mechanisms of Dexmedetomidine. Cardiovasc Drugs Ther 2024; 38:1139-1159. [PMID: 38869744 DOI: 10.1007/s10557-024-07579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Cardiovascular disease remains the leading cause of death worldwide. Dexmedetomidine is a highly selective α2 adrenergic receptor agonist with sedative, analgesic, anxiolytic, and sympatholytic properties, and several studies have shown its possible protective effects in cardiac injury. The aim of this review is to further elucidate the underlying cardioprotective mechanisms of dexmedetomidine, thus suggesting its potential in the clinical management of cardiac injury. RESULTS AND CONCLUSION Our review summarizes the findings related to the involvement of dexmedetomidine in cardiac injury and discusses the results in the light of different mechanisms. We found that numerous mechanisms may contribute to the cardioprotective effects of dexmedetomidine, including the regulation of programmed cell death, autophagy and fibrosis, alleviation of inflammatory response, endothelial dysfunction and microcirculatory derangements, improvement of mitochondrial dysregulation, hemodynamics, and arrhythmias. Dexmedetomidine may play a promising and beneficial role in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Leyu Jiang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Xiong
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Cai XQ, Yang H, Liang BQ, Deng CC, Xue HY, Zhang JJ, Wang XZ. Glutamate rescues heat stress-induced apoptosis of Sertoli cells by enhancing the activity of antioxidant enzymes and activating the Trx1-Akt pathway in vitro. Theriogenology 2024; 223:1-10. [PMID: 38642435 DOI: 10.1016/j.theriogenology.2024.04.005] [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: 06/17/2023] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Heat stress reduces the number of Sertoli cells, which is closely related to an imbalanced redox status. Glutamate functions to maintain the equilibrium of redox homeostasis. However, the role of glutamate in heat treated Sertoli cells remains unclear. Herein, Sertoli cells from 3-week-old piglets were treated at 44 °C for 30 min (heat stress). Glutamate levels increased significantly following heat stress treatment, followed by a gradual decrease during recovery, while glutathione (GSH) showed a gradual increase. The addition of exogenous glutamate (700 μM) to Sertoli cells before heat stress significantly reduced the heat stress-induced apoptosis rate, mediated by enhanced levels of antioxidant substances (superoxide dismutase (SOD), total antioxidant capacity (TAC), and GSH) and reduced levels of oxidative substances (reactive oxygen species (ROS) and malondialdehyde (MDA)). Glutamate addition to Sertoli cells before heat stress upregulated the levels of glutamate-cysteine ligase, modifier subunit (Gclm), glutathione synthetase (Gss), thioredoxin (Trx1) and B-cell leukemia/lymphoma 2 (Bcl-2), and the ratio of phosphorylated Akt (protein kinase B)/total Akt. However, it decreased the levels of Bcl2-associated X protein (Bax) and cleaved-caspase 3. Addition of the inhibitor of glutaminase (Gls1), Bptes (Bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide, 30 μM)to Sertoli cells before heat stress reversed these effects. These results inferred that glutamate rescued heat stress-induced apoptosis in Sertoli cells by enhancing activity of antioxidant enzymes and activating the Trx1-Akt pathway. Thus, glutamate supplementation might represent a novel strategy to alleviate the negative effect of heat stress.
Collapse
Affiliation(s)
- Xia-Qing Cai
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Huan Yang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Bing-Qian Liang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Cheng-Chen Deng
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Hong-Yan Xue
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Jiao-Jiao Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Xian-Zhong Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| |
Collapse
|
3
|
Li R, Zhang Y, Zhu Q, Wu Y, Song W. The role of anesthesia in peri‑operative neurocognitive disorders: Molecular mechanisms and preventive strategies. FUNDAMENTAL RESEARCH 2024; 4:797-805. [PMID: 39161414 PMCID: PMC11331737 DOI: 10.1016/j.fmre.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/21/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Peri-operative neurocognitive disorders (PNDs) include postoperative delirium (POD) and postoperative cognitive dysfunction (POCD). Children and the elderly are the two populations most vulnerable to the development of POD and POCD, which results in both high morbidity and mortality. There are many factors, including neuroinflammation and oxidative stress, that are associated with POD and POCD. General anesthesia is a major risk factor of PNDs. However, the molecular mechanisms of PNDs are poorly understood. Dexmedetomidine (DEX) is a useful sedative agent with analgesic properties, which significantly improves POCD in elderly patients. In this review, the current understanding of anesthesia in PNDs and the protective effects of DEX are summarized, and the underlying mechanisms are further discussed.
Collapse
Affiliation(s)
- Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qinxin Zhu
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Yili Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| | - Weihong Song
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| |
Collapse
|
4
|
Zhang R, Xie K, Lian Y, Hong S, Zhu Y. Dexmedetomidine ameliorates x-ray-induced myocardial injury via alleviating cardiomyocyte apoptosis and autophagy. BMC Cardiovasc Disord 2024; 24:323. [PMID: 38918713 PMCID: PMC11201331 DOI: 10.1186/s12872-024-03988-8] [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: 10/25/2023] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Radiotherapy is a primary local treatment for tumors, yet it may lead to complications such as radiation-induced heart disease (RIHD). Currently, there is no standardized approach for preventing RIHD. Dexmedetomidine (Dex) is reported to have cardio-protection effects, while its role in radiation-induced myocardial injury is unknown. In the current study, we aimed to evaluate the radioprotective effect of dexmedetomidine in X-ray radiation-treated mice. METHODS 18 male mice were randomized into 3 groups: control, 16 Gy, and 16 Gy + Dex. The 16 Gy group received a single dose of 16 Gy X-ray radiation. The 16 Gy + Dex group was pretreated with dexmedetomidine (30 µg/kg, intraperitoneal injection) 30 min before X-ray radiation. The control group was treated with saline and did not receive X-ray radiation. Myocardial tissues were collected 16 weeks after X-ray radiation. Hematoxylin-eosin staining was performed for histopathological examination. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was performed to assess the state of apoptotic cells. Immunohistochemistry staining was performed to examine the expression of CD34 molecule and von Willebrand factor. Besides, western blot assay was employed for the detection of apoptosis-related proteins (BCL2 apoptosis regulator and BCL2-associated X) as well as autophagy-related proteins (microtubule-associated protein 1 light chain 3, beclin 1, and sequestosome 1). RESULTS The findings demonstrated that 16 Gy X-ray radiation resulted in significant changes in myocardial tissues, increased myocardial apoptosis, and activated autophagy. Pretreatment with dexmedetomidine significantly protects mice against 16 Gy X-ray radiation-induced myocardial injury by inhibiting apoptosis and autophagy. CONCLUSION In summary, our study confirmed the radioprotective effect of dexmedetomidine in mitigating cardiomyocyte apoptosis and autophagy induced by 16 Gy X-ray radiation.
Collapse
MESH Headings
- Animals
- Autophagy/drug effects
- Autophagy/radiation effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/radiation effects
- Myocytes, Cardiac/metabolism
- Apoptosis/drug effects
- Male
- Dexmedetomidine/pharmacology
- Radiation Injuries, Experimental/prevention & control
- Radiation Injuries, Experimental/pathology
- Radiation Injuries, Experimental/metabolism
- Radiation Injuries, Experimental/drug therapy
- Radiation-Protective Agents/pharmacology
- Disease Models, Animal
- Signal Transduction/drug effects
- Mice
- Autophagy-Related Proteins/metabolism
- Mice, Inbred C57BL
- Apoptosis Regulatory Proteins/metabolism
Collapse
Affiliation(s)
- Runze Zhang
- Department of Anesthesiology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Kangjie Xie
- Department of Anesthesiology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Yanhong Lian
- Department of Anesthesiology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Shufang Hong
- Department of Anesthesiology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Yuntian Zhu
- Department of Anesthesiology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
5
|
Yang H, Zhao Y, Chen Y, Yang T, Dou X, Li J, Yang G, Feng G, Fang H, Fan H, Zhang S. Dexmedetomidine Alleviates Acute Stress-Induced Acute Kidney Injury by Attenuating Inflammation and Oxidative Stress via Inhibiting the P2X 7R/NF-κB/NLRP3 Pathway in Rats. Inflammation 2024:10.1007/s10753-024-02065-8. [PMID: 38896231 DOI: 10.1007/s10753-024-02065-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
This study aimed to investigate the potential protective effects of Dexmedetomidine (DEX) against acute kidney injury (AKI) induced by acute stress (AS). Wistar rats were divided into five groups: Control, DEX, AS, AS + DEX, and AS + A438079. The results showed that AS led to AKI by increasing inflammatory biomarkers and oxidative stress-related indicators. The acute stress model in rats was successfully established. Renal function, histopathology, oxidative stress, and inflammation were assessed. Localization of P2X7 receptor (P2X7R) was determined by immunofluorescence. Additionally, the key inflammatory proteins of the P2X7R/NF-κB/NLRP3 signaling pathway were measured by Western blotting. DEX significantly improved kidney function, alleviated kidney injury, and reduced oxidative stress and inflammation. DEX inhibited the activation of the P2X7R, decreased the expression of NF-κB, NLRP3 inflammasome, and Caspase-1, and inhibited the expression of interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα). Furthermore, DEX also alleviated AS-induced AKI by inhibiting the excessive production of reactive oxygen species (ROS) and reducing oxidative stress. In conclusion, DEX attenuates AS-induced AKI by mitigating inflammation and oxidative stress through the inhibition of the P2X7R/NF-κB/NLRP3 pathway in rats.
Collapse
Affiliation(s)
- Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Academy of Agricultural Science Branch of Animal Husbandry and Veterinary Branch, Qiqihar, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yongping Chen
- College of Veterinary Medicine, Agricultural University, Qingdao, China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Guiyan Yang
- Department of Pathology and Laboratory Medicine, Davis Health, University of California, Sacramento, CA, USA
| | - Guofeng Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Shuai Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
6
|
Yuan HX, Zhang LN, Li G, Qiao L. Brain protective effect of dexmedetomidine vs propofol for sedation during prolonged mechanical ventilation in non-brain injured patients. World J Psychiatry 2024; 14:370-379. [PMID: 38617978 PMCID: PMC11008391 DOI: 10.5498/wjp.v14.i3.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Dexmedetomidine and propofol are two sedatives used for long-term sedation. It remains unclear whether dexmedetomidine provides superior cerebral protection for patients undergoing long-term mechanical ventilation. AIM To compare the neuroprotective effects of dexmedetomidine and propofol for sedation during prolonged mechanical ventilation in patients without brain injury. METHODS Patients who underwent mechanical ventilation for > 72 h were randomly assigned to receive sedation with dexmedetomidine or propofol. The Richmond Agitation and Sedation Scale (RASS) was used to evaluate sedation effects, with a target range of -3 to 0. The primary outcomes were serum levels of S100-β and neuron-specific enolase (NSE) every 24 h. The secondary outcomes were remifentanil dosage, the proportion of patients requiring rescue sedation, and the time and frequency of RASS scores within the target range. RESULTS A total of 52 and 63 patients were allocated to the dexmedetomidine group and propofol group, respectively. Baseline data were comparable between groups. No significant differences were identified between groups within the median duration of study drug infusion [52.0 (IQR: 36.0-73.5) h vs 53.0 (IQR: 37.0-72.0) h, P = 0.958], the median dose of remifentanil [4.5 (IQR: 4.0-5.0) μg/kg/h vs 4.6 (IQR: 4.0-5.0) μg/kg/h, P = 0.395], the median percentage of time in the target RASS range without rescue sedation [85.6% (IQR: 65.8%-96.6%) vs 86.7% (IQR: 72.3%-95.3), P = 0.592], and the median frequency within the target RASS range without rescue sedation [72.2% (60.8%-91.7%) vs 73.3% (60.0%-100.0%), P = 0.880]. The proportion of patients in the dexmedetomidine group who required rescue sedation was higher than in the propofol group with statistical significance (69.2% vs 50.8%, P = 0.045). Serum S100-β and NSE levels in the propofol group were higher than in the dexmedetomidine group with statistical significance during the first six and five days of mechanical ventilation, respectively (all P < 0.05). CONCLUSION Dexmedetomidine demonstrated stronger protective effects on the brain compared to propofol for long-term mechanical ventilation in patients without brain injury.
Collapse
Affiliation(s)
- Hong-Xun Yuan
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Li-Na Zhang
- Central Operating Room, The Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing 100020, China
| | - Gang Li
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Li Qiao
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| |
Collapse
|
7
|
Zhu Y, He YJ, Yu Y, Xu D, Yuan SY, Yan H. Aldehyde Dehydrogenase 2 Preserves Mitochondrial Function in the Ischemic Heart: A Redox-dependent Mechanism for AMPK Activation by Thioredoxin-1. J Cardiovasc Pharmacol 2024; 83:93-104. [PMID: 37816196 DOI: 10.1097/fjc.0000000000001499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023]
Abstract
ABSTRACT Aldehyde dehydrogenase 2 (ALDH2) protects the ischemic heart by activating adenosine 5'-monophosphate-activated protein kinase (AMPK) signaling. However, the molecular mechanisms linking ALDH2 and AMPK signaling are not fully understood. This study aimed to explore the potential mechanisms linking ALDH2 and AMPK in myocardial ischemic injury. An ischemic model was established by ligating the left anterior descending coronary artery in rats. The overexpression or knockdown of ALDH2 in H9c2 cells treated with oxygen-glucose deprivation was obtained through lentivirus infection. Transferase-mediated dUTP nick-end labeling was used to evaluate apoptosis in an ischemic rat model and oxygen-glucose deprivation cells. ALDH2 activity, mitochondrial oxidative stress markers, adenosine triphosphate, respiratory control ratio, and cell viability in H9c2 cells were evaluated using a biological kit and 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide. Protein expression of ALDH2 , 4-hydroxynonenal, thioredoxin-1 (Trx-1), and AMPK-proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) signaling pathway was detected through Western blotting. ALDH2 activation reduced ischemic-induced myocardial infarct size and apoptosis. ALDH2 protected mitochondrial function by enhancing mitochondrial respiratory control ratio and adenosine triphosphate production, alleviated mitochondrial oxidative stress, and suppressed myocardial apoptosis. Moreover, ALDH2 attenuated ischemia-induced oxidative stress and maintained Trx-1 levels by reducing 4-hydroxynonenal, thereby promoting AMPK-PGC-1α signaling activation. Inhibiting Trx-1 or AMPK abolished the cardioprotective effect of ALDH2 on ischemia. ALDH2 alleviates myocardial injury through increased mitochondrial biogenesis and reduced oxidative stress, and these effects were achieved through Trx1-mediating AMPK-PGC1-α signaling activation.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Intensive Care Unit, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China; and
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Ying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Yan
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Abudurousuli G, Xu S, Che J, Ding X, Gui B, Zhu L. Role of ferroptosis in effects of anesthetics on multiple organ diseases: A literature review. Heliyon 2023; 9:e20405. [PMID: 37780755 PMCID: PMC10539942 DOI: 10.1016/j.heliyon.2023.e20405] [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: 05/17/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
Anesthesiologists are often faced with patients combined with a series of organ injuries, such as acute lung injury, myocardial ischemia-reperfusion injury, and neurodegenerative diseases. With the in-depth study of these diseases, we are more aware of the choice and rational use of anesthetics for the prognosis of these patients. Ferroptosis is a new type of programmed cell death. This unique pattern of cell death, driven by an imbalance between oxides and antioxidants, is regulated by multiple cellular metabolic events, including redox homeostasis, iron handling, mitochondrial activity, and lipids peroxidation. Numerous studies confirmed that anesthetics modulate ferroptosis by interfering its machineries such as cystine-import-glutathione-glutathione peroxidase 4 axis, Heme oxygenase 1, nuclear factor erythroid 2-related factor 2, and iron homeostasis system. In this literature review, we systemically illustrated possible involvement of ferroptosis in effects of anesthetics and adjuvant drugs on multiple organ diseases, hoping our work may serve as a basis for further studies on regulating ferroptosis through anesthetics related pharmacological modulation and promoting the rational use of anesthetics.
Collapse
Affiliation(s)
- Gulibositan Abudurousuli
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Siyang Xu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jiangsu Province Official Hospital, Nanjing, China
| | - Jinxing Che
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, The Huai'an Maternity and Child Healthcare Hospital, Huai'an, China
| | - Xiahao Ding
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Bo Gui
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Linjia Zhu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Chen Y, Chen H, Chen Y, Yang Z, Zhou T, Xu W. Dexmedetomidine Preconditioning Attenuates Myocardial Ischemia/Reperfusion Injury in Rats by Suppressing Mitophagy Via Activating Α2-Adrenergic Receptor. Arq Bras Cardiol 2023; 120:e20220750. [PMID: 37909577 DOI: 10.36660/abc.20220750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 08/16/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Dexmedetomidine (DEX), a specific α2-adrenergic receptor agonist, is protective against myocardial ischemia/reperfusion injury (MIRI). However, the association between DEX preconditioning-induced cardioprotection and mitophagy suppression remains unclear. OBJECTIVE Hence, we aimed to investigate whether DEX preconditioning alleviates MIRI by suppressing mitophagy via α2-adrenergic receptor activation. METHOD Sixty isolated rat hearts were treated with or without DEX before inducing ischemia and reperfusion; an α2-adrenergic receptor antagonist, yohimbine (YOH), was also administered before ischemia, alone or with DEX. The heart rate (HR), left ventricular diastolic pressure (LVDP), left ventricular end-diastolic pressure (LVEDP), maximal and minimal rate of left ventricular pressure development (±dp/dtmax), and myocardial infarction size were measured. The mitochondrial ultrastructure and autophagosomes were assessed using transmission electron microscopy. Mitochondrial membrane potential and reactive oxygen species (ROS) levels were measured using JC-1 and dichloride hydrofluorescein diacetate assays, respectively. The expression levels of the mitophagy-associated proteins Beclin1, LC3II/I ratio, p62, PINK1, and Parkin were detected by western blotting. RESULTS Compared with the control group, in the ischemia/reperfusion group, the HR, LVDP, and ±dp/dtmax were remarkably decreased (p< 0.05), whereas LVEDP and infarct sizes were significantly increased (p< 0.05). DEX preconditioning significantly improved cardiac dysfunction reduced myocardial infarction size, maintained mitochondrial structural integrity, increased mitochondrial membrane potential, inhibited autophagosomes formation, and decreased ROS production and Beclin1, LC3II/I ratio, PINK1, Parkin, and p62 expression(p< 0.05). When DEX and YOH were combined, YOH canceled the effect of DEX, whereas the use of YOH alone had no effect. CONCLUSION Therefore, DEX preconditioning was cardioprotective against MIRI in rats by suppressing mitophagy via α2-adrenergic receptor activation.
Collapse
Affiliation(s)
- YaHua Chen
- Guizhou Aerospace Hospital, Zunyi, Guizhou - China
- Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou - China
| | - Hui Chen
- Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou - China
| | - YuJiao Chen
- Affiliated Hospital of North Sichuan Medical College, NanChong, Sichuan - China
| | - ZaiQun Yang
- People's Hospital of Qiandongnan Miao and Dong Autonomous Prefecture, Qiandongnan, Guizhou - China
| | - Tao Zhou
- Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou - China
| | - Wei Xu
- Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou - China
| |
Collapse
|
10
|
Zhang S, Tang J, Sun C, Zhang N, Ning X, Li X, Wang J. Dexmedetomidine attenuates hepatic ischemia-reperfusion injury-induced apoptosis via reducing oxidative stress and endoplasmic reticulum stress. Int Immunopharmacol 2023; 117:109959. [PMID: 36881980 DOI: 10.1016/j.intimp.2023.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Dexmedetomidine (DEX) affords a hepatoprotective effect during ischemia-reperfusion (IR) injury (IRI); however, the underlying mechanism remains elusive. In this work, using a rat liver IR model and a BRL-3A cell hypoxia-reoxygenation (HR) model, we explored whether DEX protects the liver against IRI by decreasing oxidative stress (OS), endoplasmic reticulum stress (ERS), and apoptotic pathways. We found that DEX significantly increased SOD and GSH activity while decreasing ROS and MDA levels in BRL-3A cells, successfully preventing HR-induced OS damage. DEX administration reduced JNK, ERK, and P38 phosphorylation and blocked HR-induced MAPK signaling pathway activation. Additionally, DEX administration reduced the expression of GRP78, IRE1α, XBP1, TRAF2, and CHOP, which reduced HR-induced ERS. NAC prevented the MAPK pathway from being activated and inhibited the ERS pathway. Further research showed that DEX significantly reduced HR-induced apoptosis by suppressing the expression of Bax/Bcl-2 and cleaved caspase-3. Similarly, animal studies demonstrated DEX exerted a protective effect of the liver by alleviating histopathological injury and enhancing liver function, mechanically DEX reduced cell apoptosis in liver tissue by reducing oxidative stress and ERS. In conclusion, DEX mitigates OS and ERS during IR, thereby suppressing cell apoptosis, thus providing protection to the liver.
Collapse
Affiliation(s)
- Shixia Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jilang Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| | - Chen Sun
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Nuannuan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xiaqing Ning
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xueqin Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jiaqi Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| |
Collapse
|
11
|
Sun M, Wang R, Xia R, Xia Z, Wu Z, Wang T. Amelioration of myocardial ischemia/reperfusion injury in diabetes: A narrative review of the mechanisms and clinical applications of dexmedetomidine. Front Pharmacol 2022; 13:949754. [PMID: 36120296 PMCID: PMC9470922 DOI: 10.3389/fphar.2022.949754] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanisms contributing to the pathogenesis of myocardial ischemia-reperfusion (I/R) injury are complex and multifactorial. Many strategies have been developed to ameliorate myocardial I/R injuries based on these mechanisms. However, the cardioprotective effects of these strategies appear to diminish in diabetic states. Diabetes weakens myocardial responses to therapies by disrupting intracellular signaling pathways which may be responsible for enhancing cellular resistance to damage. Intriguingly, it was found that Dexmedetomidine (DEX), a potent and selective α2-adrenergic agonist, appears to have the property to reverse diabetes-related inhibition of most intervention-mediated myocardial protection and exert a protective effect. Several mechanisms were revealed to be involved in DEX’s protection in diabetic rodent myocardial I/R models, including PI3K/Akt and associated GSK-3β pathway stimulation, endoplasmic reticulum stress (ERS) alleviation, and apoptosis inhibition. In addition, DEX could attenuate diabetic myocardial I/R injury by up-regulating autophagy, reducing ROS production, and inhibiting the inflammatory response through HMGB1 pathways. The regulation of autonomic nervous function also appeared to be involved in the protective mechanisms of DEX. In the present review, the evidence and underlying mechanisms of DEX in ameliorating myocardial I/R injury in diabetes are summarized, and the potential of DEX for the treatment/prevention of myocardial I/R injury in diabetic patients is discussed.
Collapse
Affiliation(s)
- Meng Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhilin Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhilin Wu, ; Tingting Wang,
| | - Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhilin Wu, ; Tingting Wang,
| |
Collapse
|
12
|
Role of Keap1-Nrf2/ARE signal transduction pathway in protection of dexmedetomidine preconditioning against myocardial ischemia/reperfusion injury. Biosci Rep 2022; 42:231655. [PMID: 35959640 PMCID: PMC9446386 DOI: 10.1042/bsr20221306] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Objective: To explore the role and mechanism of the Kelch sample related protein-1-nuclear factor erythroid-2 related factor 2/antioxidant response element (Keap1-Nrf2/ARE) signaling pathway in protection of dexmedetomidine (DEX) preconditioning against myocardial ischemia/reperfusion injury (MIRI). Methods: A total of 70 male SD rats were randomly divided into seven equal groups (n=10): blank control (S group), ischemia/reperfusion injury (C group), DEX preconditioning (DEX group), tertiary butylhydroquinone (tBHQ) control (tBHQ group), combined tBHQ and DEX preconditioning (tBHQ+DEX group), all-trans retinoic acid (ATRA) control (ATRA group), and combined ATRA and DEX preconditioning (ATRA+DEX group). Serum creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) concentrations were measured by ELISA kits, and the infarct size (IS) was assessed by Evan’s blue and 2,3,5-triphenyltetrazolium chloride (TTC) staining. Oxidative stress was assessed through Western blotting for expression of Keap1-Nrf2/ARE pathway members and oxidative stress markers. Results: Cardioprotection of DEX, tBHQ, and tBHQ+DEX preconditioning treatments were shown as lower concentrations of serum CK-MB and cTnI and a smaller IS following MIRI in rats compared with those of MIRI rats without pre-treatment. In addition, tBHQ+DEX preconditioning exhibited stronger myocardial protection compared with DEX preconditioning. Mechanistically, the cardioprotection offered by DEX, tBHQ, and tBHQ+DEX preconditioning treatments was mediated via exerting antioxidant stress through activation of the Keap1-Nrf2/ARE signal transduction pathway. Conversely, the protective effects of DEX were diminished by blocking the Keap1-Nrf2/ARE pathway with inhibitor ATRA. Conclusion: DEX preconditioning protects against MIRI by exerting antioxidant stress through activation of the Keap1-Nrf2/ARE signal transduction pathway, while inhibition of the Keap1-Nrf2/ARE signal transduction pathway reverses the protective effect of DEX preconditioning on MIRI.
Collapse
|
13
|
Wang H, Yao X, Huang K, Zhang J, Xiao J, Guo J, Wei D, Xiang B. Low-dose dexamethasone in combination with luteolin improves myocardial infarction recovery by activating the antioxidative response. Biomed Pharmacother 2022; 151:113121. [PMID: 35605295 DOI: 10.1016/j.biopha.2022.113121] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
This study aimed to explore the effects of dexamethasone (DEX) and its combination with luteolin (LUT) on cardiac function during myocardial infarction (MI) in a mouse model. We evaluated whether the Keap1/Nrf2 pathway mediates the cardioprotective function of DEX both in vivo and in vitro. The MI mouse model was established by ligation of the left anterior descending coronary artery of wild-type (WT) and Nrf2 knockout mice. After recovery for 21 days, DEX or its combination with LUT was intraperitoneally administered at different doses to WT or Nrf2 knockout mice daily for 7 consecutive days. Mice treated with DEX at a low dose (50 μg/kg/day) showed better cardiac function, fewer cardiac lesions, and smaller infarct sizes compared with MI model mice. DEX (50 μg/kg/day) administration also significantly decreased the production of reactive oxygen species (ROS) and pro-inflammatory cytokines, increased the expression of antioxidative enzymes, and activated the Keap1/Nrf2/HO-1 pathway. However, in Nrf2 knockout mice, DEX treatment did not influence cardiac function, inflammation, the oxidative response, or Keap1/Nrf2/HO-1 activation. In the MI cell model, low concentrations of DEX attenuated the H2O2-induced decreases in cell viability and antioxidative enzyme levels and activated the Keap1/Nrf2/HO-1 pathway. Low doses of DEX exerted protective effects in MIR mice and MI cell models by improving cardiac function, eliminating ROS, inhibiting inflammatory responses, and activating antioxidative responses. The protective effects of DEX on myocardial tissues were mediated by the Keap1/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Xiaoqin Yao
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Keli Huang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jing Zhang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jingrong Xiao
- Nursing Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jing Guo
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Dachuang Wei
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Bo Xiang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
14
|
Peng JF, Salami OM, Lei C, Ni D, Habimana O, Yi GH. Targeted mitochondrial drugs for treatment of Myocardial ischemia-reperfusion injury. J Drug Target 2022; 30:833-844. [PMID: 35652502 DOI: 10.1080/1061186x.2022.2085728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) refers to the further damage done to ischemic cardiomyocytes when restoring blood flow. A large body of evidence shows that MI/RI is closely associated with excessive production of mitochondrial reactive oxygen species, mitochondrial calcium overload, disordered mitochondrial energy metabolism, mitophagy, mitochondrial fission, and mitochondrial fusion. According to the way it affects mitochondria, it can be divided into mitochondrial quality abnormalities and mitochondrial quantity abnormalities. Abnormal mitochondrial quality refers to the dysfunction caused by the severe destruction of mitochondria, which then affects the balance of mitochondrial density and number, causing an abnormal mitochondrial quantity. In the past, most of the reports were limited to the study of the mechanism of myocardial ischemia-reperfusion injury, some of which involved mitochondria, but no specific countermeasures were proposed. In this review, we outline the mechanisms for treating myocardial ischemia-reperfusion injury from the direction of mitochondria and focus on targeted interventions and drugs to restore mitochondrial health during abnormal mitochondrial quality control and abnormal mitochondrial quantity control. This is an update in the field of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jin-Fu Peng
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | | | - Cai Lei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Ni
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Olive Habimana
- International College, University of South China, 28 W Changsheng Road, Hengyang, Hunan 421001, China
| | - Guang-Hui Yi
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
15
|
Wang L, Wang S, Jia T, Sun X, Xing Z, Liu H, Yao J, Chen Y. Dexmedetomidine prevents cardiomyocytes from hypoxia/reoxygenation injury via modulating tetmethylcytosine dioxygenase 1-mediated DNA demethylation of Sirtuin1. Bioengineered 2022; 13:9369-9386. [PMID: 35387565 PMCID: PMC9161963 DOI: 10.1080/21655979.2022.2054762] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Myocardial hypoxia/reoxygenation (H/R) injury is a common pathological change in patients with acute myocardial infarction undergoing reperfusion therapy. Dexmedetomidine (DEX) has been found to substantially improve ischemia-mediated cell damage. Here, we focus on probing the role and mechanism of DEX in ameliorating myocardial H/R injury. Oxygen–glucose deprivation and reoxygenation (OGD/R) were applied to construct the H/R injury model in human myocardial cell lines. After different concentrations of DEX’s treatment, cell counting kit-8 (CCK-8) assay and BrdU assay were employed to test cell viability. The profiles of apoptosis-related proteins Bcl2, Bax, Bad and Caspase3, 8, 9 were determined by Western blot (WB). The expression of inflammatory factors interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) was checked by reverse transcription-polymerase chain reaction (RT-PCR). By conducting WB, we examined the expression of NF-κB, Sirt1, Tet methylcytosine dioxygenase 1 (TET1) and DNA methylation-related proteins (DNA methyltransferase 1, DNMT1; DNA methyltransferase 3 alpha, DNMT3A; and DNA methyltransferase 3 beta, DNMT3B). Our data showed that OGD/R stimulation distinctly hampered the viability and elevated apoptosis and inflammatory factor expression in cardiomyocytes. DEX treatment notably impeded myocardial apoptosis and inflammation and enhanced cardiomyocyte viability. OGD/R enhanced total DNA methylation levels in cardiomyocytes, while DEX curbed DNA methylation. In terms of mechanism, inhibiting TET1 or Sirtuin1 (Sirt1) curbed the DEX-mediated myocardial protection. TET1 strengthened demethylation of the Sirt1 promoter and up-regulated Sirt1. DEX up-regulates Sirt1 by accelerating TET1 and mediating demethylation of the Sirt1 promoter and improves H/R-mediated myocardial injury.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Shaowei Wang
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Tong Jia
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Xiaojia Sun
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Zhen Xing
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Hui Liu
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Jie Yao
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Yanlin Chen
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| |
Collapse
|
16
|
Yu P, Zhang J, Ding Y, Chen D, Sun H, Yuan F, Li S, Li X, Yang P, Fu L, Yu S, Zhang J. Dexmedetomidine post-conditioning alleviates myocardial ischemia-reperfusion injury in rats by ferroptosis inhibition via SLC7A11/GPX4 axis activation. Hum Cell 2022; 35:836-848. [PMID: 35212945 DOI: 10.1007/s13577-022-00682-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/30/2022] [Indexed: 02/08/2023]
Abstract
The SLC7A11/GPX4 axis plays an important role in ferroptosis during cardiac ischemia/reperfusion injury (IRI). The present study was designed to evaluate the impact of dexmedetomidine (DEX) post-conditioning on cardiac IRI and to explore whether the effect was achieved by SLC7A11/GPX4 signaling pathway regulation. Rat myocardial IRI was established by occluding the left anterior descending artery for 30 min followed by 2-h reperfusion. The infarct area was detected by diphenyltetrazolium chloride (TTC) staining; the cardiac function was evaluated by echocardiography. The levels of lipid peroxide biomarkers were measured to estimate the injury caused by lipid peroxide. HE staining and Sirius staining were utilized to assess myocardial damage and fibrosis. The mitochondrial morphology was observed by electron micrography. Western blot and quantitative real-time polymerase chain reaction were employed to measure the relative molecular characteristics. Our results showed that DEX administration at the beginning of reperfusion attenuated IRI-induced myocardial injury, alleviated mitochondrial dysfunction, decreased the level of reactive oxygen species (ROS), alleviated mitochondrial dysfunction, inhibited the activation of SLC7A11/GPX4, and modulated the expression of ferroptosis-related proteins, including SLC7A11, glutathione peroxidase 4 (GPX4), ferritin heavy chain (FTH), and cyclooxygenase-2 (COX-2). Conversely, the ferroptosis activator erastin partly suppressed the DEX-mediated cardio protection. Altogether, these results reveal that DEX inhibits ferroptosis by enhancing the expression of SLC7A11 and GPX4, thereby preventing cardiac I/R injury.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yi Ding
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, China
| | - Dandan Chen
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, China
| | - Haijian Sun
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Fenglai Yuan
- Department of Burns and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214125, China
| | - Siyuan Li
- Grade 2017, The Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Xiaozhong Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Linghua Fu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shuchun Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jiru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, China.
| |
Collapse
|
17
|
Wang H, Xia Y, Sun Z. Dexmedetomidine: A potential therapy for myocardial ischemia/reperfusion injury. Int J Cardiol 2021; 335:104. [PMID: 33930513 DOI: 10.1016/j.ijcard.2021.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Hongkun Wang
- Department of Cardiology, The Second People's Hospital of Liaocheng, Liaocheng 252000, PR China
| | - Ying Xia
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, PR China
| | - Zhiqiang Sun
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, PR China.
| |
Collapse
|
18
|
Xiao J, Wan W, Zhang Y, Ma J, Yan L, Luo Y, Tang J. Administration of Dexmedetomidine Does Not Produce Long-Term Protective Effect on Testicular Damage Post Testicular Ischemia-Reperfusion Injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:315-321. [PMID: 33536744 PMCID: PMC7850429 DOI: 10.2147/dddt.s293926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Background After surgical correction of testicular torsion, up to 68% of ipsilateral testes undergo atrophy due to ischemia-reperfusion injury (IRI). Recent studies have shown that dexmedetomidine (Dex) alleviates IRI in various vital organs. However, those studies evaluated its protective effect on short-term reperfusion. Purpose We aimed to investigate whether Dex has a long-term protective effect against testicular injury after IRI. Materials and Methods A total of 24 New Zealand white rabbits were randomly divided into three groups (n = 8/group): the control group (saline-infused rabbits without IRI), the IRI group (saline-injected rabbits with IRI), and the Dex group (Dex-injected rabbits with IRI). The spermatic cord of rabbits in IRI and Dex groups was ligated for 4 h, and 1 h before reperfusion, Dex was administered intraperitoneally at a dose of 50 μg/kg body weight in group Dex, whereas saline was administered at the same dose to the IRI and control groups. Rabbits were kept alive for 4 weeks post reperfusion, then the testes were harvested, and the rabbits were euthanized. Results Four weeks post reperfusion, testicular volumes of the affected side decreased considerably in the IRI and Dex groups compared to the control group, with no significant difference between the IRI and Dex groups. Compared to the control group, the Johnson score and the mean seminiferous tubular diameters were significantly decreased in the IRI and Dex groups, but no significant differences were observed after administration of Dex. There were no significant differences in malondialdehyde and superoxide dismutase levels between the groups treated with and without Dex. Conclusion Dex administration 3 h after ischemia and 1 h before reperfusion did not demonstrate a significant protective effect against testicular injury 4 weeks after IRI in rabbits. Further research is needed to confirm the potential therapeutic effects of Dex by varying the experimental conditions.
Collapse
Affiliation(s)
- Jing Xiao
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Wenbo Wan
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ying Zhang
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jun Ma
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lin Yan
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yukun Luo
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jie Tang
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|