1
|
Xi Y, Hou X, Huang Y, Zhou Y, Chen Y, Wang Y, Cheng H. Loganin attenuates the inflammation, oxidative stress, and apoptosis through the JAK2/STAT3 pathway in cerebral ischemia-reperfusion injury. J Stroke Cerebrovasc Dis 2024:108114. [PMID: 39537043 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108114] [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: 07/22/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Loganin, a monoterpene iridoid glycoside derived from Cornus officinalis Sieb. Et Zucc, has been reported to have anti-inflammatory and antioxidant activity. Nevertheless, the potential role and molecular mechanism of loganin in cerebral ischemia-reperfusion (I/R) injury are not well-understood. The purpose of the study was to explore the functional role of loganin in the inflammation, oxidative stress, and apoptosis in cerebral I/R injury in rats MATERIALS AND METHODS: Following middle cerebral artery occlusion (MCAO), 80 mg/kg of loganin was intragastrically administered for 7 consecutive days. Neuromotor function scores were performed 24 h after the last administration, and the cerebral infarction volume was determined by TTC straining. The expressions of IL-6, IL-1β, and TNF-α in the brain tissues of MCAO rats were detected by ELISA assay. The activities of ROS, SOD, and MDA were measured by ELISA assay as well. Cell apoptosis were was tested by TUNEL straining. Western blot assay was applied for measuring the protein levels RESULTS: We observed that the expressions of IL-6, IL-1β, and TNF-α were amplitude markedly elevated in the rats following MCAO. Treatment with loganin obviously reduced these expressions in the brain tissues of MCAO rats. ELISA assay showed that ROS generation and MDA activity were increased in MCAO group and it was decreased after treatment with loganin. However, loganin increased the SOD activity, which was reduced by MCAO operation. Moreover, loganin promoted neurological function improvement and inhibited cell apoptosis in the rats after MCAO. Mechanically, loganin triggered JAK2/STAT3 phosphorylation in the rats following MCAO, and activation of JAK2/STAT3 pathway rescued the inhibition effects of loganin on the inflammation, oxidative stress, and apoptosis CONCLUSIONS: These results provide evidence that loganin may alleviate the inflammation, oxidative stress, and apoptosis through the JAK2/STAT3 pathway in MCAO rats.
Collapse
Affiliation(s)
- Yunfeng Xi
- Yangzhou University Medical College; Nanjing Luhe people's Hospital, Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| | - Xiaoli Hou
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou 22500, Jiangsu Province, China.
| | - Yuan Huang
- Yangzhou University Medical College; Department of Clinical Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| | - Yan Zhou
- Yangzhou University Medical College; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Institute of Translational Medicine, Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| | - Yu Chen
- Yangzhou University Medical College; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Institute of Translational Medicine, Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| | - Yixia Wang
- Yangzhou University Medical College; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Institute of Translational Medicine, Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| | - Hong Cheng
- Yangzhou University Medical College; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Institute of Translational Medicine, Yangzhou University, Yangzhou 22500, Jiangsu Province, China.
| |
Collapse
|
2
|
Li X, Sun H, Zhang L, Liang H, Zhang B, Yang J, Peng X, Sun J, Zhou Y, Zhai M, Jiang L, Zhu H, Duan W. GDF15 attenuates sepsis-induced myocardial dysfunction by inhibiting cardiomyocytes ferroptosis via the SOCS1/GPX4 signaling pathway. Eur J Pharmacol 2024; 982:176894. [PMID: 39147013 DOI: 10.1016/j.ejphar.2024.176894] [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: 03/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome triggered by infection, presenting with symptoms such as fever, increased heart rate, and low blood pressure. In severe cases, it can lead to multiple organ dysfunction, posing a life-threatening risk. Sepsis-induced cardiomyopathy (SIC) is a critical factor in the poor prognosis of septic patients, leading to myocardial dysfunction characterized by cell death, inflammation, and diminished cardiac function. Ferroptosis, an iron-dependent form of programmed cell death, is a key mechanism causing cardiomyocyte damage in SIC. Growth differentiation factor 15 (GDF15), a member of the TGF-β superfamily, is associated with various cardiovascular diseases and can inhibit oxidative stress, reduce reactive oxygen species (ROS), and suppress ferroptosis. Elevated serum GDF15 levels in sepsis are correlated with organ injuries, suggesting its potential as a therapeutic target. However, its role and mechanisms in SIC remain unclear. Glutathione peroxidase 4 (GPX4), the only enzyme capable of reducing lipid peroxides within cells, protects cells by reducing lipid peroxidation levels and inhibiting ferroptosis. Investigating the regulatory factors of GPX4 may provide a theoretical basis for SIC treatment. In this study, a mouse SIC model revealed that elevated GDF15 exerts a protective effect. Antagonizing GDF15 exacerbates myocardial damage. Through transcriptomic analysis and other methods, we confirmed that GDF15 inhibits the expression of SOCS1 by activating the ALK5-SMAD2/3 pathway, thereby activates the JAK2/STAT3 pathway, promotes the transcription of GPX4, inhibits ferroptosis in cardiomyocytes, and plays a myocardial protective role in SIC.
Collapse
Affiliation(s)
- Xiayun Li
- College of Life Sciences, Northwest University, Xi'an, 710069, China; Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - He Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liyun Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Hongliang Liang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China; Department of Surgery, The 954th Hospital of the Chinese People's Liberation Army, Shannan, 856100, China
| | - Jiachang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Xiangyan Peng
- School of Medicine, Northwest University, Xi'an, 710069, China
| | - Jingwei Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Yang Zhou
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liqing Jiang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Zhao Y, Huang W, Liu F, Sun Q, Shen D, Fan W, Huang D, Zhang Y, Gao F, Wang B. Verapamil attenuates myocardial ischemia/reperfusion injury by inhibiting apoptosis via activating the JAK2/STAT3 signaling pathway. Biomed Pharmacother 2024; 180:117568. [PMID: 39405898 DOI: 10.1016/j.biopha.2024.117568] [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: 07/25/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 11/14/2024] Open
Abstract
Apoptosis is a crucial pathological process in myocardial ischemia/reperfusion injury (MIRI). Verapamil (Ver), normally used to treat hypertension or heart rhythm disorders, also attenuates MIRI. The potential of Ver to inhibit apoptosis and thereby attenuate MIRI remains unclear, as does the mechanism. We established an in vivo mouse ischemia/reperfusion (I/R) model by occlusion of the left anterior descending coronary. To construct a hypoxia/reoxygenation model in vitro, H9c2 cardiomyocytes were immersed in a hypoxic buffer in a hypoxia/anaerobic workstation. Ver significantly improved cardiac function and reduced myocardial infarction size in I/R mice, while decreasing apoptosis. Both in vivo and in vitro, application of Ver activated the JAK2/STAT3 signaling pathway and elevated Bcl-2 expression, while decreasing Bax and cleaved caspase-3 levels. Treatment with AG490, a JAK2 inhibitor, partially counteracted the anti-apoptotic and the cardioprotective effect of Ver. Thus, we conclude that Ver alleviates MIRI by reducing apoptosis via the JAK2/STAT3 signaling pathway activation. These findings provide a novel mechanism of Ver in the treatment of MIRI.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Weiyi Huang
- Department of Clinical Pharmacy, Shantou University Medical College, Shantou 515041, China
| | - Fang Liu
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Qiang Sun
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Daifei Shen
- Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wenjun Fan
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Bin Wang
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
4
|
Chen J, Wang B, Meng T, Li C, Liu C, Liu Q, Wang J, Liu Z, Zhou Y. Oxidative Stress and Inflammation in Myocardial Ischemia-Reperfusion Injury: Protective Effects of Plant-Derived Natural Active Compounds. J Appl Toxicol 2024. [PMID: 39482870 DOI: 10.1002/jat.4719] [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: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death among patients with cardiovascular diseases. Percutaneous coronary intervention (PCI) has been the preferred clinical treatment for AMI due to its safety and efficiency. However, research indicates that the rapid restoration of myocardial oxygen supply following PCI can lead to secondary myocardial injury, termed myocardial ischemia-reperfusion injury (MIRI), posing a grave threat to patient survival. Despite ongoing efforts, the mechanisms underlying MIRI are not yet fully elucidated. Among them, oxidative stress and inflammation stand out as critical pathophysiological mechanisms, playing significant roles in MIRI. Natural compounds have shown strong clinical therapeutic potential due to their high efficacy, availability, and low side effects. Many current studies indicate that natural compounds can mitigate MIRI by reducing oxidative stress and inflammatory responses. Therefore, this paper reviews the mechanisms of oxidative stress and inflammation during MIRI and the role of natural compounds in intervening in these processes, aiming to provide a basis and reference for future research and development of drugs for treating MIRI.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Yang Z, Li C, Wang Y, Dong W, Yang M, Jin J. A single-chain antibody construct with specificity of a natural IgM antibody reduces hepatic ischemia reperfusion injury in mice. J Cell Mol Med 2024; 28:e18291. [PMID: 38597412 PMCID: PMC11005456 DOI: 10.1111/jcmm.18291] [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: 11/03/2023] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
Natural immunoglobulin M (IgM) antibodies have been shown to recognize post-ischemic neoepitopes following reperfusion of tissues and to activate complement. Specifically, IgM antibodies and complement have been shown to drive hepatic ischemia reperfusion injury (IRI). Herein, we investigate the therapeutic effect of C2 scFv (single-chain antibody construct with specificity of a natural IgM antibody) on hepatic IRI in C57BL/6 mice. Compared with PBS-treated mice, C2 scFv-treated mice displayed almost no necrotic areas, significant reduction in serum ALT, AST and LDH levels, and significantly reduced in the number of TUNEL positive cells. Moreover, C2 scFv-treated mice exhibited a notable reduction in inflammatory cells after hepatic IRI than PBS-treated mice. The serum IL-6, IL-1β, TNF-α and MPC-1 levels were also severely suppressed by C2 scFv. Interestingly, C2 scFv reconstituted hepatic inflammation and IRI in Rag1-/- mice. We found that C2 scFv promoted hepatic cell death and increased inflammatory cytokines and infiltration of inflammatory cells after hepatic IRI in Rag1-/- mice. In addition, IgM and complement 3d (C3d) were deposited in WT mice and in Rag1-/- mice reconstituted with C2 scFv, indicating that C2 scFv can affect IgM binding and complement activation and reconstitute hepatic IRI. C3d expression was significantly lower in C57BL/6 mice treated with C2 scFv compared to PBS, indicating that excessive exogenous C2 scFv inhibited complement activation. These data suggest that C2 scFv alleviates hepatic IRI by blocking complement activation, and treatment with C2 scFv may be a promising therapy for hepatic IRI.
Collapse
Affiliation(s)
- Zhi Yang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseasesthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- China–USA Lipids in Health and Disease Research CenterGuilin Medical UniversityGuilinGuangxiChina
- Laboratory of Hepatobiliary and Pancreatic Surgerythe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| | - Chunmei Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseasesthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- China–USA Lipids in Health and Disease Research CenterGuilin Medical UniversityGuilinGuangxiChina
- Laboratory of Hepatobiliary and Pancreatic Surgerythe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| | - Yongqin Wang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| | - Wei Dong
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| | - Moujie Yang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseasesthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| | - Junfei Jin
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repairthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseasesthe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- China–USA Lipids in Health and Disease Research CenterGuilin Medical UniversityGuilinGuangxiChina
- Laboratory of Hepatobiliary and Pancreatic Surgerythe Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
| |
Collapse
|
6
|
Guo B, Yu Y, Wang M, Li R, He X, Tang S, Liu Q, Mao Y. Targeting the JAK2/STAT3 signaling pathway with natural plants and phytochemical ingredients: A novel therapeutic method for combatting cardiovascular diseases. Biomed Pharmacother 2024; 172:116313. [PMID: 38377736 DOI: 10.1016/j.biopha.2024.116313] [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: 01/06/2024] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 02/22/2024] Open
Abstract
The aim of this article is to introduce the roles and mechanisms of the JAK2/STAT3 pathway in various cardiovascular diseases, such as myocardial fibrosis, cardiac hypertrophy, atherosclerosis, myocardial infarction, and myocardial ischemiareperfusion. In addition, the effects of phytochemical ingredients and different natural plants, mainly traditional Chinese medicines, on the regulation of different cardiovascular diseases via the JAK2/STAT3 pathway are discussed. Surprisingly, the JAK2 pathway has dual roles in different cardiovascular diseases. Future research should focus on the dual regulatory effects of different phytochemical ingredients and natural plants on JAK2 to pave the way for their use in clinical trials.
Collapse
Affiliation(s)
- Bing Guo
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Yunfeng Yu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Min Wang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ronghui Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan He
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Siqin Tang
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Qili Liu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yilin Mao
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China.
| |
Collapse
|