1
|
Zhang Y, Lu Q, Hu H, Yang C, Zhao Q. Esketamine alleviates hypoxia/reoxygenation injury of cardiomyocytes by regulating TRPV1 expression and inhibiting intracellular Ca 2+ concentration. Clinics (Sao Paulo) 2024; 79:100363. [PMID: 38692008 PMCID: PMC11070684 DOI: 10.1016/j.clinsp.2024.100363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/07/2024] [Indexed: 05/03/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the effect of Esketamine (ESK) on the Hypoxia/Reoxygenation (H/R) injury of cardiomyocytes by regulating TRPV1 and inhibiting the concentration of intracellular Ca2+. METHODS The H/R injury model of H9c2 cardiomyocytes was established after 4h hypoxia and 6h reoxygenation. H9c2 cells were treated with different concentrations of ESK or TRPV1 agonist capsaicin (10 μM) or TRPV1 inhibitor capsazepine (1 μM). Cell viability was detected by CCK-8 method, and apoptosis by flow cytometry. Intracellular Ca2+ concentration was evaluated by Fluo-4 AM. LDH, MDA, SOD, and GSH-Px were detected with corresponding commercial kits. TRPV1 and p-TRPV1 proteins were detected by Western blot. RESULTS After H/R, H9c2 cell viability decreased, apoptosis increased, intracellular Ca2+ concentration increased, LDH and MDA levels increased, SOD and GSH-Px levels decreased, and p-TRPV1 expression increased. ESK treatment rescued these changes induced by H/R. After up-regulating TRPV1, the protective effect of ESK on H/R injury of H9c2 cells was weakened, while down-regulating TRPV1 could further protect against H/R injury. CONCLUSION ESK alleviates H/R injury of cardiomyocytes by regulating TRPV1 expression and inhibiting intracellular Ca2+ concentration.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - QuanMei Lu
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - HanChun Hu
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - ChunChen Yang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - QiHong Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China.
| |
Collapse
|
2
|
Yuan W, Xiao Y, Zhang Y, Xiang K, Huang T, Diaby M, Gao J. Apoptotic mechanism of development inhibition in zebrafish induced by esketamine. Toxicol Appl Pharmacol 2024; 482:116789. [PMID: 38103741 DOI: 10.1016/j.taap.2023.116789] [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: 09/30/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Esketamine, a widely used intravenous general anesthetic, is also employed for obstetric and pediatric anesthesia, and depression treatment. However, concerns regarding esketamine abuse have emerged. Moreover, the potential in vivo toxicity of esketamine on growth and development remains unclear. To address these concerns, we investigated the effects of esketamine exposure on developmental parameters, cell apoptosis, and gene expression in zebrafish. Esketamine exposure concentration-dependently decreased the heart rate and body length of zebrafish embryos/larvae while increasing the hatching rate and spontaneous movement frequency. Developmental retardation of zebrafish larvae, including shallow pigmentation, small eyes, and delayed yolk sac absorption, was also observed following esketamine treatment. Esketamine exposure altered the expression of apoptosis-related genes in zebrafish heads, primarily downregulating bax, caspase9, caspase3, caspase6, and caspase7. Intriguingly, BTSA1, a Bax agonist, reversed the anti-apoptotic and decelerated body growth effects of esketamine in zebrafish. Collectively, our findings suggest that esketamine may hinder embryonic development by inhibiting embryonic apoptosis via the Bax/Caspase9/Caspase3 pathway. To the best of our knowledge, this is the first study to report the lethal toxicity of esketamine in zebrafish. We have elucidated the developmental toxic effects of esketamine on zebrafish larvae and its potential apoptotic mechanisms. Further studies are warranted to evaluate the safety of esketamine in animals and humans.
Collapse
Affiliation(s)
- Wenjuan Yuan
- Medical College of Yangzhou University, Yangzhou, China; Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
| | - Yinggang Xiao
- Medical College of Yangzhou University, Yangzhou, China; Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
| | - Yang Zhang
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
| | - Kuilin Xiang
- College of Animal Science and Technology, Yangzhou University, Jiangsu, China
| | - Tianfeng Huang
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
| | - Mohamed Diaby
- College of Animal Science and Technology, Yangzhou University, Jiangsu, China
| | - Ju Gao
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China.
| |
Collapse
|
3
|
Wang Y, Cheng W, Wang X, He T, Liu J, Chen S, Zhang J. Integrated metabolomics and network pharmacology revealing the mechanism of arsenic-induced hepatotoxicity in mice. Food Chem Toxicol 2023:113913. [PMID: 37348806 DOI: 10.1016/j.fct.2023.113913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/20/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Endemic arsenic (As) poisoning is a severe biogeochemical disease that endangers human health. Epidemiological investigations and animal experiments have confirmed the damaging effects of As on the liver, but there is an urgent need to investigate the underlying mechanisms. This study adopted a metabolomic approach using UHPLC-QE/MS to identify the different metabolites and metabolic mechanisms associated with As-induced hepatotoxicity in mice. A network pharmacology approach was applied to predict the potential target of As-induced hepatotoxicity. The predicted targets of differential metabolites were subjected to a deep matching for elucidating the integration mechanisms. The results demonstrate that the levels of ALT and AST in plasma significantly increased in mice after As exposure. In addition, the liver tissue showed disorganized liver lobules, lax cytoplasm and inflammatory cell infiltration. Metabolomic analysis revealed that As exposure caused disturbance to 40 and 75 potential differential metabolites in plasma and liver, respectively. Further investigation led to discovering five vital metabolic pathways, including phenylalanine, tyrosine, and tryptophan biosynthesis and nicotinate and nicotinamide metabolism pathways. These pathways may responded to As-induced hepatotoxicity primarily through lipid metabolism, apoptosis, and deoxyribonucleic acid damage. The network pharmacology suggested that As could induce hepatotoxicity in mice by acting on targets including Hsp90aa1, Akt2, Egfr, and Tnf, which regulate PI3K Akt, HIF-1, MAPK, and TNF signaling pathways. Finally, the integrated metabolomics and network pharmacology revealed eight key targets associated with As-induced hepatoxicity, namely DNMT1, MAOB, PARP1, MAOA, EPHX2, ANPEP, XDH, and ADA. The results also suggest that nicotinic acid and nicotinamide metabolisms may be involved in As-induced hepatotoxicity. This research identified the metabolites, targets, and mechanisms of As-induced hepatotoxicity, offering meaningful insights and establishing the groundwork for developing antidotes for widespread As poisoning.
Collapse
Affiliation(s)
- Yazhi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Weina Cheng
- Department of Pharmaceutical Analysis, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaoning Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Tianmu He
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, China; School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Jingxian Liu
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, China
| | - Shuangshuang Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Jianyong Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
4
|
Tao Z, Lin R, Zhang R, He P, Lei C, Li Y. Ischemia reperfusion myocardium injuries in type 2 diabetic rats: Effects of ketamine and insulin on LC3-II and mTOR expression. Int J Immunopathol Pharmacol 2023; 37:3946320231196450. [PMID: 37643354 PMCID: PMC10467302 DOI: 10.1177/03946320231196450] [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: 05/10/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023] Open
Abstract
Objectives: Myocardiopathy occurs in ischemia-induced injury caused by dysregulation of autophagy of cardiac tissues. The present report evaluates the protective effect of ketamine and insulin against myocardial injury in type 2 diabetic rats (T2DM).Methods: The effects of ketamine and its combination with insulin on biochemical parameters and inflammatory cytokines in the serum of I/R-induced myocardial injury in T2DM rats were evaluated. The parameters of reactive oxygen species and the expression of autophagosome signaling pathway proteins were also determined. Using transmission electron microscopy, we investigated autophagosomes. Western blots were used to detect autophagy-associated signaling pathways. Myocardial function was determined by echocardiography and histopathological changes in myocardial tissues were also determined in I/R-induced myocardial injury in type 2 diabetic rats.Results: There was a significant reduction in glucose, AST, LDH, and CK-MB levels and cytokines (IL-1β, IL-6, and TNF-α) in serum of the ketamine (p < .05) and ketamine + insulin (p < .01) groups than in the diabetic + I/R. MDA and ROS levels were reduced with a substantial (p < .05) increase in GSH levels through improved cardiac function in the ketamine (p < .05) and ketamine + insulin (p < .01) groups than the diabetic + I/R group. There was an increase in mature autophagosomes in diabetic+I/R+Kt+In compared to diabetic+I/R+Kt alone in infarction and marginal zones. It should be noted that the significant increase (p < .01) in protein levels of the autophagy-associated intracellular signaling pathways AMPK and mTOR, as well as an increase in LC3-II and BECLIN-1, suggests that ketamine combined with insulin-activated autophagy-associated intracellular signaling AMPK and mTOR.Conclusion: The findings of the study suggest that ketamine combined with insulin administration remarkably protects I/R-induced myocardial injury in rats with T2DM by reducing the dysregulation of autophagy.
Collapse
Affiliation(s)
- Zhiguo Tao
- Department of Anaesthesiology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Rongmu Lin
- Department of Anaesthesiology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Rui Zhang
- Department of Anaesthesiology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Peng He
- Department of Anaesthesiology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Chengwen Lei
- Department of Anaesthesiology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yuanhai Li
- Department of Anaesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Xiao S, Zhou Y, Wang Q, Yang D. Ketamine Attenuates Airway Inflammation via Inducing Inflammatory Cells Apoptosis and Activating Nrf2 Pathway in a Mixed-Granulocytic Murine Asthma Model. Drug Des Devel Ther 2022; 16:4411-4428. [PMID: 36597444 PMCID: PMC9805722 DOI: 10.2147/dddt.s391010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose The use of ketamine, an anesthetic, as a treatment for asthma has been investigated in numerous studies. However, how ketamine affects asthma is unclear. The present study examined the effects of ketamine on a murine model of mixed-granulocytic asthma, and the role of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. Methods The murine model of mixed-granulocytic asthma was established using ovalbumin (OVA) for sensitization and the combination of OVA and lipopolysaccharides (LPS) for challenge. The main characteristics of asthma, oxidative stress biomarkers, and the expression of the Nrf2 pathway were examined. ML385 was administered to verify the role of the Nrf2 pathway. Results Mice in the OVA +LPS group developed asthmatic characteristics, including airway hyperresponsiveness, mixed-granulocytic airway inflammation, mucus overproduction, as well as increased levels of oxidative stress and impaired apoptosis of inflammatory cells. Among the three concentrations, ketamine at 75mg/kg effectively attenuated these asthmatic symptoms, activated the Nrf2 pathway, decreased oxidative stress, and induced apoptosis of eosinophils and neutrophils in bronchoalveolar lavage fluid (BALF) with a reducing level of myeloid cell leukemia 1(Mcl-1). ML385 (an Nrf2 inhibitor) eliminated the protective effects of ketamine on the mixed-granulocytic asthma model. Conclusion The study concluded that ketamine reduced oxidative stress and attenuated asthmatic symptoms (neutrophilic airway inflammation) by activating the Nrf2-Keap1 pathway, with 75 mg/kg ketamine showing the best results. Ketamine administration also increased neutrophil and eosinophil apoptosis in BALF, which may contribute to the resolution of inflammation. The use of ketamine as a treatment for asthma may therefore be beneficial.
Collapse
Affiliation(s)
- Shilin Xiao
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ying Zhou
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Qianyu Wang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Dong Yang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China,Correspondence: Dong Yang, Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan, Beijing, 100144, People’s Republic of China, Tel +86-13661267522, Email
| |
Collapse
|
6
|
Xu G, Wang Y, Chen Z, Zhang Y, Zhang X, Zhang G. Esketamine improves propofol-induced brain injury and cognitive impairment in rats. Transl Neurosci 2022; 13:430-439. [PMID: 36561289 PMCID: PMC9730546 DOI: 10.1515/tnsci-2022-0251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 12/13/2022] Open
Abstract
As an intravenous anesthetic, propofol has been indicated to induce neurotoxicity in both animal and human brains. It is of great significance to better understand the potential mechanism of propofol-induced neurotoxicity to eliminate the side effects of propofol. Esketamine is a sedative that has been proven to have an antidepressant effect. However, its effect on propofol-induced neurotoxicity and the underlying mechanism remain unclear. Herein, we investigated the role of esketamine in propofol-induced brain injury. A rat model of propofol-induced brain injury was established with or without the treatment of esketamine. The results demonstrated that propofol-induced impairment in spatial learning and memory of rats and promoted oxidative stress, neuronal injury and apoptosis in rat hippocampal tissues. The effects caused by propofol were attenuated by esketamine. Esketamine activated the mature brain-derived neurotrophic factor/tropomyosin receptor kinase B/phosphatidylinositide 3-kinase (mBDNF/TrkB/PI3K) signaling pathway in propofol-administrated rats. Moreover, knocking down BDNF partially reversed esketamine-mediated activation of the mBDNF/TrkB/PI3K signaling pathway and inhibition of neuronal apoptosis in propofol-induced rats. Overall, esketamine mitigates propofol-induced cognitive dysfunction and brain injury in rats by activating mBDNF/TrkB/PI3K signaling.
Collapse
Affiliation(s)
- Guiping Xu
- Department of Anesthesiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Urumqi 830001, China
| | - Yang Wang
- Department of Anesthesiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Urumqi 830001, China
| | - Zhe Chen
- Department of Anesthesiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Urumqi 830001, China
| | - Yuxuan Zhang
- Department of Anesthesiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Urumqi 830001, China
| | - Xuexue Zhang
- Graduate School of Xinjiang Medical University, Urumqi 830000, China
| | - Guichao Zhang
- Medical School, Shihezi University, Xinjiang, Shihezi, 832000, China
| |
Collapse
|
7
|
Han J, Zhang X, Xia L, Liao O, Li Q. S-ketamine promotes autophagy and alleviates neuropathic pain by inhibiting PI3K/Akt/mTOR signaling pathway. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Xu G, Lv X, Feng Y, Li H, Chen C, Lin H, Li H, Wang C, Chen J, Sun J. Study on the effect of active components of Schisandra chinensis on liver injury and its mechanisms in mice based on network pharmacology. Eur J Pharmacol 2021; 910:174442. [PMID: 34492285 DOI: 10.1016/j.ejphar.2021.174442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
The aim of this study was to analyze the active components of Schisandra chinensis on liver injury and its mechanism in mice by network pharmacology. The active components of S. chinensis were found through Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and their corresponding targets were predicted. The targets of liver injury were searched through Therapeutic Targets Database (TTD), DisGeNET and drugbank databases, and the Venn diagram was constructed to obtain the action targets. The "drug-active component-target" network and protein-protein interaction network (PPI) were constructed by using STRING database and Cytoscape software, and the key targets were further screened by the enrichment analysis of relevant KEGG pathways. Finally, a CCl4-induced mouse liver injury model was established to verify the efficacy and related targets of S. chinensis and clarify its mechanism. Eight active components and 56 related targets of S. chinensis were screened out based on their oral bioavailability (OB) and drug likeness (DL). Five targets of S. chinensis related to liver injury were found by using the Venn diagram. The key targets, namely Ptgs2 and Nos2 genes, were further screened out by constructing a PPI network, and Schisandrol B (SCB) was considered the key component most closely related to the liver injury in S. chinensis. The results indicate that SCB may play a role in the treatment of the CCl4-induced liver injury by down-regulating the expression of iNOS and COX-2, and regulating the expression of NF-κB and IL-17 signaling pathway to inhibit the expression of proinflammatory factors.
Collapse
Affiliation(s)
- Guangyu Xu
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Xi Lv
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Yanbo Feng
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Han Li
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Cong Chen
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Hao Lin
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - He Li
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Chunmei Wang
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Jianguang Chen
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Jinghui Sun
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China.
| |
Collapse
|